WO2021195602A1 - Alzheimer's diagnostic method - Google Patents

Alzheimer's diagnostic method Download PDF

Info

Publication number
WO2021195602A1
WO2021195602A1 PCT/US2021/024539 US2021024539W WO2021195602A1 WO 2021195602 A1 WO2021195602 A1 WO 2021195602A1 US 2021024539 W US2021024539 W US 2021024539W WO 2021195602 A1 WO2021195602 A1 WO 2021195602A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
pbmc
subject
stimulatory agent
memory
Prior art date
Application number
PCT/US2021/024539
Other languages
French (fr)
Inventor
Thomas MONTINE
Thanaphong PHONGPREECHA
Rosemary FERNANDEZ
Nima AGHAEEPOUR
Brice Gaudilliere
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Publication of WO2021195602A1 publication Critical patent/WO2021195602A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/27Esters, e.g. nitroglycerine, selenocyanates of carbamic or thiocarbamic acids, meprobamate, carbachol, neostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/5052Cells of the immune system involving B-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/912Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • G01N2333/91205Phosphotransferases in general
    • G01N2333/9121Phosphotransferases in general with an alcohol group as acceptor (2.7.1), e.g. general tyrosine, serine or threonine kinases
    • G01N2333/91215Phosphotransferases in general with an alcohol group as acceptor (2.7.1), e.g. general tyrosine, serine or threonine kinases with a definite EC number (2.7.1.-)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/916Hydrolases (3) acting on ester bonds (3.1), e.g. phosphatases (3.1.3), phospholipases C or phospholipases D (3.1.4)

Definitions

  • biomarkers for Alzheimers disease include cerebrospinal fluid and PET markers of amyloid-beta; and tau proteins, which are accurate in detecting the presence of AD-associated pathophysiological and neuropathological changes.
  • biomarkers the high cost, insufficient accessibility and/or invasiveness of these assays limit their use as viable first-line tools for detecting pathophysiologic patterns, aka biomarkers.
  • Blood-based and plasma- based biomarkers are being developed with some showing promises, but results in the field have been mixed because of several reasons such as selected candidates rather than data- derived candidates, reliance on just one to only a few biomarkers, inconsistencies in clinical cohorts, and discrepancies in diagnostic evaluations. The methods provided herein address these and other needs in the art.
  • FIG.s 1A-1C show the experimental and analytical workflow from obtaining PBMCs to identifying potential immune cell markers.
  • FIG. 1A shows in that the discovery cohorts, whole blood was collected from 28 individuals with AD and 17 individuals with PD; AD was compared with the samples from 53 older healthy controls (HC-I) while PD samples were compared with a subset of those with age- and sex-matched healthy controls (HC-I sub ).
  • FIG. IB illustrates PBMCs were either unstimulated or stimulated with IFN-a, IL- 6, IL-7, IL-10, IL-21, LPS, or PMA/ionomycin. PBMCs were then bound with 21 metal- conjugated antibodies to surface markers and 15 metal-conjugated antibodies to intracellular signaling molecules before analysis by CyTOF.
  • FIG. 1C shows cell abundance was evaluated on PBMCs from unstimulated condition.
  • the stimulations and antibody probes generated a total of 4200 intracellular signaling responses (35 PBMC subtypes under 8 stimulating conditions and assayed for 15 intracellular responses), which were used to identify the potential immune features with the aid of cell-signaling knowledge, machine learning methods, and statistical analysis.
  • FIG.s 2A-2F illustrate AD and PD patients exhibited different cell frequency in several cell subsets.
  • FIG. 2A is a CITRUS-derived dendrogram of unstimulated PBMC data from AD and PD.
  • PBMC subsets are represented as clusters and color coded for level of marker expression (see Methods). Red highlighted nodes are cell subtypes of CD4 + Tcemrai Mem, blue highlighted nodes are CD4 + TActivated, orange highlighted clusters are CD8 + TNaive and the green clusters are CD38 + CD16 low monocytes that were significantly different (q value ⁇ 0.05) among the two diagnostic groups.
  • FIG. 2B is a bar graph showing the significance levels of the highlighted nodes.
  • FIG.s 2C-2F are box plots presenting example values from the last node in each of the four highlighted lineages for CD4 + Tcemrai memory cells (FIG. 2C), CD4 + TActivated cells (FIG. 2D), CD8 1 T Na ive cells (FIG. 2E), and CD38 + CD16 low monocytes (FIG 2F). P values for the stratified subsets were determined using Wilcoxon rank-sum test.
  • FIG.s 3A and 3B show responses from the same intracellular signaling proteins are highly correlated to each other.
  • FIG. 3A is a correlation network (Spearman’s coefficient) of immune features obtained from CyTOF data of HC-I, AD, and PD colored by the type of stimulant. The edges of the network represent features with Spearman’s coefficient higher than 0.8.
  • FIG. 3B illustrates an unsupervised algorithm clustering the network into 24 communities, where their annotations are based on commonly shared feature attributes (PBMC subtype, stimulation, or signaling property) within the community.
  • PBMC subtype, stimulation, or signaling property commonly shared feature attributes
  • FIG.s 4A-4E illustrate that the iEN model can satisfactorily classify AD/HC-I in both discovery and validation cohorts with most important model components associated with signals from pPLCy2 and pSTATs.
  • FIG. 4A is box plots showing the predicted values from iEN model with Wilcoxon rank-sum test P value for discovery and validation cohort.
  • FIG. 4B is receiver operating characteristics (ROC) curves from the iEN model predictions of discovery and validation cohorts.
  • FIG. 4C is a correlation network colored by iEN model components with red and blue colors highlighting the components that are indicative of AD and HC-I, respectively. The size of the nodes represents the Spearman’s coefficient of the immune feature to the respective ground truths.
  • FIG. 4D illustrates a model reduction analysis looking at the effect of the number of included features on iEN performance.
  • FIG. 4E is a correlation network colored and annotated only for the top 14 features that were associated with components selected from model reduction.
  • FIG.s 5A and 5B illustrate portions of the canonical response versus responses observed experimentally in this study.
  • FIG. 5A is a correlation network illustrated by responses that were expected in literature and were found in this study (grey), those that were not expected from literature and were also not found (white), and those that were expected in literature but not found experimentally and vice versa.
  • FIG. 5B shows the portion of the responses that were observed (and not observed) according to literature and experiments in this study.
  • FIG.s 6A-6E are visualizations of features with strong signals and other features associated with 111 top iEN selected components.
  • FIG. 6A is a network correlation with all of the features associated with top 111 iEN components colored.
  • FIG. 6A are heatmaps of the selected communities with magnitude colored by Spearman’s correlation (r).
  • FIG.s 6C- 6E are box plots of the selected features in the communities for pPLCy2 (FIG. 6C), pSTATl (FIG. 6D) and pSTAT5 (FIG. 6E) responses.
  • the shaded gray area indicated that the features are also a part of reduced iEN’s top 14 components, and the box indicates features that are among the top 111 components.
  • FIG.s 7A-7H are heatmaps and box plots of the intracellular response in the PBMC of the selected communities highlight immune features for AD/HC-I classification.
  • FIG.s 7A-7C are heatmaps of the pPLCy2 (FIG. 7A), pSTATl (FIG. 7B), and pSTAT5 (FIG. 7C) responses by PBMC subtypes and stimulations. The color of the heatmap scaled with the Wilcoxon rank-sum testP value of the difference in response of the immune feature between HC-I and AD patients. The network communities annotated with these responses (community 12, 17, 18, and 20) were depicted on the left hand side of the heatmap.
  • FIG.s 7D-7H are box plots showing the significant difference of the selected immune features from the heatmap, including pPLCy2 response in unstimulated NKT cells (FIG. 7D), pPLCy2 response in LPS stimulated NKT cells (FIG. 7E), pPLCy2 response in unstimulated CD56 bnght NK cells (FIG. 7F), and pSTATl response in IFN-a stimulated plasmablast cells (FIG. 7G).
  • FIG.s 8A-8G illustrate that responses of pPLCy2. pSTATl, and pSTAT5 show expected expressions according literature.
  • FIG. 8A are heatmaps of the mean values of normalized responses in each cell subset and stimulation for all AD patients in the discovery cohorts before batch correction.
  • FIG. 8B is similar to FIG. 8A but for HC-I participants.
  • FIG. 8C is a similar heatmap with only AD patients of the biggest batch in the discovery cohort.
  • FIG. 8D is similar to FIG. 8C but for HC-I participants.
  • FIG.s 8E-8G are examples of raw gated signals and cell frequency with the corresponding histograms from a pair of participants. Flow cytometry data for pPLCy2 response of unstimulated NKTs (FIG. 8E), pSTATl response of IFN-a stimulated DCs (FIG. 8F), and pSTATl response of IFN-a stimulated monocytes (FIG. 8F) are shown.
  • FIG.s 9A-9F are correlation networks for AD/HC-I separated by sex and APOE :-:4 allele.
  • FIGs. 9A and 9B is the correlation network with node size corresponding to the Wilcoxon rank-sum testP value of each feature for AD/HC-I diagnosis in male (FIG. 9A) and female participants (FIG. 9B) (discovery cohort).
  • FIGs. 9C and 9D show a similar correlation network for all sex of healthy cohorts (HC-I) but for sex-specific AD.
  • FIG. 9E is a similar correlation network for all sex of healthy cohorts (HC-I) with no APOE :-:4 allele and all sex of AD with at least one APOE e4 allele.
  • FIG. 9F is similar to FIG. 9E but for HC-I and AD both groups with no APOE e4 allele.
  • the color of each node represented the magnitude and direction of the associated iEN components developed from AD/HC-I classification model using all AD and HC-I data.
  • FIG.s 10A and 10B are correlation networks of HC-II/HC-I and HC-II/AD indicating that aging does not share key differential pSTATs and pPLCy2 signals highlighted by HC-I/ AD.
  • FIG. 10A is a correlation network with node size corresponding to the Wilcoxon rank-sum testP value of each feature for HC-II/HC-I. The color of each node represented the magnitude and direction of the associated iEN components developed from AD/HC-I classification.
  • FIG. 10B is similar to FIG. 10A however for the HC-II/AD pair.
  • FIG.s 11A-11C illustrate that cross disease prediction reveals similarities between AD and PD.
  • FIG. 11A is a bar graph showing performance of the disease cross-prediction using iEN components developed from AD/HC-I diagnosis to classify PD/HC-I sub and AD/PD.
  • FIG. 11B shows the iEN predicted values for each diagnostic group.
  • FIG. 11C is a the correlation network with node size corresponding to the Wilcoxon rank-sum test P value of each feature for PD/HC-I sub diagnosis, with the color of each node representing the magnitude and direction of the associated iEN components developed from AD/HC-I.
  • the network highlighted possible regions, such as in the labeled clusters, where AD and PD signals can overlap.
  • FIG. 12 illustrates the gating process to obtain different cell subsets.
  • Cell subsets (35) were gated from DNA high, cleaned up PBMC and a matrix of 12 phospho-epitopes and 3 endosomal proteins with 8 stimuli conditions were evaluated from each subset.
  • FIG. 13 illustrates additional non-gender specific signals including first, pS6 expression in IFN-a and IL-6 stimulated CD4+ T cells, most CD8+ T cells, and NK cells. Second, pSTAT3 signals from specific subset of B cells including memory B cells, Naive B cells, and Switched memory B cells.
  • culture when referring to cell culture itself or the process of culturing, can be used interchangeably to mean that a cell is maintained outside the body (e.g., ex vivo) under conditions suitable for survival.
  • Cultured cells are allowed to survive, and culturing can result in cell growth, differentiation, or division. The term does not imply that all cells in the culture survive or grow or divide, as some may naturally senesce, etc.
  • Cells are typically cultured in media, which can be changed during the course of the culture.
  • media and “culture solution” refer to the cell culture milieu.
  • Media is typically an isotonic solution, and can be liquid, gelatinous, or semi-solid, e.g., to provide a matrix for cell adhesion or support.
  • Media as used herein, can include the components for nutritional, chemical, and structural support necessary for culturing a cell.
  • condition to allow growth in culture and the like refers to conditions of temperature (typically at about 37° C for mammalian cells), humidity, CO2 (typically around 5%), in appropriate media (including salts, buffer, serum), such that the cells are able to undergo cell division or at least maintain viability for at least 24 hours, preferably longer (e.g., for days, weeks or months).
  • Suitable culture conditions are described herein, and can include standard tissue culture conditions.
  • PBMCs can be cultured in a buffered media that includes amino acids, nutrients, growth factors, etc., as will be understood in the art.
  • a cell derived from an individual when referring to cells or a biological sample, indicates that the cell or sample was obtained from the stated source at some point in time.
  • a cell derived from an individual can represent a primary cell obtained directly from the individual (i.e., unmodified), or can be modified, e.g., by introduction of a recombinant vector, by culturing under particular conditions, or immortalization.
  • a cell derived from a given source will undergo cell division and / or differentiation such that the original cell is no longer exists, but the continuing cells will be understood to derive from the same source.
  • amino acid residue in a protein "corresponds" to a given residue when it occupies the same essential structural position within the protein as the given residue.
  • nucleic acid or protein when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It can be, for example, in a homogeneous state and may be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, g- carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i. e..
  • R group e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium.
  • Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • non-naturally occurring amino acid” and “unnatural amino acid” refer to amino acid analogs, synthetic amino acids, and amino acid mimetics which are not found in nature.
  • amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • polypeptide peptide
  • protein protein
  • the terms "polypeptide,” “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues, wherein the polymer may In embodiments be conjugated to a moiety that does not consist of amino acids. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers.
  • a "fusion protein” refers to a chimeric protein encoding two or more separate protein sequences that are recombinantly expressed as a single moiety.
  • a “detectable agent” or “detectable moiety” is a compound or composition detectable by appropriate means such as spectroscopic, photochemical, biochemical, immunochemical, chemical, magnetic resonance imaging, or other physical means.
  • a detectable moiety is a monovalent detectable agent or a detectable agent bound (e.g. covalently and directly or via a linking group) with another compound, e.g., a nucleic acid.
  • Exemplary detectable agents/moieties for use in the present disclosure include an antibody ligand, a peptide, a nucleic acid, radioisotopes, paramagnetic metal ions, fluorophore (e.g.
  • DYNABEADS® by ThermoFisher encompassing functionalized magnetic beads such as DYNABEADS® M-270 amine by ThermoFisher
  • paramagnetic molecules paramagnetic molecules, paramagnetic nanoparticles, ultrasmall superparamagnetic iron oxide nanoparticles, ultrasmall superparamagnetic iron oxide nanoparticle aggregates, superparamagnetic iron oxide nanoparticles, superparamagnetic iron oxide nanoparticle aggregates, monocrystalline iron oxide nanoparticles, monocrystalline iron oxide, nanoparticle contrast agents, liposomes or other delivery vehicles containing Gadolinium chelate molecules, gadolinium, radionuclides (e.g.
  • microbubbles e.g. including microbubble shells including albumin, galactose, lipid, and/or polymers; microbubble gas core including air, heavy gas(es), perfluorcarbon, nitrogen, octafluoropropane, perflexane lipid microsphere, perflutren, etc.
  • iodinated contrast agents e.g.
  • the detectable agent is a detectable fluorescent agent. In embodiments, the detectable agent is a detectable phosphorescent agent.
  • the detectable agent is a detectable radioactive agent. In embodiments, the detectable agent is a detectable metalloenzyme. In embodiments, the detectable agent is a detectable colorimetric agent. In embodiments, the detectable agent is a detectable luminescent agent. In embodiments, the detectable agent is a detectable spectrophotometric agent. In embodiments, the detectable agent is a detectable metal-organic framework. In embodiments, the detectable agent is detectable by means other than by spectroscopy. In embodiments, the detectable agent comprises a fluorophore linked to biotin, avidin, or streptavidin. In embodiments, the detectable agent comprises a fluorophore linked to streptavidin.
  • the detectable agent comprises a fluorophore linked to avidin. In embodiments, the detectable agent comprises a fluorophore linked to avidin linked to biotin. In embodiments, the detectable agent comprises a fluorophore linked to streptavidin linked to biotin.
  • antibody is used according to its commonly known meaning in the art. Antibodies exist, e.g., as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)'2, a dimer of Fab which itself is a light chain joined to VH-CHI by a disulfide bond. The F(ab)'2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)'2 dimer into an Fab' monomer.
  • the Fab' monomer is essentially Fab with part of the hinge region (see Fundamental Immunology (Paul ed., 3d ed. 1993). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology. Thus, the term antibody, as used herein, also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv) or those identified using phage display libraries (see, e.g., McCafferty et al, Nature 348:552-554 (1990)).
  • An exemplary immunoglobulin (antibody) structural unit comprises a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD).
  • the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the terms variable light chain (VL), variable light chain (VL) domain or light chain variable region and variable heavy chain (VH), variable heavy chain (VH) domain or heavy chain variable region refer to these light and heavy chain regions, respectively.
  • variable light chain (VL), variable light chain (VL) domain and light chain variable region as referred to herein may be used interchangeably.
  • variable heavy chain VH
  • variable heavy chain domain VH domain
  • heavy chain variable region variable heavy chain variable region
  • the Fc i.e. fragment crystallizable region; also referred to herein as “Fc domain”
  • Fc domain is the "base” or “tail” of an immunoglobulin and is typically composed of two heavy chains that contribute two or three constant domains depending on the class of the antibody. By binding to specific proteins, the Fc region ensures that each antibody generates an appropriate immune response for a given antigen.
  • the Fc region also binds to various cell receptors, such as Fc receptors, and other immune molecules, such as complement proteins.
  • the Fc region includes a constant heavy chain domain 3 (CH3 domain) and a constant heavy chain domain 2 (CH2 domain).
  • the epitope of an antibody is the region of its antigen to which the antibody binds.
  • Two antibodies bind to the same or overlapping epitope if each competitively inhibits (blocks) binding of the other to the antigen. That is, a lx, 5x, lOx, 20x or lOOx excess of one antibody inhibits binding of the other by at least 30% but preferably 50%, 75%, 90% or even 99% as measured in a competitive binding assay (see, e.g., Junghans etal, Cancer Res. 50:1495, 1990).
  • two antibodies have the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • CD Cluster of Differentiation
  • Each numeric antigen is a specific protein that is recognized in the art by its CD designation.
  • CD3 as referred to herein is a protein complex comprising four chain including CD3y chain, a CD35 chain, and two CD3s chains.
  • An example sequences of CD3 complex chains include: Epsilon chain precursor (GENBANK® Accession No.
  • NP_000724.1 Gamma chain precursor (GENBANK® Accession No. NP_000064.1); Delta chain precursor (GENBANK® Accession No. NP_000723.1) which are incorporated herein by reference. Multiple isoforms are possible for each of the chains of CD3.
  • CD4 as referred to herein is a glycoprotein expressed on the surface of T helper cells, regulatory T cells, monocytes, macrophages, and dendritic cells.
  • CD4 was originally known as leu-3 and T4 (after the OKT4 monoclonal antibody).
  • CD4 as referred to herein has four immunoglobulin domains (Di to D4) that are exposed on the extracellular surface of the cell, see ENTREZ No. 920, UNIPROT No. P01730, and GENBANK® Accession No. NP 000607, which are incorporated by reference.
  • CD8 is a transmembrane glycoprotein that serves as a co-receptor for the T cell receptor (TCR). Like the TCR, CD8 binds to a major histocompatibility complex ⁇ MS 1C) molecule, but is specific for the class I MHC protein, see ENTREZ No. 925 and UNIPROT No. R0G732, which are incorporated by reference herein.
  • TCR T cell receptor
  • CD45RA refers to the CD45 Receptor antigen also known as Protein tyrosine phosphatase, receptor type, C (PTPRC).
  • exemplary amino acid sequences for CD45RA include GENBANK® Accession Nos. NP_002829.3, NP_563578.2, NP_563578.2, and NP_002829.3, which are all incorporated herein by reference.
  • CD45RA is expressed on naive T cells, as well as on CD8- and CD4-expressing effector cells. After antigen interaction, T cells gain expression of CD45RO and lose expression of CD45RA. Thus, either CD45RA or CD45RO is used to generally differentiate the naive from memory T cell populations.
  • a “CD45RA-negative CD8 T cell” as provided herein is a CD8 T cell which lacks expression of detectable levels of CD45RA.
  • the CD45RA- negative CD8 T cell is a memory T cell.
  • a "CD45RA-negative CD4 T cell” as provided herein is a CD4 T cell which lacks expression of detectable amounts of CD45RA.
  • the CD45RA-negative CD4 T cell is a memory T cell.
  • the CD45RA-negative CD8 T cell is a memory T cell.
  • CD19 protein or “CD19” as used herein includes any of the recombinant or naturally-occurring forms of B-lymphocyte antigen CD 19, also known as CD 19 molecule (Cluster of Differentiation 19), B-Lymphocyte Surface Antigen B4, T-Cell Surface Antigen Leu- 12 and CVID3, or variants or homologs thereof that maintain CD 19 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to CD 19).
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring CD 19 protein.
  • the CD 19 protein is substantially identical to the protein identified by the UniProt reference number P15391 or a variant or homolog having substantial identity thereto.
  • CD20 protein or “CD20” as used herein includes any of the recombinant or naturally-occurring forms of B-lymphocyte antigen CD20 or Cluster of Differentiation 20 (CD20), or variants or homologs thereof that maintain CD20 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to CD20).
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring CD20 protein.
  • the CD20 protein is substantially identical to the protein identified by the UniProt reference number PI 1836 or a variant or homolog having substantial identity thereto.
  • CD20 is involved in regulating early steps in the activation and differentiation process of B cells (Tedder et al., Eur. J. Immunol. 16:881-887, 1986) and can function as a calcium ion channel (Tedder et al., J. Cell. Biochem.14D:195, 1990).
  • Exemplary amino acid sequences for CD20 are provided in GENBANK® Accession Nos. NP 068769.2 (human), NP 690605.1 (human), and NP 031667.1 (mouse), which are incorporated by reference herein.
  • PLC-y2 includes any of the recombinant or naturally-occurring forms of the 1- Phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2 (PLC-y2) enzyme or variants or homologs thereof that maintain PLC-y2 enzyme activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to PLC-y2).
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring PLC- y2 polypeptide.
  • PLC-y2 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference PI 6885, homolog or functional fragment thereof.
  • PLC-y2 is the protein as identified by the NCBI Reference Sequence: NP_002652.2, homolog or functional fragment thereof.
  • PLC-y2 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_002661.5, homolog or functional fragment thereof.
  • AKT or "AKT protein” as provided herein includes any of the recombinant or naturally-occurring forms of the Protein kinase B (PKB), also known as AKT or variants or homologs thereof that maintain AKT kinase activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to AKT).
  • PKT Protein kinase B
  • variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring AKT polypeptide.
  • AKT is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P31749, homolog or functional fragment thereof.
  • AKT is encoded by a nucleic acid sequence identified by NCBI Reference Sequence:
  • NM_005163.2 homolog or functional fragment thereof.
  • STAT5 or "STAT5 protein” as provided herein includes any of the recombinant or naturally-occurring forms of the Signal transducer and Activator of transcription 5 (STAT5) or variants or homologs thereof that maintain STAT5 protein activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to STAT5).
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring STAT5 polypeptide.
  • STAT5 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P42229, homolog or functional fragment thereof. In embodiments, STAT5 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_003152.4, homolog or functional fragment thereof.
  • STAT1 or "STAT1 protein” as provided herein includes any of the recombinant or naturally-occurring forms of the Signal transducer and Activator of transcription 1 (STAT1) or variants or homologs thereof that maintain STAT1 protein activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to STAT1).
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring STAT1 polypeptide.
  • STAT1 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P42224.2, homolog or functional fragment thereof. In embodiments, STAT1 is the protein as identified by the NCBI Reference Sequence: NP 644671, homolog or functional fragment thereof. In embodiments, STAT1 is the protein as identified by the NCBI Reference Sequence: NP_009330, homolog or functional fragment thereof. In embodiments, STAT1 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_007315.4, homolog or functional fragment thereof. In embodiments, STAT1 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_139266.3, homolog or functional fragment thereof.
  • interferon a includes any of the recombinant or naturally-occurring forms of the interferon alpha protein (IFN-a) or variants or homologs thereof that maintain IFN-a protein activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IFN-a).
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g.
  • IFN-a is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P01562, homolog or functional fragment thereof.
  • IFN-a is the protein as identified by the NCBI Reference Sequence: NP_076918.1, homolog or functional fragment thereof.
  • IFN-a is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_024013.3, homolog or functional fragment thereof.
  • interleukin-6 includes any of the recombinant or naturally-occurring forms of the cytokine interleukin 6 (IL-6) or variants or homologs thereof that maintain IL-6 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IL-6).
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring IL-6 polypeptide.
  • IL-6 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P05231, homolog or functional fragment thereof. In embodiments, IL-6 is the protein as identified by the NCBI Reference Sequence: NP_001358025.1, homolog or functional fragment thereof. In embodiments, IL-6 is the protein as identified by the NCBI Reference Sequence: NP_001305024.1, homolog or functional fragment thereof. In embodiments, IL-6 is the protein as identified by the NCBI Reference Sequence:
  • IL-6 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001371096.1, homolog or functional fragment thereof. In embodiments, IL-6 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001318095.2, homolog or functional fragment thereof. In embodiments, IL-6 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_000600.5, homolog or functional fragment thereof.
  • interleukin-7 includes any of the recombinant or naturally-occurring forms of the cytokine interleukin 7 (IL-7) or variants or homologs thereof that maintain IL-7 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IL-7).
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring IL-7 polypeptide.
  • IL-7 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference PI 3232, homolog or functional fragment thereof. In embodiments, IL-7 is the protein as identified by the NCBI Reference Sequence: NP_001186817.1, homolog or functional fragment thereof. In embodiments, IL-7 is the protein as identified by the NCBI Reference Sequence: NP_001186816.1, homolog or functional fragment thereof. In embodiments, IL-7 is the protein as identified by the NCBI Reference Sequence:
  • IL-7 is the protein as identified by the NCBI Reference Sequence: NP_000871.1, homolog or functional fragment thereof. In embodiments, IL-7 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001199888.2, homolog or functional fragment thereof. In embodiments, IL-7 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001199887.2, homolog or functional fragment thereof. In embodiments, IL-7 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence:
  • IL-7 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_000880.4, homolog or functional fragment thereof.
  • interleukin- 10 includes any of the recombinant or naturally-occurring forms of the cytokine interleukin 10 (IL-10) or variants or homologs thereof that maintain IL-10 activity (e.g. within at least 50%, 80%,
  • IL-10 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P22301, homolog or functional fragment thereof.
  • IL-10 is the protein as identified by the NCBI Reference Sequence: NP_000563.1, homolog or functional fragment thereof.
  • IL-10 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_000572.3, homolog or functional fragment thereof.
  • interleukin-21 includes any of the recombinant or naturally-occurring forms of the cytokine interleukin 21 (IL-21) or variants or homologs thereof that maintain IL-21 activity (e.g. within at least 50%, 80%,
  • IL-21 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference Q9HBE4, homolog or functional fragment thereof.
  • IL-21 is the protein as identified by the NCBI Reference Sequence: NP_068575.1, homolog or functional fragment thereof.
  • IL-21 is the protein as identified by the NCBI Reference Sequence: NP 001193935.1, homolog or functional fragment thereof.
  • IL- 21 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001207006.3, homolog or functional fragment thereof.
  • IL-21 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_021803.4, homolog or functional fragment thereof.
  • LPS lipopoly saccharide
  • the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% structural identity across the whole polymer or a portion thereof compared to a naturally occurring LPS.
  • LPSs Lipopolysaccharides as provided herein are components of the cell wall of Gram-negative bacteria.
  • LPS and its lipid A moiety stimulate cells of the innate immune system by the Toll-like receptor 4 (TLR4), a member of the Toll-like receptor protein family, which recognizes common pathogen-associated molecular-patterns (PAMPs).
  • TLR4 Toll-like receptor 4
  • PAMPs common pathogen-associated molecular-patterns
  • PMA or "phorbol myristate acetate” as provided herein includes any of the recombinant or naturally-occurring forms of 12-O-Tetradecanoylphorbol-l 3-acetate (TP A), also commonly known as tetradecanoylphorbol acetate, tetradecanoyl phorbol acetate, and phorbol 12-myristate 13-acetate (PMA) or variants or homologs thereof that maintain PMA activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to PMA).
  • PMA is the compound identified by Cas Registry Number 16561-29-8.
  • a "cell” as used herein, refers to a cell carrying out metabolic or other function sufficient to preserve or replicate its genomic DNA.
  • a cell can be identified by well-known methods in the art including, for example, presence of an intact membrane, staining by a particular dye, ability to produce progeny or, in the case of a gamete, ability to combine with a second gamete to produce a viable offspring.
  • Cells may include prokaryotic and eukaroytic cells.
  • Prokaryotic cells include but are not limited to bacteria.
  • Eukaryotic cells include but are not limited to yeast cells and cells derived from plants and animals, for example mammalian, insect (e.g., spodoptera) and human cells.
  • B Cells may be useful when they are naturally nonadherent or have been treated not to adhere to surfaces, for example by trypsinization.
  • B cells are lymphocytes, a type of white blood cell (leukocyte), that develops into a plasma cell (a “mature B cell”), which produces antibodies.
  • An “immature B cell” is a cell that can develop into a mature B cell.
  • pro-B cells undergo immunoglobulin heavy chain rearrangement to become pro B pre B cells, and further undergo immunoglobulin light chain rearrangement to become an immature B cells.
  • Immature B cells include T1 and T2 B cells.
  • T cells or "T lymphocytes” as used herein are a type of lymphocyte (a subtype of white blood cell) that plays a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells, by the presence of a T-cell receptor on the cell surface. T cells include, for example, natural killer T (NKT) cells, cytotoxic T lymphocytes (CTLs), regulatory T (Treg) cells, and T helper cells.
  • NKT natural killer T
  • CTLs cytotoxic T lymphocytes
  • Treg regulatory T helper cells.
  • T cells Different types can be distinguished by use of T cell detection agents.
  • CD4 + T lymphocyte or "CD4 T cell” as referred to herein is lymphocyte that expresses the CD4 glycoprotein on its surface.
  • CD4 T cells include helper T cells, which are T cells that help orchestrate the immune response, including antibody responses and killer T cell responses.
  • CD4 T cell precursors differentiate into one of several subtypes, including TH1 (type 1 helper T cell), TH2 (type 2 helper T cell), TH3 (T helper 3 cells), TH17 (T helper 17 cells) or TFH (Follicular B helper T cells). These subtypes of helper T cells are characterized by their secretion of different cytokines to facilitate different types of immune responses.
  • a CD4 T cell is an effector T cell.
  • An "effector T cell” as referred to herein is a T cell that has been activated by its cognate antigen, and is actively involved in eliminating a pathogen. Thus, an effector T cell actively responds to a stimulus (a pathogen or a co-stimulation) and carries out a cell-mediated immune response.
  • Non- limiting examples of effector T cells as referred to herein include helper T cells, killer T cells (cytotoxic T cells) and regulatory T cells.
  • a "CD8 + T lymphocyte” or "CD8 T cell” as referred to herein is a lymphocyte that expresses the CD8 glycoprotein on its surface.
  • Examples of CD8 T cells include cytotoxic T cells and natural killer cells.
  • a CD8 T cell is a cytotoxic T cell.
  • a CDS T cell is a suppressor T cell.
  • a "memory T cell” is a T cell that has previously encountered and responded to its cognate antigen during prior infection, encounter with cancer or previous vaccination. At a second encounter with its cognate antigen memory T cells can reproduce (divide) to mount a faster and stronger immune response than the first time the immune system responded to the pathogen.
  • the memory T cell is a CD45RA-negative CD4 T cell. In embodiments, the memory T cell is a CD45RA-negative CD8 T cell.
  • a “regulatory T cell” or “suppressor T cell” is a lymphocyte which modulates the immune system, maintains tolerance to self-antigens, and prevents autoimmune disease. Regulatory T cells express the CD4, FOXP3, and CD25 and are thought to be derived from the same lineage as naive CD4 cells.
  • PBMC peripheral blood mononuclear cell
  • PBMCs peripheral blood mononuclear cell
  • PBMCs are a cell population including lymphocytes (T cells, B cells, NK cells) and monocytes.
  • PBMCs do not include cells that lack a nucleus.
  • PBMCs do not include erythrocytes or platelets.
  • PBMCs do not include erythrocytes.
  • PBMCs do not include platelets.
  • PBMCs do not include cells with a multi-lobed nucleus.
  • PBMCs do not include granulocytes.
  • PBMCs do not include neutrophils.
  • PBMCs do not include basophils.
  • PBMCs do not include eosinophils.
  • lymphocytes make up the majority of the PBMC population, followed by monocytes, and only a small percentage of dendritic cells.
  • a "natural killer T cell” or “NKT cell” is a heterogeneous group of T cells including T cells and natural killer cells (Nk cells).
  • natural killer T cells bind to non- polymorphic CD Id molecule.
  • Natural killer T cells constitute approximately 1% of all peripheral blood T cells.
  • the natural killer T cell is aNKl.l+ T cell.
  • the natural killer T cell is a NK1.1-
  • the natural killer T cell is a CD4+ T cell.
  • the natural killer T cell is a CD4- T cell.
  • the natural killer T cell is a CD8+ T cell.
  • the natural killer T cell is aNKl.l+ T cell CD8- T cell.
  • the natural killer T cell is CD16+ and CD56+ T cell.
  • the natural killer T cell is granzyme producing T cell.
  • a "natural killer cell”, "NK cell” or "large granular lymphocyte (LGL)” is a type of cytotoxic lymphocyte critical to the innate immune system.
  • a NK cell is identified by the expression of CD56 and the lack of expression of CD3 (CD56+, CD3-).
  • a NK cell is a CD56+ and CD3- lymphocyte.
  • a NK cell is a CD56bright or CD56dim lymphocyte.
  • CD56bright NK cells are similar to T helper cells in exerting their influence by releasing cytokines.
  • CD56bright NK cells constitute the majority of NK cells in bone marrow, secondary lymphoid tissue, liver, and skin.
  • CD56dim NK cells form part of the peripheral blood and are capable of cell killing.
  • CD56dim NK cells are CD 16 positive lymphocytes.
  • NK cells may eliminate virus -infected cells by CD16-mediated ADCC (antibody-dependent cellular cytotoxicity).
  • a monocyte refers to a leukocyte that is able to differentiate in macrophages or dendritic cells.
  • a monocyte is a CD14++ and CD 16- leukocyte.
  • a monocyte is a CD 14+ and CD 16+ leukocyte.
  • a monocyte is a CD14++ and CD 16+ leukocyte.
  • basophil refers to a leukocyte that is responsible for inflammatory reactions during immune response, as well as in the formation of acute and chronic allergic diseases, including anaphylaxis, asthma, atopic dermatitis and hay fever.
  • a basophil is aFcsRI+, CD123+, CD49b(DX-5)+, CD69+, Thy-1.2+, 2B4+, CDllbdull, CD117(c-kit)-, CD24-, CD19-, CD80-, CD14-, CD23-, Ly49c-, CD122- CDllc-, Gr-1-, NK1.1-, B220-, CD3-, gd TCR-, ajlTCR-. a4 and 4-integrin negative leukocyte.
  • a monocyte is a CD14++ and CD16+ cell.
  • a basophil is a cell positive for CD13, CD44, CD54, CD63, CD69, CD107a, CD123, CD164, CD 193/ CCR3, CD203c, TLR-4, and FcsRI.
  • a basophil is an activated basophil and expresses increased levels of CD13, CD107a, CD164, or surface-exposed CD63 and the ectoenzyme CD203c relative to a non-activated basophil.
  • dendritic cell refers to an antigen- presenting cells (APC) or accessory cell of the mammalian immune system.
  • a dendritic cell processes antigen material and presents it on the cell surface to the T cells of the immune system.
  • Dendritic cells act as messengers between the innate and the adaptive immune systems.
  • the term dendritic cells include "myeloid DCs” (mDCs) and “plasmacytoid dendritic cells” (pDCs). In embodiments, the dendritic cell is an mDC.
  • an mDC is a mDC-1 and is a major stimulator of T cells.
  • an mDC is an mDC- 2 and involved in wound infection.
  • the mDC expresses Interleukin 12 (IL- 12), Interleukin 6 (IL-6), TNF, and/or chemokines.
  • the mDC expresses TLR2 and/or TLR4.
  • the dendritic cell is a pDC.
  • the pDC produces high amounts of IFN-a.
  • the pDC expresses TLR7 and/or TLR9.
  • the dendritic cell is a CDlc+ myeloid DC. In embodiments, the dendritic cell is a CD141+ myeloid DC. In embodiments, the dendritic cell is a CD303+ plasmacytoid DC.
  • a plasmablast as provided herein is used according to its conventional meaning in the biological arts.
  • a plasmablast is a cell in a short-lived differentiation stage between a post germinal center B-cell and a mature plasma cell. Plasmablasts retain a proliferative capability together with an almost fully mature plasma cell phenotype.
  • a plasmablast is a CD19+, CD20-, IG+/-, CD27++ and CD38++ cell.
  • a plasma cell is a CD19+/-, CD20-, Ig-, CD27++, CD38+++ and CD138+ cell.
  • T cell detection agents refers to a chemical or molecular moiety capable of identifying T cells.
  • a T cell detection agent can be an antibody to a T cell specific surface maker (e.g. an antibody against CD3, and antibody against CD4, or an antibody against CD8).
  • T cell detection agents can be used alone or in combination.
  • T cell detection agents can further be detected by fluorescence activated cell sorting (FACS).
  • cell subset detection agent refers to a chemical or molecule detection agent that can be used to identify and distinguish a specific subset of cells (e.g. senescent cells, naive cells, effector cells, memory cells etc.).
  • Example cell subset detection agents include “naive cell detection agents”, “memory cell detection agents”, and “effector cell detection agent.”
  • Cell subset detection agents can include antibodies against distinguishing cell surface markers.
  • cell subset detection agents include antibodies against CD27 or antibodies against CD45RA.
  • “Selective” or “selectivity” or the like of a compound refers to the compound’s ability to discriminate between molecular targets (e.g. a compound having selectivity toward HMT SUV39H1 and/or HMT G9a).
  • An “inhibitor” refers to a compound (e.g. compounds described herein) that reduces activity when compared to a control, such as absence of the compound or a compound with known inactivity.
  • Contacting is used in accordance with its plain ordinary meaning and refers to the process of allowing at least two distinct species (e.g. chemical compounds including biomolecules or cells) to become sufficiently proximal to react, interact or physically touch.
  • species e.g. chemical compounds including biomolecules or cells
  • the resulting reaction product can be produced directly from a reaction between the added reagents or from an intermediate from one or more of the added reagents that can be produced in the reaction mixture.
  • contacting may include allowing two species to react, interact, or physically touch, wherein the two species may be a compound as described herein and a protein or enzyme. In some embodiments contacting includes allowing a compound described herein to interact with a protein or enzyme that is involved in a signaling pathway.
  • activation means positively affecting (e.g. increasing) the activity or function of the protein relative to the activity or function of the protein in the absence of the activator.
  • activation means positively affecting (e.g. increasing) the concentration or levels of the protein relative to the concentration or level of the protein in the absence of the activator.
  • the terms may reference activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein decreased in a disease.
  • activation may include, at least in part, partially or totally increasing stimulation, increasing or enabling activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein associated with a disease (e.g., a protein which is decreased in a disease relative to a non-diseased control).
  • Activation may include, at least in part, partially or totally increasing stimulation, increasing or enabling activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein
  • agonist refers to a substance capable of detectably increasing the expression or activity of a given gene or protein.
  • the agonist can increase expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the agonist.
  • expression or activity is 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold or higher than the expression or activity in the absence of the agonist.
  • the term “inhibition”, “inhibit”, “inhibiting” and the like in reference to a protein-inhibitor interaction means negatively affecting (e.g. decreasing) the activity or function of the protein relative to the activity or function of the protein in the absence of the inhibitor. In embodiments inhibition means negatively affecting (e.g. decreasing) the concentration or levels of the protein relative to the concentration or level of the protein in the absence of the inhibitor. In embodiments inhibition refers to reduction of a disease or symptoms of disease. In embodiments, inhibition refers to a reduction in the activity of a particular protein target.
  • inhibition includes, at least in part, partially or totally blocking stimulation, decreasing, preventing, or delaying activation, or inactivating, desensitizing, or down-regulating signal transduction or enzymatic activity or the amount of a protein.
  • inhibition refers to a reduction of activity of a target protein resulting from a direct interaction (e.g. an inhibitor binds to the target protein).
  • inhibition refers to a reduction of activity of a target protein from an indirect interaction (e.g. an inhibitor binds to a protein that activates the target protein, thereby preventing target protein activation).
  • inhibitor refers to a substance capable of detectably decreasing the expression or activity of a given gene or protein.
  • the antagonist can decrease expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the antagonist. In certain instances, expression or activity is 1.5-fold, 2-fold, 3- fold, 4-fold, 5-fold, 10-fold or lower than the expression or activity in the absence of the antagonist.
  • expression includes any step involved in the production of the polypeptide including, but not limited to, transcription, post-transcriptional modification, translation, post-translational modification, and secretion. Expression can be detected using conventional techniques for detecting protein (e.g., ELISA, Western blotting, flow cytometry, immunofluorescence, immunohistochemistry, etc.).
  • recombinant when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified.
  • recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
  • Transgenic cells and animals are those that express a heterologous gene or coding sequence, typically as a result of recombinant methods.
  • exogenous refers to a molecule or substance (e.g., a compound, nucleic acid or protein) that originates from outside a given cell or organism.
  • endogenous refers to a molecule or substance that is native to, or originates within, a given cell or organism.
  • modulator refers to a composition that increases or decreases the level of a target molecule or the function of a target molecule or the physical state of the target of the molecule relative to the absence of the modulator.
  • modulate is used in accordance with its plain ordinary meaning and refers to the act of changing or varying one or more properties. “Modulation” refers to the process of changing or varying one or more properties. For example, as applied to the effects of a modulator on a target protein, to modulate means to change by increasing or decreasing a property or function of the target molecule or the amount of the target molecule.
  • Control or “control experiment” is used in accordance with its plain ordinary meaning and refers to an experiment in which the subjects or reagents of the experiment are treated as in a parallel experiment except for omission of a procedure, reagent, or variable of the experiment. In some instances, the control is used as a standard of comparison in evaluating experimental effects. In some embodiments, a control is the measurement of the activity of a protein in the absence of a compound as described herein (including embodiments and examples).
  • a “standard control” as provided herein refers to a sample that serves as a reference, usually a known reference, for comparison to a test sample.
  • a test sample can be taken from a patient suspected of having a disease (e.g., Alzheimer’s disease, Parkinson’s disease) and compared to samples from a patient known to have the disease, or a known normal (non-disease) individual.
  • a control can also represent an average value gathered from a population of similar individuals, e.g., disease patients or healthy individuals with a similar medical background, same age, weight, etc.
  • a control value can also be obtained from the same individual, e.g., from an earlier-obtained sample, prior to disease, or prior to treatment.
  • controls can be designed for assessment of any number of parameters.
  • One of skill in the art will understand which controls are valuable in a given situation and be able to analyze data based on comparisons to control values. Controls are also valuable for determining the significance of data. For example, if values for a given parameter are widely variant in controls, variation in test samples will not be considered as significant.
  • a standard control is a level of phosphorylation of an intracellular signaling molecule from a sample or subject lacking the disease, a sample or subject at a selected stage of the disease or disease state, or in the absence of a particular variable such as a therapeutic agent.
  • the control includes a known level of phosphorylation of an intracellular signaling molecule. Such a known level correlates with an average level of subjects lacking the disease, at a selected stage of the disease or disease state, or in the absence of a particular variable such as a therapeutic agent.
  • a control also includes the level of phosphorylation of an intracellular signaling molecule from one or more selected samples or subjects as described herein.
  • a control includes the level of phosphorylation of an intracellular signaling molecule in a sample from a subject that does not have the disease, is at a selected stage of disease or disease state, or has not received treatment for the disease.
  • Another exemplary control level includes an assessment of the level of phosphorylation of an intracellular signaling molecule in samples taken from multiple subjects that do not have the disease, are at a selected stage of the disease, or have not received treatment for the disease.
  • control level of phosphorylation of an intracellular signaling molecule includes the level of phosphorylation of an intracellular signaling molecule in a sample or subject in the absence of a therapeutic agent
  • control sample or subject is optionally the same sample or subject to be tested before or after treatment with a therapeutic agent or is a selected sample or subject in the absence of the therapeutic agent.
  • a standard control is an average expression level calculated from a number of subjects without a particular disease.
  • a control level also includes a known control level or value known in the art.
  • a “standard control” as used herein in reference to the level of phosphorylation of one or more intracellular signaling molecules refers to the level measured in a control subject (e.g. in a sample from the control subject) or population of control subjects.
  • the control subject is a healthy control subject relative to the subject being tested, wherein the healthy control subject does not have a neurological disease.
  • the control subject is the test subject prior to treatment of the test subject, wherein the test subject and control subject have a neurological disease.
  • the test subject has been treated for a neurological disease with a neurological disease treatment and the control subject is the test subject prior to treatment.
  • the population of control subjects is a diverse collection of healthy subjects and diseased subjects, wherein the level of phosphorylation of an intracellular signaling molecule in the test subject is compared to the levels of the population of control subjects (e.g. an average of phosphorylation levels of the population of control subjects).
  • the population of control subjects is a collection of healthy subjects that do not have a neurological disease, wherein the level of the test subject is compared to the levels of the population of control subjects (e.g. an average of expression levels of the population of control subjects).
  • the population of control subjects is a collection of subjects that have been treated for a neurological disease, wherein the level of the test subject is compared to the levels of the population of control subjects (e.g. an average of levels of the population of control subjects).
  • the control subject and the test subject are the same.
  • the standard control is the level of phosphorylation of an intracellular signaling molecule in a sample from healthy tissue.
  • sample refers to a composition that is obtained or derived from a subject and/or individual of interest that contains a cellular and/or other molecular entity that is to be characterized and/or identified, for example based on physical, biochemical, chemical and/or physiological characteristics.
  • disease sample and variations thereof refers to any sample obtained from a subject of interest that would be expected or is known to contain the cellular and/or molecular entity that is to be characterized.
  • tissue extracts such as homogenized tissue, tumor tissue, cellular extracts, primary or cultured cells or cell lines, cell supernatants, cell lysates, platelets, serum, plasma, vitreous fluid, lymph fluid, synovial fluid, follicular fluid, seminal fluid, amniotic fluid, milk, whole blood, blood-derived cells, urine, cerebro spinal fluid, saliva, sputum, tears, perspiration, mucus, tumor lysates, and tissue culture medium, and combinations thereof.
  • tissue sample or “cell sample” is meant a collection of similar cells obtained from a tissue of a subject or individual.
  • the source of the tissue or cell sample may be solid tissue as from a FFPE, FF, fresh, frozen, and/or preserved organ, tissue sample, biopsy, and/or aspirate; blood or any blood constituents such as plasma; bodily fluids such as cerebral spinal fluid, amniotic fluid, peritoneal fluid, or interstitial fluid; cells from any time in gestation or development of the subject.
  • the tissue sample may also be primary or cultured cells or cell lines.
  • the tissue or cell sample is obtained from a disease (e.g., cancer) tissue/organ.
  • the tissue sample may contain compounds which are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like.
  • a disease e.g. a protein associated disease, a cancer (e.g., cancer, inflammatory disease, autoimmune disease, or infectious disease)
  • a disease e.g. cancer, inflammatory disease, autoimmune disease, or infectious disease
  • the disease e.g. cancer, inflammatory disease, autoimmune disease, or infectious disease
  • a symptom of the disease is caused by (in whole or in part) the substance or substance activity or function.
  • a causative agent could be a target for treatment of the disease.
  • aberrant refers to different from normal. When used to describe enzymatic activity or protein function, aberrant refers to activity or function that is greater or less than a normal control or the average of normal non-diseased control samples. Aberrant activity may refer to an amount of activity that results in a disease, wherein returning the aberrant activity to a normal or non-disease-associated amount (e.g. by administering a compound or using a method as described herein), results in reduction of the disease or one or more disease symptoms.
  • Pathway refers to a set of system components involved in two or more sequential molecular interactions that result in the production of a product or activity.
  • a pathway can produce a variety of products or activities that can include, for example, intermolecular interactions, changes in expression of a nucleic acid or polypeptide, the formation or dissociation of a complex between two or more molecules, accumulation or destruction of a metabolic product, activation or deactivation of an enzyme or binding activity.
  • the term "pathway” includes a variety of pathway types, such as, for example, a biochemical pathway, a gene expression pathway, a regulatory pathway, or a combination thereof.
  • signaling pathway refers to a series of interactions between cellular and optionally extra-cellular components (e.g. proteins, nucleic acids, small molecules, ions, lipids) that conveys a change in one component to one or more other components, which in turn may convey a change to additional components, which is optionally propagated to other signaling pathway components.
  • extra-cellular components e.g. proteins, nucleic acids, small molecules, ions, lipids
  • diagnosis refers to an identification or likelihood of the presence of a particular type of neurological disease (e.g., Parkinson’s disease, Alzheimer’s disease) or outcome in a subject.
  • prognosis refers to the likelihood or risk of a subject developing a particular outcome or particular event (e.g., Parkinson’s disease, Alzheimer’s disease).
  • biological sample encompasses essentially any sample type obtained from a subject that can be used in a diagnostic or prognostic method described herein.
  • the biological sample may be any bodily fluid, tissue or any other suitable sample.
  • the definition encompasses blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom and the progeny thereof.
  • the definition also includes samples that have been manipulated in any way after their procurement, such as by treatment with reagents, solubilization, or enrichment for certain components, such as cells (e.g., cancer cells), polypeptides, or proteins.
  • biological sample encompasses a clinical sample, but also, includes cells in culture, cell supernatants, cell lysates, blood, serum, plasma, urine, cerebral spinal fluid, biological fluid, and tissue samples.
  • the sample may be pretreated as necessary by dilution in an appropriate buffer solution or concentrated, if desired. Any of a number of standard aqueous buffer solutions, employing one of a variety of buffers, such as phosphate, Tris, or the like, preferably at physiological pH can be used.
  • Biological samples can be derived from patients using well- known techniques such as venipuncture, lumbar puncture, fluid sample such as saliva or urine, or tissue biopsy and the like.
  • the sample is a blood sample (e.g., containing PBMCs).
  • neurological disorder refers to a disease or condition in which the function of a subject’s nervous system becomes impaired.
  • neurological diseases include Alexander's disease, Alper's disease, Alzheimer's disease, Amyotrophic lateral sclerosis, Ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), Bovine spongiform encephalopathy (BSE), Canavan disease, chronic fatigue syndrome, Cockayne syndrome, Corticobasal degeneration, Creutzfeldt-Jakob disease, frontotemporal dementia, Gerstmann- Straussler-Scheinker syndrome, Huntington's disease, HIV-associated dementia, Kennedy's disease, Krabbe's disease, kuru, Lewy body dementia, Machado-Joseph disease (Spinocerebellar ataxi
  • treating refers to any indicia of success in the therapy or amelioration of an injury, disease, pathology or condition (e.g., Parkinson’s disease, Alzheimer’s disease) including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the injury, pathology or condition more tolerable to the patient; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; improving a patient’s physical or mental well-being.
  • the treatment or amelioration of symptoms can be based on objective or subjective parameters; including the results of a physical examination, neuropsychiatric exams, and/or a psychiatric evaluation.
  • the term "treating" and conjugations thereof, may include prevention of an injury, pathology, condition, or disease.
  • treating is preventing.
  • treating does not include preventing.
  • Treating” or “treatment” as used herein also broadly includes any approach for obtaining beneficial or desired results in a subject’s condition (e.g., Parkinson’s disease, Alzheimer’s disease), including clinical results.
  • beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of the extent of a disease, stabilizing (i.e., not worsening) the state of disease, prevention of a disease’s transmission or spread, delay or slowing of disease progression, amelioration or palliation of the disease state, diminishment of the reoccurrence of disease, and remission, whether partial or total and whether detectable or undetectable.
  • treatment includes any cure, amelioration, or prevention of a disease. Treatment may prevent the disease from occurring; inhibit the disease’s spread; relieve the disease’s symptoms, fully or partially remove the disease’s underlying cause, shorten a disease’s duration, or do a combination of these things.
  • Treating” and “treatment” as used herein include prophylactic treatment.
  • Treatment methods include administering to a subject a therapeutically effective amount of an active agent.
  • the administering step may consist of a single administration or may include a series of administrations.
  • the length of the treatment period depends on a variety of factors, such as the severity of the condition, the age of the patient, the concentration of active agent, the activity of the compositions used in the treatment, or a combination thereof.
  • the effective dosage of an agent used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art.
  • chronic administration may be required.
  • the compositions are administered to the subject in an amount and for a duration sufficient to treat the patient.
  • the treating or treatment is no prophylactic treatment.
  • prevention refers to a decrease in the occurrence of disease symptoms in a patient. As indicated above, the prevention may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment.
  • “Patient” or “subject in need thereof’ refers to a living organism suffering from or prone to a disease or condition that can be treated by administration of a pharmaceutical composition as provided herein.
  • Non-limiting examples include humans, other mammals, bovines, rats, mice, dogs, monkeys, goat, sheep, cows, deer, and other non-mammalian animals.
  • a patient is human.
  • a “effective amount” is an amount sufficient for a compound to accomplish a stated purpose relative to the absence of the compound (e.g. achieve the effect for which it is administered, treat a disease, reduce enzyme activity, increase enzyme activity, reduce a signaling pathway, or reduce one or more symptoms of a disease or condition).
  • An example of an “effective amount” is an amount sufficient to contribute to the treatment, prevention, or reduction of a symptom or symptoms of a disease, which could also be referred to as a “therapeutically effective amount.”
  • a “reduction” of a symptom or symptoms means decreasing of the severity or frequency of the symptom(s), or elimination of the symptom(s).
  • a “prophylactically effective amount” of a drug is an amount of a drug that, when administered to a subject, will have the intended prophylactic effect, e.g., preventing or delaying the onset (or reoccurrence) of an injury, disease, pathology or condition, or reducing the likelihood of the onset (or reoccurrence) of an injury, disease, pathology, or condition, or their symptoms.
  • the full prophylactic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses.
  • a prophylactically effective amount may be administered in one or more administrations.
  • An “activity decreasing amount,” as used herein, refers to an amount of antagonist required to decrease the activity of an enzyme relative to the absence of the antagonist.
  • a “function disrupting amount,” as used herein, refers to the amount of antagonist required to disrupt the function of an enzyme or protein relative to the absence of the antagonist. The exact amounts will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); and Remington: The Science and Practice of Pharmacy, 20th Edition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins).
  • the therapeutically effective amount can be initially determined from cell culture assays.
  • Target concentrations will be those concentrations of active compound(s) that are capable of achieving the methods described herein, as measured using the methods described herein or known in the art.
  • therapeutically effective amounts for use in humans can also be determined from animal models.
  • a dose for humans can be formulated to achieve a concentration that has been found to be effective in animals.
  • the dosage in humans can be adjusted by monitoring compounds effectiveness and adjusting the dosage upwards or downwards, as described above. Adjusting the dose to achieve maximal efficacy in humans based on the methods described above and other methods is well within the capabilities of the ordinarily skilled artisan.
  • a therapeutically effective amount refers to that amount of the therapeutic agent (e.g., a Parkinson’s disease therapeutic agent, Alzheimer’s disease therapeutic agent) sufficient to ameliorate the disorder, as described above.
  • a therapeutically effective amount will show an increase or decrease of at least 5%, 10%, 15%, 20%, 25%, 40%, 50%, 60%, 75%, 80%, 90%, or at least 100%.
  • Therapeutic efficacy can also be expressed as “-fold” increase or decrease.
  • a therapeutically effective amount can have at least a 1.2-fold, 1.5-fold, 2-fold, 5-fold, or more effect over a control.
  • Dosages may be varied depending upon the requirements of the patient and the compound being employed.
  • the dose administered to a patient should be sufficient to effect a beneficial therapeutic response in the patient over time.
  • the size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached. Dosage amounts and intervals can be adjusted individually to provide levels of the administered compound effective for the particular clinical indication being treated. This will provide a therapeutic regimen that is commensurate with the severity of the individual's disease state.
  • administering means oral administration, administration as a suppository, topical contact, intravenous, parenteral, intraperitoneal, intramuscular, intralesional, intrathecal, intranasal or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject.
  • Administration is by any route, including parenteral and transmucosal (e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal).
  • Parenteral administration includes, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial.
  • Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc.
  • the administering does not include administration of any active agent other than the recited active agent.
  • compositions described herein are administered at the same time, just prior to, or just after the administration of one or more additional therapies.
  • the compounds provided herein can be administered alone or can be coadministered to the patient. Coadministration is meant to include simultaneous or sequential administration of the compounds individually or in combination (more than one compound).
  • the preparations can also be combined, when desired, with other active substances (e.g. to reduce metabolic degradation).
  • the compositions of the present disclosure can be delivered transdermally, by a topical route, or formulated as applicator sticks, solutions, suspensions, emulsions, gels, creams, ointments, pastes, jellies, paints, powders, and aerosols.
  • the methods provided herein are, inter alia, useful for diagnosing neurological disease (e.g., Alzheimer’s Disease, Parkinson’s disease).
  • the methods provided herein may be used to detect specific phosphorylation levels of an intracellular signaling molecules such as, for example, PLC-y2, AKT, STAT1 or STAT5, in a subject having or being at risk of developing a neurological disease (e.g., Alzheimers disease or Parkinson’s disease).
  • the phosphorylation levels of intracellular signaling molecules are detected in peripheral blood mononuclear cells (PBMCs) and subpopulations thereof, which have been stimulated with a stimulatory agent ex vivo (in, for example, a cell culture container after the PBMCs have been isolated from a subject).
  • PBMCs peripheral blood mononuclear cells
  • Stimulatory agents include, for example, interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA).
  • IFN-a interferon a
  • IL-6 interleukin-6
  • IL-7 interleukin-7
  • IL-10 interleukin- 10
  • IL-21 interleukin-21
  • LPS lipopoly saccharides
  • PMA phorbol myristate acetate
  • a “level of phosphorylation” as provided herein and in reference to intracellular signaling molecules is a detectable amount of phosphorylated intracellular signaling molecules or a detectable amount of phosphorylation of intracellular signaling molecules that are present in a PBMC after said PBMC has been contacted with a stimulatory agent.
  • a level of phosphorylation may be increased relative to a standard control or it may be decreased relative to a standard control.
  • the level of phosphorylation of an intracellular signaling molecule is increased relative to a standard control.
  • the level of phosphorylation of an intracellular signaling molecule is decreased relative to a standard control.
  • the standard control is a level of phosphorylation of an intracellular signaling molecule in the absence of a stimulatory agent. In embodiments, the level of phosphorylation of an intracellular signaling molecule is increased subsequent to the contacting of the PBMC with the stimulatory agent. In embodiments, the level of phosphorylation of an intracellular signaling molecule is decreased subsequent to the contacting of the PBMC with the stimulatory agent.
  • An “intracellular signaling molecule” as provided herein refers to a protein inside a cell that forms part of a cellular signaling pathway. In embodiments, the intracellular signaling molecule is PLC-y2. AKT, STAT1 or STAT5. In embodiments, the intracellular signaling molecule is PLC-y2.
  • the intracellular signaling molecule is AKT. In embodiments, the intracellular signaling molecule is STAT1. In embodiments, the intracellular signaling molecule is STAT5. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2, phosphorylated AKT, phosphorylated STAT1 or phosphorylated STAT5. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2. In embodiments, the intracellular signaling molecule is phosphorylated AKT. In embodiments, the intracellular signaling molecule is phosphorylated STAT1. In embodiments, the intracellular signaling molecule is phosphorylated STAT5.
  • the “expression level,” “amount,” or “level,” are used herein interchangeably, of a biomarker is a detectable level in a biological sample. “Expression” generally refers to the process by which information (e.g., gene-encoded and/or epigenetic) is converted into the structures present and operating in the cell. Therefore, as used herein, “expression” may refer to transcription into a polynucleotide, translation into a polypeptide, or even polynucleotide and/or polypeptide modifications (e.g., posttranslational modification of a polypeptide).
  • Fragments of the transcribed polynucleotide, the translated polypeptide, or polynucleotide and/or polypeptide modifications shall also be regarded as expressed whether they originate from a transcript generated by alternative splicing or a degraded transcript, or from a post-translational processing of the polypeptide, e.g., by proteolysis.
  • “Expressed genes” include those that are transcribed into a polynucleotide as mRNA and then translated into a polypeptide, and also those that are transcribed into RNA but not translated into a polypeptide (for example, transfer and ribosomal RNAs).
  • Expression levels can be measured by methods known to one skilled in the art and also disclosed herein.
  • the expression level or amount of a biomarker can be used to identify/characterize a subject having a neurological disease.
  • the expression level or amount of a biomarker provided herein in a subject having a neurological disease described herein can also be used to determine and/or track the benefit of an administered neurological therapy over time.
  • the biomarker is a level of phosphorylation of an intracellular signaling molecule.
  • biomarker refers to an indicator, e.g., a predictive, prognostic, and/or a pharmacodynamic indicator which can be detected in a sample (e.g., a tissue sample, e.g., a peripheral blood sample).
  • the biomarker may serve as an indicator of a particular type of a neurological disease or disorder (e.g., Parkinson’s disease, Alzheimer’s disease) characterized by certain molecular, pathological, histological, and/or clinical features.
  • a biomarker is a level of phosphorylation of an intracellular signaling molecule. Exemplary sets of biomarkers are found in Table 7.
  • a “stimulatory agent” as provided herein refers to an agent (e.g., interferon a (IFN- a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA) that positively affects (e.g. increases) the level of phosphorylation of an intracellular signaling molecule (e.g., PLC-y2, AKT, STAT1 or STAT5) relative to the level of phosphorylation of the intracellular signaling molecule in the absence of the agent.
  • stimulating means positively affecting (e.g.
  • stimulating means positively affecting (e.g. increasing) the amount of phosphorylated intracellular signaling molecules relative to the amount of phosphorylated intracellular signaling molecules in the absence of the agent.
  • the stimulatory agent can increase the level of phosphorylation of an intracellular signaling molecule or the amount of phosphorylated intracellular signaling molecule 10%, 20%, 30%, 40%, 50%, 60%, 70%,
  • level or amount is 1.5-fold, 2-fold, 3-fold, 4-fold, 5- fold, 10-fold or higher than the level or amount in the absence of the stimulatory agent.
  • the stimulatory agent is any one of the agents listed in Table 5. In embodiments, the stimulatory agent is any one of the agents listed in Table 5 and is administered at a concentration as set forth in Table 5.
  • the stimulatory agent is IFN-a.
  • IFN-a is administered at a concentration of about 10,000 units/ ml. In embodiments, IFN-a is administered at a concentration of 10,000 units/ ml. In embodiments, IFN-a is administered at a concentration of about 5,000 units/ ml. In embodiments, IFN-a is administered at a concentration of 5,000 units/ ml. In embodiments, IFN-a is administered at a concentration of about 15,000 units/ ml. In embodiments, IFN-a is administered at a concentration of 15,000 units/ ml. In embodiments, IFN-a is administered at a concentration of 15,000 units/ ml. In embodiments, IFN-a is administered at a concentration of about 20,000 units/ ml.
  • IFN-a is administered at a concentration of 20,000 units/ ml. In embodiments, IFN-a is administered at a concentration from about 5,000 units/ ml to about 20,000 units/ ml. In embodiments, IFN-a is administered at a concentration from 5,000 units/ ml to 20,000 units/ ml.
  • the stimulatory agent is IL-6.
  • IL-6 is administered at a concentration of about 50 ng/ml. In embodiments, IL-6 is administered at a concentration of 50 ng/ml. In embodiments, IL-6 is administered at a concentration of about 10 ng/ml. In embodiments, IL-6 is administered at a concentration of 10 ng/ml. In embodiments, IL-6 is administered at a concentration of about 15 ng/ml. In embodiments, IL-6 is administered at a concentration of 15 ng/ml. In embodiments, IL-6 is administered at a concentration of about 20 ng/ml. In embodiments, IL-6 is administered at a concentration of 20 ng/ml.
  • IL-6 is administered at a concentration of about 25 ng/ml. In embodiments, IL-6 is administered at a concentration of 25 ng/ml. In embodiments, IL-6 is administered at a concentration of about 30 ng/ml. In embodiments, IL-6 is administered at a concentration of 30 ng/ml. In embodiments, IL-6 is administered at a concentration of about 35 ng/ml. In embodiments, IL-6 is administered at a concentration of 35 ng/ml. In embodiments, IL-6 is administered at a concentration of about 40 ng/ml. In embodiments, IL-6 is administered at a concentration of 40 ng/ml.
  • IL-6 is administered at a concentration of about 45 ng/ml. In embodiments, IL-6 is administered at a concentration of 45 ng/ml. In embodiments, IL-6 is administered at a concentration of about 55 ng/ml. In embodiments, IL-6 is administered at a concentration of 55 ng/ml. In embodiments, IL-6 is administered at a concentration of about 60 ng/ml. In embodiments, IL-6 is administered at a concentration of 60 ng/ml. In embodiments, IL-6 is administered at a concentration of about 65 ng/ml. In embodiments, IL-6 is administered at a concentration of 65 ng/ml.
  • IL-6 is administered at a concentration of about 70 ng/ml. In embodiments, IL-6 is administered at a concentration of 70 ng/ml. In embodiments, IL-6 is administered at a concentration of about 80 ng/ml. In embodiments, IL-6 is administered at a concentration of 80 ng/ml. In embodiments, IL-6 is administered at a concentration of about 90 ng/ml. In embodiments, IL-6 is administered at a concentration of 90 ng/ml. In embodiments, IL-6 is administered at a concentration of about 100 ng/ml. In embodiments, IL-6 is administered at a concentration of 100 ng/ml. In embodiments, IL-6 is administered at a concentration from about 10 ng/ml to about 100 ng/ml. In embodiments, IL-6 is administered at a concentration from about 10 ng/ml to 100 ng/ml. In embodiments, IL-6 is administered at a concentration from about 10 ng/ml to 100 ng/m
  • the stimulatory agent is IL-7.
  • IL-7 is administered at a concentration of about 50 ng/ml. In embodiments, IL-7 is administered at a concentration of 50 ng/ml. In embodiments, IL-7 is administered at a concentration of about 10 ng/ml. In embodiments, IL-7 is administered at a concentration of 10 ng/ml. In embodiments, IL-7 is administered at a concentration of about 15 ng/ml. In embodiments, IL-7 is administered at a concentration of 15 ng/ml. In embodiments, IL-7 is administered at a concentration of about 20 ng/ml. In embodiments, IL-7 is administered at a concentration of 20 ng/ml.
  • IL-7 is administered at a concentration of about 25 ng/ml. In embodiments, IL-7 is administered at a concentration of 25 ng/ml. In embodiments, IL-7 is administered at a concentration of about 30 ng/ml. In embodiments, IL-7 is administered at a concentration of 30 ng/ml. In embodiments, IL-7 is administered at a concentration of about 35 ng/ml. In embodiments, IL-7 is administered at a concentration of 35 ng/ml. In embodiments, IL-7 is administered at a concentration of about 40 ng/ml. In embodiments, IL-7 is administered at a concentration of 40 ng/ml.
  • IL-7 is administered at a concentration of about 45 ng/ml. In embodiments, IL-7 is administered at a concentration of 45 ng/ml. In embodiments, IL-7 is administered at a concentration of about 55 ng/ml. In embodiments, IL-7 is administered at a concentration of 55 ng/ml. In embodiments, IL-7 is administered at a concentration of about 60 ng/ml. In embodiments, IL-7 is administered at a concentration of 60 ng/ml. In embodiments, IL-7 is administered at a concentration of about 65 ng/ml. In embodiments, IL-7 is administered at a concentration of 65 ng/ml.
  • IL-7 is administered at a concentration of about 70 ng/ml. In embodiments, IL-7 is administered at a concentration of 70 ng/ml. In embodiments, IL-7 is administered at a concentration of about 80 ng/ml. In embodiments, IL-7 is administered at a concentration of 80 ng/ml. In embodiments, IL-7 is administered at a concentration of about 90 ng/ml. In embodiments, IL-7 is administered at a concentration of 90 ng/ml. In embodiments, IL-7 is administered at a concentration of about 100 ng/ml. In embodiments, IL-7 is administered at a concentration of 100 ng/ml. In embodiments, IL-7 is administered at a concentration from about 10 ng/ml to about 100 ng/ml. In embodiments, IL-7 is administered at a concentration from about 10 ng/ml to 100 ng/ml. In embodiments, IL-7 is administered at a concentration from about 10 ng/ml to 100 ng/m
  • the stimulatory agent is IL-10.
  • IL-10 is administered at a concentration of about 50 ng/ml. In embodiments, IL-10 is administered at a concentration of 50 ng/ml. In embodiments, IL-10 is administered at a concentration of about 10 ng/ml. In embodiments, IL-10 is administered at a concentration of 10 ng/ml. In embodiments, IL-10 is administered at a concentration of about 15 ng/ml. In embodiments, IL-10 is administered at a concentration of 15 ng/ml. In embodiments, IL-10 is administered at a concentration of about 20 ng/ml. In embodiments, IL-10 is administered at a concentration of 20 ng/ml.
  • IL-10 is administered at a concentration of about 25 ng/ml. In embodiments, IL-10 is administered at a concentration of 25 ng/ml. In embodiments, IL-10 is administered at a concentration of about 30 ng/ml. In embodiments, IL-10 is administered at a concentration of 30 ng/ml. In embodiments, IL-10 is administered at a concentration of about 35 ng/ml. In embodiments, IL-10 is administered at a concentration of 35 ng/ml. In embodiments, IL-10 is administered at a concentration of about 40 ng/ml. In embodiments, IL-10 is administered at a concentration of 40 ng/ml.
  • IL-10 is administered at a concentration of about 45 ng/ml. In embodiments, IL-10 is administered at a concentration of 45 ng/ml. In embodiments, IL-10 is administered at a concentration of about 55 ng/ml. In embodiments, IL-10 is administered at a concentration of 55 ng/ml. In embodiments, IL-10 is administered at a concentration of about 60 ng/ml. In embodiments, IL-10 is administered at a concentration of 60 ng/ml. In embodiments, IL-10 is administered at a concentration of about 65 ng/ml. In embodiments, IL-10 is administered at a concentration of 65 ng/ml.
  • IL-10 is administered at a concentration of about 70 ng/ml. In embodiments, IL-10 is administered at a concentration of 70 ng/ml. In embodiments, IL-10 is administered at a concentration of about 80 ng/ml. In embodiments, IL-10 is administered at a concentration of 80 ng/ml. In embodiments, IL-10 is administered at a concentration of about 90 ng/ml. In embodiments, IL-10 is administered at a concentration of 90 ng/ml. In embodiments, IL-10 is administered at a concentration of about 100 ng/ml. In embodiments, IL-10 is administered at a concentration of 100 ng/ml. In embodiments, IL-10 is administered at a concentration from about 10 ng/ml to about 100 ng/ml. In embodiments, IL-10 is administered at a concentration from about 10 ng/ml to 100 ng/ml. In embodiments, IL-10 is administered at a concentration from about 10 ng/ml to 100 ng/m
  • the stimulatory agent is IL-21.
  • IL-21 is administered at a concentration of about 50 ng/ml. In embodiments, IL-21 is administered at a concentration of 50 ng/ml. In embodiments, IL-21 is administered at a concentration of about 10 ng/ml. In embodiments, IL-21 is administered at a concentration of 10 ng/ml. In embodiments, IL-21 is administered at a concentration of about 15 ng/ml. In embodiments, IL-21 is administered at a concentration of 15 ng/ml. In embodiments, IL-21 is administered at a concentration of about 20 ng/ml. In embodiments, IL-21 is administered at a concentration of 20 ng/ml.
  • IL-21 is administered at a concentration of about 25 ng/ml. In embodiments, IL-21 is administered at a concentration of 25 ng/ml. In embodiments, IL-21 is administered at a concentration of about 30 ng/ml. In embodiments, IL-21 is administered at a concentration of 30 ng/ml. In embodiments, IL-21 is administered at a concentration of about 35 ng/ml. In embodiments, IL-21 is administered at a concentration of 35 ng/ml. In embodiments, IL-21 is administered at a concentration of about 40 ng/ml. In embodiments, IL-21 is administered at a concentration of 40 ng/ml.
  • IL-21 is administered at a concentration of about 45 ng/ml. In embodiments, IL-21 is administered at a concentration of 45 ng/ml. In embodiments, IL-21 is administered at a concentration of about 55 ng/ml. In embodiments, IL-21 is administered at a concentration of 55 ng/ml. In embodiments, IL-21 is administered at a concentration of about 60 ng/ml. In embodiments, IL-21 is administered at a concentration of 60 ng/ml. In embodiments, IL-21 is administered at a concentration of about 65 ng/ml. In embodiments, IL-21 is administered at a concentration of 65 ng/ml.
  • IL-21 is administered at a concentration of about 70 ng/ml. In embodiments, IL-21 is administered at a concentration of 70 ng/ml. In embodiments, IL-21 is administered at a concentration of about 80 ng/ml. In embodiments, IL-21 is administered at a concentration of 80 ng/ml. In embodiments, IL-21 is administered at a concentration of about 90 ng/ml. In embodiments, IL-21 is administered at a concentration of 90 ng/ml. In embodiments, IL-21 is administered at a concentration of about 100 ng/ml. In embodiments, IL-21 is administered at a concentration of 100 ng/ml. In embodiments, IL-21 is administered at a concentration from about 10 ng/ml to about 100 ng/ml. In embodiments, IL-21 is administered at a concentration from about 10 ng/ml to 100 ng/ml. In embodiments, IL-21 is administered at a concentration from about 10 ng/ml to 100 ng/m
  • the stimulatory agent is LPS.
  • LPS is administered at a concentration of about 1 ug/ml. In embodiments, LPS is administered at a concentration of 1 ug/ml. In embodiments, LPS is administered at a concentration of about 2 ug/ml. In embodiments, LPS is administered at a concentration of 2 ug/ml. In embodiments, LPS is administered at a concentration of about 3 ug/ml. In embodiments, LPS is administered at a concentration of 3 ug/ml. In embodiments, LPS is administered at a concentration of about 4 ug/ml. In embodiments, LPS is administered at a concentration of 4 ug/ml.
  • LPS is administered at a concentration of about 5 ug/ml. In embodiments, LPS is administered at a concentration of 5 ug/ml. In embodiments, LPS is administered at a concentration of about 10 ug/ml. In embodiments, LPS is administered at a concentration of 10 ug/ml. In embodiments, LPS is administered at a concentration from about 1 ug/ml to about 10 ug/ml. In embodiments, LPS is administered at a concentration from 1 ug/ml to 10 ug/ml.
  • the stimulatory agent is PMA.
  • PMA is administered at a concentration of about 100 ng/ml. In embodiments, PMA is administered at a concentration of 100 ng/ml. In embodiments, PMA is administered at a concentration of about 10 ng/ml. In embodiments, PMA is administered at a concentration of 10 ng/ml. In embodiments, PMA is administered at a concentration of about 50 ng/ml. In embodiments, PMA is administered at a concentration of 50 ng/ml. In embodiments, PMA is administered at a concentration of about 150 ng/ml. In embodiments, PMA is administered at a concentration of 150 ng/ml. In embodiments, PMA is administered at a concentration from about 10 ng/ml to about 150 ng/ml. In embodiments, PMA is administered at a concentration from 10 ng/ml to 150 ng/ml. In embodiments, PMA is administered at a concentration from 10 ng/ml to 150 ng/ml. In
  • a method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease includes (i) obtaining or having obtained a sample from a subject having or being at risk of developing a neurological disease; (ii) isolating a peripheral blood mononuclear cell (PBMC) from the sample; wherein the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC); (iii) contacting the PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10
  • a method of treating a neurological disease in a subject in need thereof includes (i) obtaining or having obtained a sample from a subject having a neurological disease; (ii) isolating a peripheral blood mononuclear cell (PBMC) from the sample; wherein the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC); (iii) contacting the PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myr
  • IFN-a interferon a
  • a method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject undergoing treatment for a neurological disease includes (i) obtaining or having obtained a sample from a subject undergoing treatment for a neurological disease; (ii) isolating a peripheral blood mononuclear cell (PBMC) from the sample; wherein the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC); (iii) contacting the PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21
  • IFN-a interferon a
  • a method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease including: (i) obtaining or having obtained a sample from a subject having or being at risk of developing a neurological disease; (ii) isolating a peripheral blood mononuclear cell (PBMC) from the sample; wherein the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC); (iii) contacting the PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10),
  • IFN-a interferon
  • the PBMC is a CD4+ T cell. In embodiments, the PBMC is a CD8+ T cell. In embodiments, the PBMC is a natural killer T cell. In embodiments, the PBMC is a natural killer (NK) cell. In embodiments, the PBMC is a B cell. In embodiments, the PBMC is a monocyte. In embodiments, the PBMC is a basophil. In embodiments, the PBMC is a plasmablast. In embodiments, the PBMC is a dendritic cell (DC).
  • DC dendritic cell
  • the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast and a dendritic cell (DC).
  • a CD4+ T cell a CD8+ T cell
  • a natural killer T cell a natural killer (NK) cell
  • B cell a monocyte, a basophil, a plasmablast and a dendritic cell (DC).
  • DC dendritic cell
  • the stimulatory agent is interferon a (IFN-a).
  • the stimulatory agent is interleukin-6 (IL-6).
  • the stimulatory agent is interleukin-7 (IL-7).
  • the stimulatory agent is interleukin- 10 (IL-10).
  • the stimulatory agent is interleukin-21 (IL-21).
  • the stimulatory agent is lipopolysaccharides (LPS).
  • the stimulatory agent is phorbol myristate acetate (PMA).
  • the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL- 21), lipopolysaccharides (LPS) and phorbol myristate acetate (PMA).
  • IFN-a interferon a
  • IL-6 interleukin-6
  • IL-7 interleukin-7
  • IL-10 interleukin- 10
  • IL-21 interleukin-21
  • LPS lipopolysaccharides
  • PMA phorbol myristate acetate
  • the PBMCs are contacted with the stimulatory agent in a non-natural environment, such as an in vitro cell culture container.
  • the cell culture container e.g., petri dish
  • the intracellular signaling molecule is PLC-y2. In embodiments, the intracellular signaling molecule is AKT. In embodiments, the intracellular signaling molecule is STAT1. In embodiments, the intracellular signaling molecule is STAT5. In embodiments, the intracellular signaling molecule is PLC-y2, AKT, STAT1 and STAT5. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2. In embodiments, the intracellular signaling molecule is phosphorylated AKT. In embodiments, the intracellular signaling molecule is phosphorylated STAT1. In embodiments, the intracellular signaling molecule is phosphorylated STAT5.
  • the intracellular signaling molecule is phosphorylated PLC-y2, phosphorylated AKT, phosphorylated STAT1 and phosphorylated STAT5.
  • an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the method includes based at least in part on the level of phosphorylation in step (v) administering a neurological disease treatment to the subject.
  • the neurological disease treatment is an Alzheimer’s disease treatment.
  • the neurological disease treatment is a dementia treatment.
  • the neurological disease treatment is a Parkinson’s disease treatment.
  • the PBMCs are contacted with a non stimulatory (inert, non-active) agent (e.g., a cell culture buffer).
  • a non stimulatory agent as provided herein is an agent that upon contacting with the PBMC does not detectably increase or decrease the level of phosphorylation of an intracellular signaling molecule provided herein.
  • the level of phosphorylation of the intracellular signaling molecule in presence of a non-stimulatory agent is the same as the level of phosphorylation of the intracellular signaling molecule in the absence of the non-stimulatory agent.
  • the intracellular signaling molecule is PLC-y2. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell.
  • the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL- 10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
  • the stimulatory agent is unstimulated and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell.
  • the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
  • the stimulatory agent is IL-10 and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell.
  • the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
  • the stimulatory agent is IL-21 and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell.
  • the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
  • the stimulatory agent is LPS and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell.
  • the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
  • the stimulatory agent is IL-6 and the PBMC is a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
  • the stimulatory agent is IL-6 and the PBMC is a CD8+ activated T cell.
  • the stimulatory agent is IL- 6 and the PBMC is a CD4+ CD8+ T cell or a natural killer T cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD56bright NK cell, or a CD16high monocyte.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a natural killer T cell.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD8+ activated T cell or a CD16high monocyte.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, or a CD56bright NK cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD56bright NK cell, or a CD16high monocyte.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a natural killer T cell.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD8+ activated T cell or a CD16high monocyte.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, or a CD56bright NK cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD56bright NK cell, or a CD16high monocyte.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a natural killer T cell.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD8+ activated T cell or a CD16high monocyte.
  • the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, or a CD56bright NK cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the intracellular signaling molecule is STAT1. In embodiments, the intracellular signaling molecule is phosphorylated STAT1.
  • the stimulatory agent is IFN-a and the PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD56bright NK cell, a myeloid dendritic cell (mDC) or a plasmacytoid dendritic cell (pDC).
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the stimulatory agent is LPS and the PBMC is a CD8+ central memory T cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the intracellular signaling molecule is STAT5.
  • the intracellular signaling molecule is phosphorylated STAT5.
  • the stimulatory agent is IFN-a and the PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, a CD16high monocyte or a CD161ow monocyte.
  • the stimulatory agent is IFN-a and the PBMC is a CD16high monocyte or a CD161ow monocyte.
  • the stimulatory agent is IFN-a and the PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, or a CD4- CD8- T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, or a CD8+ effector memory T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the stimulatory agent is LPS and the PBMC is a CD4+ CD8+ T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the subject is a male subject.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dimNK cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is unstimulated and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-10 and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dimNK cell, a CD16high monocyte or a CD161ow monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-21 and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dimNK cell, a CD16high monocyte or a CD161ow monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is LPS and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
  • the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IFN-a, IL-6 or IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-6 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is PMA and the PBMC is a CD4+ activated T cell, a CD4+ naive T cell, a CD8+ activated T cell, a natural killer T cell, a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a B switched memory cell, or a B translational cell.
  • the stimulatory agent is PMA and the PBMC is a CD4+ activated T cell, a CD4+ naive T cell, or a natural killer T cell.
  • the stimulatory agent is PMA and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a B switched memory cell, or a B translational cell.
  • the stimulatory agent is PMA and the PBMC is a CD8+ activated T cell or a CD4+ CD8+ T cell.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the intracellular signaling molecule is AKT. In embodiments, the intracellular signaling molecule is phosphorylated AKT.
  • the subject is a male subject.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-6 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-10 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-21 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is LPS and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-6 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-10 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-21 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is LPS and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell.
  • the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the stimulatory agent is PMA and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a mDC, a pDC, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
  • the subject is a female subject.
  • the stimulatory agent is unstimulated, IL-7, IL-10 or LPS and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is unstimulated and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-7 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-10 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is LPS and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is unstimulated and the PBMC is a B naive cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is unstimulated, IL-7, IL-10 or LPS and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is unstimulated and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-7 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-10 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is LPS and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a CD4- CD8- T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a mDC, a pDC, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, or a pDC.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD4+ CD8+ T cell, a CD4- CD8- T cell, a natural killer T cell, a plasmablast cell, or a CD56bright NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a mDC, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject. [0199] In embodiments, the stimulatory agent is IL-6 and the PBMC is an IgD- CD27- B cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, or a B naive cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-21 and the PBMC is an IgD- CD27- B cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the subject is a female subject.
  • the intracellular signaling molecule is STAT5.
  • the intracellular signaling molecule is phosphorylated STAT5.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD8+ effector memory T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ effector T cell, a CD8+ central memory T cell, a CD8+ naive T cell, or a natural killer T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ effector T cell, a CD4- CD8- T cell, a CD4+ CD8+ T cell, a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD4- CD8- T cell, a CD4+ CD8+ T cell or a natural killer T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD8+ central memory T cell, or a CD8+ effector memory T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the standard control is a healthy female subject.
  • the stimulatory agent is IL-7 and the PBMC is a natural killer T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the stimulatory agent is IL-7 and the PBMC is a CD8+ effector T cell, a CD4- CD8- T cell, or a CD4+ CD8+ T cell.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the intracellular signaling molecule is PLC-y2. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2. [0213] In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-10, IL-21 or LPS and the PBMC is a basophil.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-10 and the PBMC is a basophil.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-21 and the PBMC is a basophil.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS and the PBMC is a basophil.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the intracellular signaling molecule is STAT1.
  • the stimulatory agent is IFN-a and the PBMC is a plasmablast or CD56bright NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the intracellular signaling molecule is STAT5.
  • the stimulatory agent is IFN-a and the PBMC is a CD16high monocyte or a CD161ow monocyte.
  • the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the neurological disease is Alzheimer’s disease.
  • the intracellular signaling molecule is PLC-y2.
  • the stimulatory agent is IL-10, IL-21, LPS or PMA and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-10 and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-21 and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is PMA and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-10, IL-21, LPS or PMA and the PBMC is an IgA- B cell or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease. [0230] In embodiments, the stimulatory agent is IL-10 and the PBMC is an IgA- B cell or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-21 and the PBMC is an IgA- B cell or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS and the PBMC is an IgA- B cell or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is PMA and the PBMC is an IgA- B cell or a B translational cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-10, IL-21, LPS or PMA and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-10 and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-21 and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is PMA and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-7, IL-10, or IL-21 and the PBMC is a CD8+ activated T cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-7 and the PBMC is a CD8+ activated T cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-10 and the PBMC is a CD8+ activated T cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-21 and the PBMC is a CD8+ activated T cell.
  • the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the intracellular signaling molecule is STAT1.
  • the stimulatory agent is unstimulated
  • the PBMC is an IgD+ B memory cell, an IgD- CD27- B cell or a B naive cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-6 or IL-7 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-7 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS or PMA and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is PMA and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS or PMA and the PBMC is a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS and the PBMC is a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is PMA and the PBMC is a CD56dimCD16dim NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease.
  • the standard control is a healthy subject.
  • the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS or PMA and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, or a CD56bright NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is PMA and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, or a CD56bright NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the stimulatory agent is LPS and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, or a CD56bright NK cell.
  • the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • the sample from the subject is a blood sample or a plasma sample.
  • the sample from the subject is a blood sample.
  • the sample from the subject is a plasma sample.
  • the detecting includes contacting the ex vivo stimulated PBMC with one or more immune cell specific antibodies, thereby forming a labeled ex vivo stimulated PBMC.
  • the one or more immune cell-specific antibodies includes a detectable moiety.
  • the one or more immune cell-specific antibodies are an anti-CD3, an anti-CD4 antibody, an anti-CD7 antibody, an anti-CD8 antibody, an anti-CDllb antibody, an anti-CDllc antibody, an anti-CD14 antibody, an anti-CD16 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti-CD24 antibody, an anti-CD25 antibody, an anti- CD27 antibody, an anti-CD38 antibody, an anti-CD45RA antibody, an anti-CD56 antibody, an anti-CD 123 antibody, an anti-CD 127 antibody, an anti -IgA antibody, an anti-IgD antibody or an anti-HLA-Dr antibody.
  • the one or more immune cell-specific antibodies are selected from a group of antibodies consisting of an anti-CD3, an anti-CD4 antibody, an anti-CD7 antibody, an anti-CD8 antibody, an anti-CDllb antibody, an anti- CDllc antibody, an anti-CD14 antibody, an anti-CD16 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti-CD24 antibody, an anti-CD25 antibody, an anti-CD27 antibody, an anti-CD38 antibody, an anti-CD45RA antibody, an anti-CD56 antibody, an anti-CD123 antibody, an anti-CD 127 antibody, an anti-IgA antibody, an anti-IgD antibody and an anti- HLA-Dr antibody.
  • the detecting further includes contacting the labeled ex vivo stimulated PBMC with one or more anti-intracellular signaling molecule antibodies, thereby forming a intracellularly labeled ex vivo stimulated PBMC.
  • the one or more anti-intracellular signaling molecule antibodies are an anti-pERKl/2 antibody, an anti-IkBa antibody, an anti-NF-kB antibody, an anti-p38 antibody, an anti-pAKT antibody, an anti- pCREB antibody, an anti-pLCK antibody, an anti-pPLC-y2 antibody, an anti-pS6 antibody, an anti-pSTATl antibody, an anti-pSTAT3 antibody, an anti-pSTAT5 antibody, an anti- Lamp2 antibody, an anti-EEAl antibody, or an anti-Rab5 antibody.
  • the one or more anti-intracellular signaling molecule antibodies are selected from a group of antibodies consisting of an anti-pERKl/2 antibody, an anti-IkBa antibody, an anti-NF-kB antibody, an anti-p38 antibody, an anti-pAKT antibody, an anti-pCREB antibody, an anti- pLCK antibody, an anti-pPLC-y2 antibody, an anti-pS6 antibody, an anti-pSTATl antibody, an anti-pSTAT3 antibody, an anti-pSTAT5 antibody, an anti-Lamp2 antibody, an anti-EEAl antibody, and an anti-Rab5 antibody.
  • the detecting a level of phosphorylation includes detecting binding of the one or more anti-intracellular signaling molecule antibodies to the labeled ex vivo stimulated PBMC.
  • the ex vivo stimulated PBMC is a permeabilized PBMC.
  • the ex vivo stimulated PBMC is in a detection device.
  • the neurological disease treatment is a PLC-y2 pathway activator.
  • the neurological disease treatment is galantamine, rivastigmine, donepezil, memantine, levodopa, carbidopa, amantadine, a dopamine agonist, a monoamine oxidase B inhibitor, a Catechol-O-methyltransferase inhibitor, or an anticholinergic.
  • the neurological disease treatment is galantamine.
  • the neurological disease treatment is rivastigmine.
  • the neurological disease treatment is donepezil.
  • the neurological disease treatment is memantine. In embodiments, the neurological disease treatment is levodopa. In embodiments, the neurological disease treatment is carbidopa. In embodiments, the neurological disease treatment is amantadine. In embodiments, the neurological disease treatment is a dopamine agonist. In embodiments, the neurological disease treatment is a monoamine oxidase B inhibitor. In embodiments, the neurological disease treatment is a Catechol-O-methyltransferase inhibitor. In embodiments, the neurological disease treatment is an anticholinergic.
  • methods for measuring the propensity of a patient to be diagnosed with Alzheimer’s disease include: a) conducting a measurement on a patient sample to determine frequency and functional signals of pPLC-gamma;2 responses from unstimulated and after LPS -stimulated in most peripheral blood mononuclear cells but especially in T cell subsets, B cells, NK cells, and monocytes, pSTATl responses in IFN- alpha; stimulated peripheral plasmablast, CD4+CD8+ T cells, and CD4-CD8+ T cells, and IL-6 stimulated basophils and CD8+ central memory T cells, and pSTAT5 responses in IFN- alpha; stimulated CD4+ activated T cells, CD4-CD8+ T cells, CD4+ activated T cells, and monocytes b) feeding the measurement to the developed multivariate machine learning model and statistical analyses to achieve diagnosis likelihood.
  • the methods provided herein including embodiments thereof may be used to develop tools for diagnosis and early detection of Alzheimer
  • Applicants have built a novel data set that comprises a large sample set (for discovery of biomarkers) from well-matched participants with research quality annotation. Applicants have employed cutting-edge high-throughput mass cytometry technique to screen over four thousand cell signals. The selected biomarkers, with the developed models, can also be transferred to separate Parkinson’s patients from healthy cohort. They retain their capability to distinguish Alzheimer’s patients from Parkinson’s as well as dominantly inherited Alzheimer’s patients.
  • PBMCs Peripheral blood mononuclear cells
  • AD Alzheimer’s disease
  • PD Parkinson’s disease
  • PBMCs peripheral blood mononuclear cells
  • AD Alzheimer’s disease
  • PD Parkinson’s disease
  • PBMCs peripheral blood mononuclear cells
  • AD Alzheimer’s disease
  • PD Parkinson’s disease
  • HC-I refers to matched healthy controls for AD
  • HC- I sub refers to a subset of HC-I selected as matched healthy controls for PD (Table 1).
  • AD-V AD patients
  • HC-V matched healthy controls
  • Stimulant response was detected with additional probes directed at intracellular signaling molecules.
  • Signals obtained from AD, PD, and HC-I PBMCs in unstimulated state or in response to IFN-a, IL-6, and IL-7 were strongly correlated to each other as indicated by proximity and the communities formed by these features in the correlation network (FIG.
  • one of the inputs to the iEN model was a list of immune features that represent literature-based canonical signaling pathways to prioritize during model optimization. Additionally, we have tried using signals that were experimentally present in this study (-10% discrepancy compared to canonical features; FIG.s 5 A and 5B), and it showed no significant effect on the model performance. Other conventional algorithms were also attempted but obtained less accuracy. Next, we proceeded to focus on the iEN model’s components developed from AD and HC-I.
  • the coefficients assigned to the immune features were components of the iEN model that could be used as a proxy to examine biological plausibility. These coefficients were displayed on a correlation network with red color indicating immune features that increased with the likelihood of carrying the AD diagnosis, and vice versa for blue (FIG. 4C). Piecewise regression analysis of the model revealed that only about 14 of the features with the highest coefficient magnitudes were necessary, and 111 components were needed to get comparable performance to using the entire set of immune features (4200 components; FIG. 4D). These top 14 and 111 features were depicted on the correlation networks (FIG. 4E and FIG. 6A). We next explored the four communities that contained the top 14 features, since features within the same community were highly correlated, and exhibited high univariate P value of the immune feature to diagnosis: community 18, community 12, community 17, and community 20 (FIG. 4C).
  • Community 18 comprised features with pPLCy2 intracellular signal.
  • Visualizing the differences in pPLCy2 signal between the HC-I and the AD participants by PBMC subtypes and stimulant highlighted strong differences from diverse cell types (with the exception of most CD4 + T cells and DCs) in unstimulated and various stimulated conditions (FIG. 7A and FIG. 6C), where all of the responses tended to be lower in the AD group (FIG. 6B), particularly in NKT cells (FIG.s 7D and 7E).
  • FIG.s 7A and FIG. 6C shows that the most informative components of the iEN model, including pPLCy2 response in unstimulated NKT,
  • the prediction accuracy of the developed iEN models from AD/HC-I could be used as a proxy to determine the similarities between the diagnostic groups.
  • the correlation network of PD/HC-I sub diagnostic groups illustrated potential regions containing features associated with the developed iEN components from AD/HC-I (strong colors) that were also significantly different between PD and HC-I sub (sizable nodes) such as those in community 20 (FIG. 11C).
  • the accurate PD/HC-I sub prediction may indicate co morbidity, shared risk factors, or shared mechanisms between the two neurodegenerative diseases.
  • other communities whose features were important in the AD/HC-I iEN model particularly communities 12, 17 (pSTATs region), and 18 (pPLCy2 region), did not overlap with signal differences in PD/HC-I sub .
  • Reduced pPLCy2 activation was a strong feature of AD in all major subclasses of cells except dendritic cells and most CD4 + T cells, and under all stimulation conditions except IL-6 (FIG. 7A). Although pPLCy2 differential response was observed across most stimulants, it appeared strongest with LPS. pSTATl (community 20) and pSTAT5 (community 17) differential responses were more selective and largely restricted to IFN-a stimulation. pSTAT5 differential response was focused strongly in monocytes (FIG. 7C), while pSTATl differential response was weaker and more broadly observed in CD8+ T cells, B cells, and NK cells but not in monocytes (FIG. 7B).
  • TLR toll-like receptor 4 4
  • STAT1 tissue concentration is increased in diseased regions of AD brain (35) and can regulate expression of beta secretase 1, one of the endoproteases that sequentially catalyzes the hydrolysis of amyloid precursor protein to generate Ab peptides (36).
  • STAT5 activation regulates microglial activation and is required for monocyte-mediated synaptic degradation (37).
  • a recent pathway analysis that combined multiple GWAS data with clinical annotation nominated JAK-STAT signaling abnormalities as prominent contributors to AD etiology or pathogenesis (38).
  • both T cells and monocytes can traffic from peripheral blood into brain, at least raising the possibility that the differential responses observed in these subsets of PBMCs might access brain, rather than simply being peripheral biomarkers of brain immune responses.
  • the iEN algorithm was unable to develop a predictive model for PD despite a similarly sized sample set as AD. Although disappointing, these results enhance our confidence in the predictive model for AD by controlling for non-specific features of age- related neurodegenerative disease, like reduced physical activity.
  • Our disease cross prediction analysis showed that the AD/HC-I model much more weakly, but significantly, predicted PD/HC-I sub . There are several possible explanations for this outcome including partially shared genetic or environmental risk factors, or partially shared neurodegenerative mechanisms. The latter is supported by the now well-established observation that the neuropathological hallmark of PD (Lewy body disease) is present in about one-third to one- half of people diagnosed clinically with AD despite using the most rigorous research criteria.
  • this disease cross-prediction may indicate partially shared alterations in the peripheral immune response between people with AD or PD.
  • disease cross-prediction of PD was driven largely by community 20, and not by community 18 (pPLCy2 region; FIG. 11C); consonant with the finding that the genetic variant in PLCG2 that lowers risk for AD does not modulate risk of PD (40).
  • the aim of this study was to determine whether differences in peripheral immune responses between healthy participants and participants with neurodegenerative diseases are detectable from the CyTOF analysis of PBMCs.
  • Participants were research volunteers at Stanford University in the Alzheimer’s Disease Research Center (ADRC) or the Pacific Udall Center (PUC). The work was approved by the IRB. Clinical diagnosis was made by consensus criteria (46, 47, 48), see Supplemental Materials for details.
  • Blood was collected from volunteers after informed consent.
  • the discovery cohort consisted of four groups: AD, PD, and two different HC groups.
  • the first healthy controls were older and were matched for AD with a subgroup of these people (HC-I sub ; age between 63-80 for male and 67-73 for female) matched for PD.
  • the second healthy controls were younger.
  • a separate validation cohort also was assembled: AD-V and HC-V.
  • No validation cohort was assembled for PD due to no generalizable model identified from the discovery cohort.
  • a separate technical control of PBMCs was prepared from a single healthy individual (70-year-old man) not included in the discovery or validation cohorts, and frozen into multiple aliquots and included in every CyTOF run.
  • a panel of 37 metal -tagged monoclonal antibodies was used to probe PBMCs (Table 4). All pre-conjugated antibodies were purchased from Fluidigm. All other antibodies were purchased in carrier protein free PBS and conjugated in house with the respective metal isotope using the MaxPar antibody conjugation kit (Fluidigm). Metal labelled antibodies were diluted to 0.5 mg/ml in Candor PBS Antibody Stabilization solution (Candor Bioscience GmbH) for storage at 4 °C.
  • PBMCs were isolated from freshly drawn whole blood using density gradient centrifugation (Ficoll-Paque PLUS; GE Healthcare) in Sepmate tubes (49). The isolated and washed whole blood PBMCs were resuspended in 10% DMSO, 90% FBS and cryopreserved in liquid nitrogen. Frozen aliquots of PBMCs (batch of ⁇ 10 samples and an aliquot of technical control) were washed twice in RPMI at 37 °C. The samples were brought up in 1 ml RPMI and viability checked. One hundred pi of each sample were aliquoted and rested for 1 hr at 37 °C.
  • PBMCs were then incubated in RPMI (unstimulated) or one of the seven specific stimulants, either a chemokine/cytokine (IFN-a, IL-6, IL-7, IL-10, IL-21), LPS, or PMA plus Ionomycin, for 15 min at 37 °C as shown in Table 5 and described previously (50). Following incubation under these eight conditions, PBMCs were fixed for 15 min with 4% PFA at room temperature, washed, permeabilized and barcoded exactly according to published methods to facilitate processing and minimize batch effects (51).
  • a chemokine/cytokine IFN-a, IL-6, IL-7, IL-10, IL-21
  • LPS chemokine/cytokine
  • All 8 stimulation conditions from a barcoded sample were pooled and incubated with titrated metal-labelled antibodies directed at cell surface markers designed to identify 35 immune cell subsets (52, 53).
  • Cell types were identified by surface antibody signal for the following lineage markers: CD3, CD4, CD7, CD8, CDllb, CDllc, CD14, CD16, CD19, CD20, CD24, CD25, CD27, CD38, CD45RA, CD56, CD123, CD127, IgA, IgD and HLA- Dr. Pooled sample was then permeabilized with methanol and stored at -80 °C.
  • Intracellular signaling markers were pERKl/2, IkBa, NF-kB, p38, pAKT, pCREB, pLCK, pPLCy2, pS6, pSTATl, pSTAT3, pSTAT5, and endosomal proteins, Lamp2, EEA1, and Rab5 (55-57).
  • the iEN model was employed as a multivariate model to examine generalizability and predictive power.
  • the iEN added on to the commonly used EN algorithm the capability to incorporate knowledge of intracellular signal transduction on the generation of the mass cytometry data and was shown to yield superior performance (25).
  • the iEN algorithm optimized the coefficient (b) for each associated feature by minimizing the cost function:
  • X is a matrix of size m x n, where m is the number of samples and n is the number of all immune features (intracellular signals from cells under different stimulating conditions), Y is a vector of ground truths (diagnostic groups) with length n, b is a vector of the model’s coefficients with length n, l and a together control the magnitude of the model regularization.
  • F is a diagonal matrix of size n x n containing a prioritization value of 1 for elements associated with canonical immune feature or e -ip for other lower priority (non- canonical) features. The list of canonical signals that were prioritized was tabulated in Table 6
  • a consensus panel consisting of one board-certified movement disorders neurologist or behavioral neurologist, one board-certified neuropsychologist, and other study personnel adjudicated the diagnosis for each participant.
  • PD diagnosis was based on UK PD Society Brain Bank clinical diagnostic criteria (46), as previously reported (47).
  • the layout of the correlation network was obtained from applying dimension reduction algorithm, t-SNE (61), on the Spearman correlation matrix of all available immune features.
  • the edge of the graph represents those with Spearman’s P value ⁇ 0.05 after Bonferroni adjustment.
  • the communities were identified by using uniform manifold approximation and projection (UMAP) to reduce the dimension to 10% of the original immune features (62), followed by unsupervised clustering using A-means algorithm.
  • the optimized number of clusters (24 clusters) was determined based on optimized C-index and Baker-Hubert gamma index.
  • Table 2 Demographics (age, sex, and counts of APOE e4 gene) of participants in discovery cohort by diagnostic groups.
  • Table 3 Demographics (age, sex, and counts of APOE e4 gene) of participants in validation cohort by diagnostic groups.
  • Table 4 Target, clone, conjugation information, and product identifier.
  • Table 7 Exemplary embodiments of intracellular signaling molecules, stimulatory agents and PBMCs contemplated for the methods provided herein including embodiments thereof.
  • Phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph, Alzheimer's Research & Therapy 11,
  • Embodiment 1 A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease, said method comprising:
  • PBMC peripheral blood mononuclear cell
  • said PBMC is a CD4 + T cell, a CD8 + T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
  • IFN-a interferon a
  • IL-6 interleukin-6
  • IL-7 interleukin-7
  • IL-10 interleukin- 10
  • IL-21 interleukin-21
  • LPS lipopolysaccharides
  • PMA phorbol myristate acetate
  • Embodiment 2 A method of treating a neurological disease in a subject in need thereof, said method comprising:
  • PBMC peripheral blood mononuclear cell
  • said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
  • IFN-a interferon a
  • IL-6 interleukin-6
  • IL-7 interleukin-7
  • IL-10 interleukin- 10
  • IL-21 interleukin-21
  • LPS lipopolysaccharides
  • PMA phorbol myristate acetate
  • Embodiment 3 A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject undergoing treatment for a neurological disease, said method comprising:
  • PBMC peripheral blood mononuclear cell
  • said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
  • IFN-a interferon a
  • IL-6 interleukin-6
  • IL-7 interleukin-7
  • IL-10 interleukin- 10
  • IL-21 interleukin-21
  • LPS lipopolysaccharides
  • PMA phorbol myristate acetate
  • Embodiment 4 A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease, said method comprising:
  • PBMC peripheral blood mononuclear cell
  • said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
  • Embodiment 5 The method of embodiment 4, wherein an increased level of phosphorylation relative to said standard control indicates that said subject has or is at risk of developing a neurological disease.
  • Embodiment 6 The method of embodiment 4, wherein a decreased level of phosphorylation relative to said standard control indicates that said subject has or is at risk of developing a neurological disease.
  • Embodiment 7 The method of any one of embodiments 4-6, comprising based at least in part on said level of phosphorylation in step (v) administering a neurological disease treatment to said subject.
  • Embodiment 8 The method of any one of embodiments 1-7, wherein said intracellular signaling molecule is PLC-y2.
  • Embodiment 9 The method of embodiment 8, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD4 + activated T cell, a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4 + CD8 + T cell, a natural killer T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD 16 h '" h NK cell, a CD56 bnght NK cell, a CD56 dim CD16 dim NK cell or a CD16 hl " h monocyte.
  • Embodiment 10 The method of embodiment 8 or 9, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a CD4 + activated T cell, a CD8 + activated T cell, a CD4 + CD8 + T cell, or a natural killer T cell.
  • Embodiment 11 The method of embodiment 8 or 9, wherein said stimulatory agent is IL-21 or LPS and said PBMC is a CDie 1 ⁇ 11 monocyte.
  • Embodiment 12 The method of embodiment 8 or 9, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD 16 high NK cell, a CD56 bnght NK cell, or a CD56 dim CD16 dim NK cell.
  • Embodiment 13 The method of embodiment 8, wherein said stimulatory agent is IL-6 and said PBMC is a CD8 + activated T cell, a CD4 + CD8 + T cell or a natural killer T cell.
  • Embodiment 14 The method of embodiment 8 or 13, wherein said stimulatory agent is IL-6 and said PBMC is a CD8 + activated T cell.
  • Embodiment 15 The method of embodiment 8 or 13, wherein said stimulatory agent is IL-6 and said PBMC is a CD4 + CD8 + T cell or a natural killer T cell.
  • Embodiment 16 The method of embodiment 8, wherein said stimulatory agent is IFN-a or IL-7 and said PBMC is a CD4 + activated T cell, a CD8 + activated T cell, a CD4 + CD8 + T cell, a natural killer T cell, a CD56 bnght NK cell, or a CDie 1 ⁇ 11 monocyte.
  • Embodiment 17 The method of embodiment 8 or 16, wherein said stimulatory agent is IFN-a or IL-7 and said PBMC is a natural killer T cell.
  • Embodiment 18 The method of embodiment 8 or 16, wherein said stimulatory agent is IFN-a or IL-7 and said PBMC is a CD8 + activated T cell or a CDlh ⁇ 811 monocyte.
  • Embodiment 19 The method of embodiment 8 or 16, wherein said stimulatory agent is IFN-a or IL-7 and said PBMC is a CD4 + activated T cell, a CD4 + CD8 + T cell, or a CD56 bright NK cell.
  • Embodiment 20 The method of embodiment 8, wherein said stimulatory agent is PMA and said PBMC is a CD4 + activated T cell, a CD8 + activated T cell, a natural killer T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
  • said stimulatory agent is PMA and said PBMC is a CD4 + activated T cell, a CD8 + activated T cell, a natural killer T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
  • Embodiment 21 The method of embodiment 8 or 20, wherein said stimulatory agent is PMA and said PBMC is a CD4 + activated T cell, a natural killer T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
  • Embodiment 22 The method of embodiment 8 or 20, wherein said stimulatory agent is PMA and said PBMC is a CD8 + activated T cell.
  • Embodiment 23 The method of any one of embodiments 1 -7, wherein said intracellular signaling molecule is STAT1.
  • Embodiment 24 The method of embodiment 23, wherein said stimulatory agent is IFN-a and said PBMC is a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4 CD8 T cell, a CD4 + CD8 + T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD56 bnght NK cell, a myeloid dendritic cell (mDC) or a plasmacytoid dendritic cell (pDC).
  • mDC myeloid dendritic cell
  • pDC plasmacytoid dendritic cell
  • Embodiment 25 The method of embodiment 23, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • Embodiment 26 The method of embodiment 23, wherein said stimulatory agent is LPS and said PBMC is a CD8 + central memory T cell.
  • Embodiment 27 The method of any one of embodiments 1-7, wherein said intracellular signaling molecule is STAT5.
  • Embodiment 28 The method of embodiment 27, wherein said stimulatory agent is IFN-a and said PBMC is a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4 CD8 T cell, a CD16 high monocyte or a CD16 low monocyte.
  • said stimulatory agent is IFN-a and said PBMC is a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4 CD8 T cell, a CD16 high monocyte or a CD16 low monocyte.
  • Embodiment 29 The method of embodiment 27 or 28, wherein said stimulatory agent is IFN-a and said PBMC is a CD 16 hlgh monocyte or a CD16 low monocyte.
  • Embodiment 30 The method of embodiment 27 or 28, wherein said stimulatory agent is IFN-a and said PBMC is a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, or a CD4 CD8 T cell.
  • Embodiment 31 The method of embodiment 27, wherein said stimulatory agent is IL-7 and said PBMC is a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, or a CD8 + effector memory T cell.
  • Embodiment 32 The method of embodiment 27, wherein said stimulatory agent is LPS and said PBMC is a CD4 + CD8 + T cell.
  • Embodiment 33 The method of embodiment 8, wherein said subject is a male subject.
  • Embodiment 34 The method of embodiment 33, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD4 + activated T cell, a regulatory T cell, a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4 + CD8 + T cell, a natural killer T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16 1 ’ 1 " 1 ’ NK cell, a CD56 bright NK cell, a CD56 dim CD16 dim NK cell, a CD16 1 ' 1 " 1 ' monocyte or a CD16 low monocyte.
  • said stimulatory agent is unstim
  • Embodiment 35 The method of embodiment 33 or 34, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a CD4 + activated T cell, a CD8 + activated T cell, a CD4 + CD8 + T cell or a natural killer T cell.
  • Embodiment 36 The method of embodiment 33 or 34, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a regulatory T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CDie ⁇ NK cell, a CD56 bright NK cell, a CD56 dim CD16 dim NK cell, a CD16 hlgh monocyte or a CD16 low monocyte.
  • said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a regulatory T cell, a CD8 + central memory T cell, a CD8 + effector
  • Embodiment 37 The method of embodiment 33, wherein said stimulatory agent is IFN-a, IL-6 or IL-7 and said PBMC is a CD4 + activated T cell, a CD4 + CD8 + T cell, a natural killer T cell, a CD8 + activated T cell, a CD16 high monocyte or a CD16 low monocyte.
  • said stimulatory agent is IFN-a, IL-6 or IL-7 and said PBMC is a CD4 + activated T cell, a CD4 + CD8 + T cell, a natural killer T cell, a CD8 + activated T cell, a CD16 high monocyte or a CD16 low monocyte.
  • Embodiment 38 The method of embodiment 33, wherein said stimulatory agent is PMA and said PBMC is a CD4 + activated T cell, a CD4 + naive T cell, a CD8 + activated T cell, a natural killer T cell, a CD4 + CD8 + T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
  • Embodiment 39 The method of embodiment 33 or 38, wherein said stimulatory agent is PMA and said PBMC is a CD4 + activated T cell, a CD4 + naive T cell, or a natural killer T cell.
  • Embodiment 40 The method of embodiment 33 or 38, wherein said stimulatory agent is PMA and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
  • Embodiment 41 The method of embodiment 33 or 38, wherein said stimulatory agent is PMA and said PBMC is a CD8 + activated T cell or a CD4 + CD8 + T cell.
  • Embodiment 42 The method of any one of embodiments 1 -7, wherein said intracellular signaling molecule is AKT.
  • Embodiment 43 The method of embodiment 42, wherein said subject is a male subject.
  • Embodiment 44 The method of embodiment 43, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4 + activated T cell, a CD4 + central memory T cell, a CD4 + effector T cell, a CD4 + effector memory T cell, a
  • CD4 + naive T cell or a regulatory T cell.
  • Embodiment 45 The method of embodiment 43 or 44, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4 + activated T cell, a CD4 + central memory T cell, a CD4 + effector T cell, a CD4 + naive T cell, or a regulatory T cell.
  • said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4 + activated T cell, a CD4 + central memory T cell, a CD4 + effector T cell, a CD4 + naive T cell, or a regulatory T cell.
  • Embodiment 46 The method of embodiment 43 or 44, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4 + effector memory T cell.
  • Embodiment 47 The method of embodiment 43, wherein said stimulatory agent is unstimulated and said PBMC is a CD4 + activated T cell, a CD4 + central memory T cell, a CD4 + effector T cell, a CD4 + naive T cell, or a regulatory T cell.
  • Embodiment 48 The method of embodiment 43, wherein said stimulatory agent is IL-7 and said PBMC is a CD4 + activated T cell.
  • Embodiment 49 The method of embodiment 43, wherein said stimulatory agent is PMA and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a mDC, a pDC, a CD 16 1 "" 1 ' monocyte or a CD16 low monocyte.
  • Embodiment 50 The method of embodiment 23, wherein said subject is a female subject.
  • Embodiment 51 The method of embodiment 50, wherein said stimulatory agent is unstimulated, IL-7, IL-10 or LPS and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, or a B translational cell.
  • Embodiment 52 The method of embodiment 50 or 51, wherein said stimulatory agent is unstimulated and said PBMC is a B naive cell.
  • Embodiment 53 The method of embodiment 50 or 51, wherein said stimulatory agent is unstimulated, IL-7, IL-10 or LPS and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, or a B translational cell.
  • Embodiment 54 The method of embodiment 50, wherein said stimulatory agent is IFN-a and said PBMC is a CD4 + activated T cell, a CD4 + effector T cell, a CD4 + naive T cell, a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4 + CD8 + T cell, a CD4 CD8 T cell, a natural killer T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CDie ⁇ NK cell, a CD56 bnght NK cell, a mDC, a pDC, a CD 16 1 "" 1 ' monocyte or a CD16 low monocyte.
  • said stimulatory agent
  • Embodiment 55 The method of embodiment 50 or 51, wherein said stimulatory agent is IFN-a and said PBMC is a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, or a pDC.
  • Embodiment 56 The method of embodiment 50 or 51, wherein said stimulatory agent is IFN-a and said PBMC is a CD4 + activated T cell, a CD4 + effector T cell, a CD8 + activated T cell, a CD8 + central memory T cell, a CD4 + CD8 + T cell, a CD4 CD8 T cell, a natural killer T cell, a plasmablast cell, or a CD56 bnght NK cell.
  • said stimulatory agent is IFN-a and said PBMC is a CD4 + activated T cell, a CD4 + effector T cell, a CD8 + activated T cell, a CD8 + central memory T cell, a CD4 + CD8 + T cell, a CD4 CD8 T cell, a natural killer T cell, a plasmablast cell, or a CD56 bnght NK cell.
  • Embodiment 57 The method of embodiment 50 or 51, wherein said stimulatory agent is IFN-a and said PBMC is a CD4 + naive T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, a B translational cell, a CD 16 h '" h NK cell, a mDC, a CD 16 h '" h monocyte or a CD16 low monocyte.
  • said stimulatory agent is IFN-a and said PBMC is a CD4 + naive T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, a B translational cell, a CD 16 h '" h NK cell, a mDC, a CD 16 h '" h monocyte or a CD16 low monocyte.
  • Embodiment 58 The method of embodiment 50, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • Embodiment 59 The method of embodiment 50 or 58, wherein said stimulatory agent is IL-6 and said PBMC is an IgD CD27 B cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • Embodiment 60 The method of embodiment 50 or 58, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD + B memory cell, or a B naive cell.
  • Embodiment 61 The method of embodiment 50, wherein said stimulatory agent is IL-21 and said PBMC is an IgD CD27 B cell.
  • Embodiment 62 The method of embodiment 27, wherein said subject is a female subject.
  • Embodiment 63 The method of embodiment 62, wherein said stimulatory agent is
  • IFN-a and said PBMC is a CD4 + activated T cell, a CD4 + effector T cell, a CD4 + effector memory T cell, a CD4 + naive T cell, a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4 CD8 T cell, a CD4 + CD8 + T cell, a natural killer T cell, a CD 16 h '" h monocyte or a CD16 low monocyte.
  • Embodiment 64 The method of embodiment 62 or 63, wherein said stimulatory agent is IFN-a and said PBMC is a CD8 + effector memory T cell.
  • Embodiment 65 The method of embodiment 62 or 63, wherein said stimulatory agent is IFN-a and said PBMC is a CD4 + effector T cell, a CD8 + central memory T cell, a CD8 + naive T cell, or a natural killer T cell.
  • Embodiment 66 The method of embodiment 62 or 63, wherein said stimulatory agent is IFN-a and said PBMC is a CD4 + activated T cell, a CD4 + effector memory T cell, a CD4 + naive T cell, a CD8 + activated T cell, a CD8 + effector T cell, a CD4 CD8 T cell, a CD4 + CD8 + T cell, a CD 16 h '" h monocyte or a CD16 low monocyte.
  • Embodiment 67 Embodiment 67.
  • said stimulatory agent is IL-7 and said PBMC is a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD4 CD8 T cell, a CD4 + CD8 + T cell or a natural killer T cell.
  • Embodiment 68 The method of embodiment 62 or 66, wherein said stimulatory agent is IL-7 and said PBMC is a CD8 + central memory T cell, or a CD8 + effector memory T cell.
  • Embodiment 69 The method of embodiment 62 or 66, wherein said stimulatory agent is IL-7 and said PBMC is a natural killer T cell.
  • Embodiment 70 The method of embodiment 62 or 66, wherein said stimulatory agent is IL-7 and said PBMC is a CD8 + effector T cell, a CD4 CD8 T cell, or a CD4 + CD8 + T cell.
  • Embodiment 71 The method of any one of embodiments 1-70, wherein said neurological disease is Alzheimer’s disease or Parkinson’s disease.
  • Embodiment 72 The method of embodiment 71, wherein said intracellular signaling molecule is PLC-y2.
  • Embodiment 73 The method of embodiment 71 or 72, wherein said stimulatory agent is unstimulated and said PBMC is a CD56 bnght NK cell.
  • Embodiment 74 The method of embodiment 71 or 72, wherein said stimulatory agent is IL-10, IL-21 or LPS and said PBMC is a basophil.
  • Embodiment 75 The method of embodiment 71, wherein said intracellular signaling molecule is STAT1.
  • Embodiment 76 The method of embodiment 71 or 75, wherein said stimulatory agent is IFN-a and said PBMC is a plasmablast or CD56 bnght NK cell.
  • Embodiment 77 The method of embodiment 71, wherein said intracellular signaling molecule is STAT5.
  • Embodiment 78 The method of embodiment 71 or 77, wherein said stimulatory agent is IFN-a and said PBMC is a CD 16 h '" h monocyte or a CD16 low monocyte.
  • Embodiment 79 The method of any one of embodiments 1-71, wherein said neurological disease is Alzheimer’s disease.
  • Embodiment 80 The method of embodiment 79, wherein said intracellular signaling molecule is PLC-y2.
  • Embodiment 81 The method of embodiment 80, wherein said stimulatory agent is IL-10, IL-21, LPS or PMA and said PBMC is a CD4 + CD8 + T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • said stimulatory agent is IL-10, IL-21, LPS or PMA
  • said PBMC is a CD4 + CD8 + T cell, an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • Embodiment 82 The method of embodiment 80 or 81, wherein said stimulatory agent is IL-10, IL-21, LPS or PMA and said PBMC is an IgA B cell or a B translational cell.
  • Embodiment 83 The method of embodiment 80 or 81, wherein said stimulatory agent is IL-10, IL-21, LPS or PMA and said PBMC is a CD4 + CD8 + T cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, or a B switched memory cell.
  • Embodiment 84 The method of embodiment 80, wherein said stimulatory agent is IL-7, IL-10, or IL-21 and said PBMC is a CD8 + activated T cell.
  • Embodiment 85 The method of embodiment 79, wherein said intracellular signaling molecule is STAT1.
  • Embodiment 86 The method of embodiment 85, wherein said stimulatory agent is unstimulated, and said PBMC is an IgD + B memory cell, an IgD CD27 B cell or a B naive cell.
  • Embodiment 87 The method of embodiment 85, wherein said stimulatory agent is IL-6 or IL-7 and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16 1 "" 1 ' NK cell, a CDSe ⁇ NK cell, or a CD56 dim CD16 dim NK cell.
  • said stimulatory agent is IL-6 or IL-7 and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16 1 "" 1 ' NK cell, a CDSe ⁇ NK cell, or a CD56 dim CD16 dim NK cell.
  • Embodiment 88 The method of embodiment 85, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD + B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
  • Embodiment 89 The method of embodiment 85, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD4 + effector T cell, a CD4 + effector memory T cell, a CD4 + naive T cell, a regulatory T cell, a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4 CD8- T cell, an IgD CD27 B cell, a B naive cell, a O ⁇ 11 NK cell, a 0056 ⁇ NK cell, or a CD56 dim CD16 dim NK cell.
  • said stimulatory agent is LPS or PMA and said PBMC is a CD4 + effector T cell, a CD4 + effector memory T cell, a CD4 + naive T cell, a regulatory T cell, a CD8 + activated T cell, a CD8 +
  • Embodiment 90 The method of embodiment 85 or 89, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD56 dim CD16 dim NK cell.
  • Embodiment 91 The method of embodiment 85 or 89, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD4 + effector T cell, a CD4 + effector memory T cell, a CD4 + naive T cell, a regulatory T cell, a CD8 + activated T cell, a CD8 + central memory T cell, a CD8 + effector T cell, a CD8 + effector memory T cell, a CD8 + naive T cell, a CD4- CD8- T cell, an IgD CD27 B cell, a B naive cell, a CD 16 ⁇ NK cell, or a CD56 bri ⁇ ht NK cell.
  • said stimulatory agent is LPS or PMA and said PBMC is a CD4 + effector T cell, a CD4 + effector memory T cell, a CD4 + naive T cell, a regulatory T cell, a CD8 + activated T cell, a CD8 + central memory T cell,
  • Embodiment 92 The method of any one of embodiments 1-91, wherein said sample from said subject is a blood sample or a plasma sample.
  • Embodiment 93 The method of any one of embodiments 1-92, wherein said detecting comprises contacting said ex vivo stimulated PBMC with one or more immune cell specific antibodies, thereby forming a labeled ex vivo stimulated PBMC.
  • Embodiment 94 The method of embodiment 93, wherein said one or more immune cell-specific antibodies comprises a detectable moiety.
  • Embodiment 95 The method of embodiment 93 or 94, wherein said one or more immune cell-specific antibodies are an anti-CD3, an anti-CD4 antibody, an anti-CD7 antibody, an anti-CD8 antibody, an anti-CDllb antibody, an anti-CDllc antibody, an anti- CD 14 antibody, an anti-CD 16 antibody, an anti-CD 19 antibody, an anti-CD20 antibody, an anti-CD24 antibody, an anti-CD25 antibody, an anti-CD27 antibody, an anti-CD38 antibody, an anti-CD45RA antibody, an anti-CD56 antibody, an anti-CD123 antibody, an anti-CD127 antibody, an anti-IgA antibody, an anti-IgD antibody or an anti-HLA-Dr antibody.
  • said one or more immune cell-specific antibodies are an anti-CD3, an anti-CD4 antibody, an anti-CD7 antibody, an anti-CD8 antibody, an anti-CDllb antibody, an anti-CDllc antibody, an anti- CD 14 antibody, an anti-CD 16 antibody, an anti-CD 19 antibody,
  • Embodiment 96 The method of any one of embodiments 93-95, wherein said detecting further comprises contacting said labeled ex vivo stimulated PBMC with one or more anti-intracellular signaling molecule antibodies, thereby forming a intracellularly labeled ex vivo stimulated PBMC.
  • Embodiment 97 The method of embodiment 96, wherein said one or more anti- intracellular signaling molecule antibodies are an anti-pERKl/2 antibody, an anti-IkBa antibody, an anti-NF-kB antibody, an anti-p38 antibody, an anti-pAKT antibody, an anti- pCREB antibody, an anti-pLCK antibody, an anti-pPLC-y2 antibody, an anti-pS6 antibody, an anti-pSTATl antibody, an anti-pSTAT3 antibody, an anti-pSTAT5 antibody, an anti- Lamp2 antibody, an anti-EEAl antibody, or an anti-Rab5 antibody.
  • Embodiment 98 The method of any one of embodiments 96-97, wherein said detecting a level of phosphorylation comprises detecting binding of said one or more anti- intracellular signaling molecule antibodies to said labeled ex vivo stimulated PBMC.
  • Embodiment 99 The method of any one of embodiments 1-98, wherein said ex vivo stimulated PBMC is a permeabilized PBMC.
  • Embodiment 100 The method of any one of embodiments 1-99, wherein said ex vivo stimulated PBMC is in a detection device.
  • Embodiment 101 The method of any one of embodiments 2 or 7-100, wherein said neurological disease treatment is a PLC-y2 pathway activator.
  • Embodiment 102 The method of any one of embodiments 2 or 7-100, wherein said neurological disease treatment is galantamine, rivastigmine, donepezil, memantine, levodopa, carbidopa, amantadine, a dopamine agonist, a monoamine oxidase B inhibitor, a Catechol-O- methyltransferase inhibitor, or an anticholinergic.
  • said neurological disease treatment is galantamine, rivastigmine, donepezil, memantine, levodopa, carbidopa, amantadine, a dopamine agonist, a monoamine oxidase B inhibitor, a Catechol-O- methyltransferase inhibitor, or an anticholinergic.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Epidemiology (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Emergency Medicine (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Provided herein are, inter alia, methods of detecting phosphorylation levels of, for example, PLC-γ2, AKT, STAT1 or STAT5, in a subject having or being at risk of developing a neurological disease (e.g., Alzheimers disease or Parkinsons disease). The methods include, inter alia, isolation of peripheral blood mononuclear cells (PBMCs) and stimulating the PBMCs with a stimulatory agent (e.g., interferon a (IFN-α), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin-10 (IL-10), interleukin-21 (IL-21), lipopolysaccharides (LPS) or phorbol myristate acetate (PMA) ex vivo.

Description

ALZHEIMER’S DIAGNOSTIC METHOD
CROSS-REFERENCES TO RELATED APPLICATIONS [0001] This application claims priority to U.S. Provisional Application No. 63/001,195, filed March 27, 2020, which is hereby incorporated by reference in its entirety and for all purposes.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT [0002] This invention was made with Government support under contracts AG047366 and NS062684 awarded by the National Institutes of Health. The Government has certain rights in the invention.
BACKGROUND OF THE INVENTION
[0003] Existing biomarkers for Alzheimers disease include cerebrospinal fluid and PET markers of amyloid-beta; and tau proteins, which are accurate in detecting the presence of AD-associated pathophysiological and neuropathological changes. However, the high cost, insufficient accessibility and/or invasiveness of these assays limit their use as viable first-line tools for detecting pathophysiologic patterns, aka biomarkers. Blood-based and plasma- based biomarkers are being developed with some showing promises, but results in the field have been mixed because of several reasons such as selected candidates rather than data- derived candidates, reliance on just one to only a few biomarkers, inconsistencies in clinical cohorts, and discrepancies in diagnostic evaluations. The methods provided herein address these and other needs in the art.
BRIEF DESCRIPTION OF THE DRAWINGS [0004] FIG.s 1A-1C show the experimental and analytical workflow from obtaining PBMCs to identifying potential immune cell markers. FIG. 1A shows in that the discovery cohorts, whole blood was collected from 28 individuals with AD and 17 individuals with PD; AD was compared with the samples from 53 older healthy controls (HC-I) while PD samples were compared with a subset of those with age- and sex-matched healthy controls (HC-Isub).
A different set of 10 younger healthy controls (HC-II) was included for examining age effects. Additionally, an independent cohort of 9 individuals with AD (AD-V) and 15 healthy controls (HC-V) was used for validation of the developed machine learning models without retraining. FIG. IB illustrates PBMCs were either unstimulated or stimulated with IFN-a, IL- 6, IL-7, IL-10, IL-21, LPS, or PMA/ionomycin. PBMCs were then bound with 21 metal- conjugated antibodies to surface markers and 15 metal-conjugated antibodies to intracellular signaling molecules before analysis by CyTOF. FIG. 1C shows cell abundance was evaluated on PBMCs from unstimulated condition. The stimulations and antibody probes generated a total of 4200 intracellular signaling responses (35 PBMC subtypes under 8 stimulating conditions and assayed for 15 intracellular responses), which were used to identify the potential immune features with the aid of cell-signaling knowledge, machine learning methods, and statistical analysis.
[0005] FIG.s 2A-2F illustrate AD and PD patients exhibited different cell frequency in several cell subsets. FIG. 2A is a CITRUS-derived dendrogram of unstimulated PBMC data from AD and PD. PBMC subsets are represented as clusters and color coded for level of marker expression (see Methods). Red highlighted nodes are cell subtypes of CD4+ Tcemrai Mem, blue highlighted nodes are CD4+ TActivated, orange highlighted clusters are CD8+ TNaive and the green clusters are CD38+CD16low monocytes that were significantly different (q value < 0.05) among the two diagnostic groups. FIG. 2B is a bar graph showing the significance levels of the highlighted nodes. FIG.s 2C-2F are box plots presenting example values from the last node in each of the four highlighted lineages for CD4+ Tcemrai memory cells (FIG. 2C), CD4+ TActivated cells (FIG. 2D), CD81 TNaive cells (FIG. 2E), and CD38+CD16low monocytes (FIG 2F). P values for the stratified subsets were determined using Wilcoxon rank-sum test.
[0006] FIG.s 3A and 3B show responses from the same intracellular signaling proteins are highly correlated to each other. FIG. 3A is a correlation network (Spearman’s coefficient) of immune features obtained from CyTOF data of HC-I, AD, and PD colored by the type of stimulant. The edges of the network represent features with Spearman’s coefficient higher than 0.8. FIG. 3B illustrates an unsupervised algorithm clustering the network into 24 communities, where their annotations are based on commonly shared feature attributes (PBMC subtype, stimulation, or signaling property) within the community.
[0007] FIG.s 4A-4E illustrate that the iEN model can satisfactorily classify AD/HC-I in both discovery and validation cohorts with most important model components associated with signals from pPLCy2 and pSTATs. FIG. 4A is box plots showing the predicted values from iEN model with Wilcoxon rank-sum test P value for discovery and validation cohort. FIG. 4B is receiver operating characteristics (ROC) curves from the iEN model predictions of discovery and validation cohorts. FIG. 4C is a correlation network colored by iEN model components with red and blue colors highlighting the components that are indicative of AD and HC-I, respectively. The size of the nodes represents the Spearman’s coefficient of the immune feature to the respective ground truths. FIG. 4D illustrates a model reduction analysis looking at the effect of the number of included features on iEN performance. FIG. 4E is a correlation network colored and annotated only for the top 14 features that were associated with components selected from model reduction.
[0008] FIG.s 5A and 5B illustrate portions of the canonical response versus responses observed experimentally in this study. FIG. 5A is a correlation network illustrated by responses that were expected in literature and were found in this study (grey), those that were not expected from literature and were also not found (white), and those that were expected in literature but not found experimentally and vice versa. FIG. 5B shows the portion of the responses that were observed (and not observed) according to literature and experiments in this study.
[0009] FIG.s 6A-6E are visualizations of features with strong signals and other features associated with 111 top iEN selected components. FIG. 6A is a network correlation with all of the features associated with top 111 iEN components colored. FIG. 6A are heatmaps of the selected communities with magnitude colored by Spearman’s correlation (r). FIG.s 6C- 6E are box plots of the selected features in the communities for pPLCy2 (FIG. 6C), pSTATl (FIG. 6D) and pSTAT5 (FIG. 6E) responses. The shaded gray area indicated that the features are also a part of reduced iEN’s top 14 components, and the box indicates features that are among the top 111 components.
[0010] FIG.s 7A-7H are heatmaps and box plots of the intracellular response in the PBMC of the selected communities highlight immune features for AD/HC-I classification. FIG.s 7A-7C are heatmaps of the pPLCy2 (FIG. 7A), pSTATl (FIG. 7B), and pSTAT5 (FIG. 7C) responses by PBMC subtypes and stimulations. The color of the heatmap scaled with the Wilcoxon rank-sum testP value of the difference in response of the immune feature between HC-I and AD patients. The network communities annotated with these responses (community 12, 17, 18, and 20) were depicted on the left hand side of the heatmap. The size of the nodes in the community corresponds to the Spearman’s coefficient of the immune feature. The features within the communities that were selected by reduced iEN model (14 components) retained their red/blue colors corresponding to the direction of the component. FIG.s 7D-7H are box plots showing the significant difference of the selected immune features from the heatmap, including pPLCy2 response in unstimulated NKT cells (FIG. 7D), pPLCy2 response in LPS stimulated NKT cells (FIG. 7E), pPLCy2 response in unstimulated CD56bnght NK cells (FIG. 7F), and pSTATl response in IFN-a stimulated plasmablast cells (FIG. 7G).
These are mostly features associated with the most informing components of the iEN model.
[0011] FIG.s 8A-8G illustrate that responses of pPLCy2. pSTATl, and pSTAT5 show expected expressions according literature. FIG. 8A are heatmaps of the mean values of normalized responses in each cell subset and stimulation for all AD patients in the discovery cohorts before batch correction. FIG. 8B is similar to FIG. 8A but for HC-I participants.
FIG. 8C is a similar heatmap with only AD patients of the biggest batch in the discovery cohort. FIG. 8D is similar to FIG. 8C but for HC-I participants. FIG.s 8E-8G are examples of raw gated signals and cell frequency with the corresponding histograms from a pair of participants. Flow cytometry data for pPLCy2 response of unstimulated NKTs (FIG. 8E), pSTATl response of IFN-a stimulated DCs (FIG. 8F), and pSTATl response of IFN-a stimulated monocytes (FIG. 8F) are shown.
[0012] FIG.s 9A-9F are correlation networks for AD/HC-I separated by sex and APOE :-:4 allele. FIGs. 9A and 9B is the correlation network with node size corresponding to the Wilcoxon rank-sum testP value of each feature for AD/HC-I diagnosis in male (FIG. 9A) and female participants (FIG. 9B) (discovery cohort). FIGs. 9C and 9D show a similar correlation network for all sex of healthy cohorts (HC-I) but for sex-specific AD. FIG. 9E is a similar correlation network for all sex of healthy cohorts (HC-I) with no APOE :-:4 allele and all sex of AD with at least one APOE e4 allele. FIG. 9F is similar to FIG. 9E but for HC-I and AD both groups with no APOE e4 allele. The color of each node represented the magnitude and direction of the associated iEN components developed from AD/HC-I classification model using all AD and HC-I data.
[0013] FIG.s 10A and 10B are correlation networks of HC-II/HC-I and HC-II/AD indicating that aging does not share key differential pSTATs and pPLCy2 signals highlighted by HC-I/ AD. FIG. 10A is a correlation network with node size corresponding to the Wilcoxon rank-sum testP value of each feature for HC-II/HC-I. The color of each node represented the magnitude and direction of the associated iEN components developed from AD/HC-I classification. FIG. 10B is similar to FIG. 10A however for the HC-II/AD pair.
[0014] FIG.s 11A-11C illustrate that cross disease prediction reveals similarities between AD and PD. FIG. 11A is a bar graph showing performance of the disease cross-prediction using iEN components developed from AD/HC-I diagnosis to classify PD/HC-Isub and AD/PD. FIG. 11B shows the iEN predicted values for each diagnostic group. FIG. 11C is a the correlation network with node size corresponding to the Wilcoxon rank-sum test P value of each feature for PD/HC-Isub diagnosis, with the color of each node representing the magnitude and direction of the associated iEN components developed from AD/HC-I. The network highlighted possible regions, such as in the labeled clusters, where AD and PD signals can overlap.
[0015] FIG. 12 illustrates the gating process to obtain different cell subsets. Cell subsets (35) were gated from DNA high, cleaned up PBMC and a matrix of 12 phospho-epitopes and 3 endosomal proteins with 8 stimuli conditions were evaluated from each subset.
[0016] FIG. 13 illustrates additional non-gender specific signals including first, pS6 expression in IFN-a and IL-6 stimulated CD4+ T cells, most CD8+ T cells, and NK cells. Second, pSTAT3 signals from specific subset of B cells including memory B cells, Naive B cells, and Switched memory B cells.
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS
[0017] The practice of the technology described herein will employ, unless indicated specifically to the contrary, conventional methods of chemistry, biochemistry, organic chemistry, molecular biology, microbiology, recombinant DNA techniques, genetics, immunology, and cell biology that are within the skill of the art, many of which are described below for the purpose of illustration. Examples of such techniques are available in the literature.
[0018] Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art. See, e.g., Singleton et al., DICTIONARY OF MICROBIOLOGY AND MOLECULAR BIOLOGY 2nd ed., J. Wiley & Sons (New York, NY 1994); Sambrook et al., MOLECULAR CLONING, A LABORATORY MANUAL, Cold Springs Harbor Press (Cold Springs Harbor, NY 1989). Any methods, devices and materials similar or equivalent to those described herein can be used in the practice of this invention. The following definitions are provided to facilitate understanding of certain terms used frequently herein and are not meant to limit the scope of the present disclosure. [0019] The terms “culture,” “culturing,” “grow,” “growing,” “maintain,” “maintaining,” “expand,” “expanding,” etc., when referring to cell culture itself or the process of culturing, can be used interchangeably to mean that a cell is maintained outside the body (e.g., ex vivo) under conditions suitable for survival. Cultured cells are allowed to survive, and culturing can result in cell growth, differentiation, or division. The term does not imply that all cells in the culture survive or grow or divide, as some may naturally senesce, etc. Cells are typically cultured in media, which can be changed during the course of the culture.
[0020] The terms “media” and “culture solution” refer to the cell culture milieu. Media is typically an isotonic solution, and can be liquid, gelatinous, or semi-solid, e.g., to provide a matrix for cell adhesion or support. Media, as used herein, can include the components for nutritional, chemical, and structural support necessary for culturing a cell.
[0021] As used herein, "conditions to allow growth" in culture and the like refers to conditions of temperature (typically at about 37° C for mammalian cells), humidity, CO2 (typically around 5%), in appropriate media (including salts, buffer, serum), such that the cells are able to undergo cell division or at least maintain viability for at least 24 hours, preferably longer (e.g., for days, weeks or months).
[0022] Suitable culture conditions are described herein, and can include standard tissue culture conditions. For example, PBMCs can be cultured in a buffered media that includes amino acids, nutrients, growth factors, etc., as will be understood in the art.
[0023] The term “derived from,” when referring to cells or a biological sample, indicates that the cell or sample was obtained from the stated source at some point in time. For example, a cell derived from an individual can represent a primary cell obtained directly from the individual (i.e., unmodified), or can be modified, e.g., by introduction of a recombinant vector, by culturing under particular conditions, or immortalization. In some cases, a cell derived from a given source will undergo cell division and / or differentiation such that the original cell is no longer exists, but the continuing cells will be understood to derive from the same source.
[0024] In this disclosure, “comprises,” “comprising,” “containing” and “having” and the like can have the meaning ascribed to them in U.S. Patent law and can mean “ includes,” “including,” and the like. “Consisting essentially of or “consists essentially” likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments.
[0025] An amino acid residue in a protein "corresponds" to a given residue when it occupies the same essential structural position within the protein as the given residue.
[0026] The term "isolated", when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It can be, for example, in a homogeneous state and may be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified.
[0027] The term "amino acid" refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, g- carboxyglutamate, and O-phosphoserine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i. e.. an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid. The terms “non-naturally occurring amino acid” and “unnatural amino acid” refer to amino acid analogs, synthetic amino acids, and amino acid mimetics which are not found in nature.
[0028] Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes. [0029] The terms "polypeptide," "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues, wherein the polymer may In embodiments be conjugated to a moiety that does not consist of amino acids. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. A "fusion protein" refers to a chimeric protein encoding two or more separate protein sequences that are recombinantly expressed as a single moiety.
[0030] A “detectable agent” or “detectable moiety” is a compound or composition detectable by appropriate means such as spectroscopic, photochemical, biochemical, immunochemical, chemical, magnetic resonance imaging, or other physical means. A detectable moiety is a monovalent detectable agent or a detectable agent bound (e.g. covalently and directly or via a linking group) with another compound, e.g., a nucleic acid. Exemplary detectable agents/moieties for use in the present disclosure include an antibody ligand, a peptide, a nucleic acid, radioisotopes, paramagnetic metal ions, fluorophore (e.g. fluorescent dyes), electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, a biotin-avidin complex, a biotin-streptavidin complex, digoxigenin, magnetic beads (e.g., DYNABEADS® by ThermoFisher, encompassing functionalized magnetic beads such as DYNABEADS® M-270 amine by ThermoFisher), paramagnetic molecules, paramagnetic nanoparticles, ultrasmall superparamagnetic iron oxide nanoparticles, ultrasmall superparamagnetic iron oxide nanoparticle aggregates, superparamagnetic iron oxide nanoparticles, superparamagnetic iron oxide nanoparticle aggregates, monocrystalline iron oxide nanoparticles, monocrystalline iron oxide, nanoparticle contrast agents, liposomes or other delivery vehicles containing Gadolinium chelate molecules, gadolinium, radionuclides (e.g. carbon-11, nitrogen-13, oxygen-15, fluorine-18, rubidium-82), fluorodeoxyglucose (e.g. fluorine- 18 labeled), any gamma ray emitting radionuclides, positron-emitting radionuclide, radiolabeled glucose, radiolabeled water, radiolabeled ammonia, biocolloids, microbubbles (e.g. including microbubble shells including albumin, galactose, lipid, and/or polymers; microbubble gas core including air, heavy gas(es), perfluorcarbon, nitrogen, octafluoropropane, perflexane lipid microsphere, perflutren, etc.), iodinated contrast agents (e.g. iohexol, iodixanol, ioversol, iopamidol, ioxilan, iopromide, diatrizoate, metrizoate, ioxaglate), barium sulfate, thorium dioxide, gold, gold nanoparticles, gold nanoparticle aggregates, fluorophores, two-photon fluorophores, or haptens and proteins or other entities which can be made detectable, e.g., by incorporating a radiolabel into a peptide or antibody specifically reactive with a target peptide. In embodiments, the detectable agent is a detectable fluorescent agent. In embodiments, the detectable agent is a detectable phosphorescent agent. In embodiments, the detectable agent is a detectable radioactive agent. In embodiments, the detectable agent is a detectable metalloenzyme. In embodiments, the detectable agent is a detectable colorimetric agent. In embodiments, the detectable agent is a detectable luminescent agent. In embodiments, the detectable agent is a detectable spectrophotometric agent. In embodiments, the detectable agent is a detectable metal-organic framework. In embodiments, the detectable agent is detectable by means other than by spectroscopy. In embodiments, the detectable agent comprises a fluorophore linked to biotin, avidin, or streptavidin. In embodiments, the detectable agent comprises a fluorophore linked to streptavidin. In embodiments, the detectable agent comprises a fluorophore linked to avidin. In embodiments, the detectable agent comprises a fluorophore linked to avidin linked to biotin. In embodiments, the detectable agent comprises a fluorophore linked to streptavidin linked to biotin.
[0031] The terms “selective” or “selectivity” or the like of a compound refers to the compound’s ability to discriminate between molecular targets.
[0032] The terms “specific”, “specifically”, “specificity”, or the like of a compound refers to the compound’s ability to cause a particular action, such as inhibition, to a particular molecular target with minimal or no action to other proteins in the cell.
[0033] The term "antibody" is used according to its commonly known meaning in the art. Antibodies exist, e.g., as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)'2, a dimer of Fab which itself is a light chain joined to VH-CHI by a disulfide bond. The F(ab)'2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)'2 dimer into an Fab' monomer. The Fab' monomer is essentially Fab with part of the hinge region (see Fundamental Immunology (Paul ed., 3d ed. 1993). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology. Thus, the term antibody, as used herein, also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv) or those identified using phage display libraries (see, e.g., McCafferty et al, Nature 348:552-554 (1990)).
[0034] An exemplary immunoglobulin (antibody) structural unit comprises a tetramer.
Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL), variable light chain (VL) domain or light chain variable region and variable heavy chain (VH), variable heavy chain (VH) domain or heavy chain variable region refer to these light and heavy chain regions, respectively. The terms variable light chain (VL), variable light chain (VL) domain and light chain variable region as referred to herein may be used interchangeably. The terms variable heavy chain (VH), variable heavy chain (VH) domain and heavy chain variable region as referred to herein may be used interchangeably. The Fc (i.e. fragment crystallizable region; also referred to herein as “Fc domain”) is the "base" or "tail" of an immunoglobulin and is typically composed of two heavy chains that contribute two or three constant domains depending on the class of the antibody. By binding to specific proteins, the Fc region ensures that each antibody generates an appropriate immune response for a given antigen. The Fc region also binds to various cell receptors, such as Fc receptors, and other immune molecules, such as complement proteins. In embodiments, the Fc region includes a constant heavy chain domain 3 (CH3 domain) and a constant heavy chain domain 2 (CH2 domain).
[0035] The epitope of an antibody is the region of its antigen to which the antibody binds. Two antibodies bind to the same or overlapping epitope if each competitively inhibits (blocks) binding of the other to the antigen. That is, a lx, 5x, lOx, 20x or lOOx excess of one antibody inhibits binding of the other by at least 30% but preferably 50%, 75%, 90% or even 99% as measured in a competitive binding assay (see, e.g., Junghans etal, Cancer Res. 50:1495, 1990). Alternatively, two antibodies have the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other. Two antibodies have overlapping epitopes if some amino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other. [0036] The term "CD" or "Cluster of Differentiation" refers to a nomenclature system for antigens found on lymphocytes, although CD antigens can be found on cells other than lymphocytes. This nomenclature is used to name antigens recognized by monoclonal antibodies that specifically bind an antigen on B cells, T cells or antigen presenting cells.
Each numeric antigen is a specific protein that is recognized in the art by its CD designation.
[0037] The term "CD3" as referred to herein is a protein complex comprising four chain including CD3y chain, a CD35 chain, and two CD3s chains. An example sequences of CD3 complex chains include: Epsilon chain precursor (GENBANK® Accession No.
NP_000724.1); Gamma chain precursor (GENBANK® Accession No. NP_000064.1); Delta chain precursor (GENBANK® Accession No. NP_000723.1) which are incorporated herein by reference. Multiple isoforms are possible for each of the chains of CD3.
[0038] The term "CD4" as referred to herein is a glycoprotein expressed on the surface of T helper cells, regulatory T cells, monocytes, macrophages, and dendritic cells. CD4 was originally known as leu-3 and T4 (after the OKT4 monoclonal antibody). CD4 as referred to herein has four immunoglobulin domains (Di to D4) that are exposed on the extracellular surface of the cell, see ENTREZ No. 920, UNIPROT No. P01730, and GENBANK® Accession No. NP 000607, which are incorporated by reference.
[0039] The term "CD8" as referred to herein is a transmembrane glycoprotein that serves as a co-receptor for the T cell receptor (TCR). Like the TCR, CD8 binds to a major histocompatibility complex {MS 1C) molecule, but is specific for the class I MHC protein, see ENTREZ No. 925 and UNIPROT No. R0G732, which are incorporated by reference herein.
[0040] The term "CD45RA" as provided herein refers to the CD45 Receptor antigen also known as Protein tyrosine phosphatase, receptor type, C (PTPRC). Exemplary amino acid sequences for CD45RA include GENBANK® Accession Nos. NP_002829.3, NP_563578.2, NP_563578.2, and NP_002829.3, which are all incorporated herein by reference. CD45RA is expressed on naive T cells, as well as on CD8- and CD4-expressing effector cells. After antigen interaction, T cells gain expression of CD45RO and lose expression of CD45RA. Thus, either CD45RA or CD45RO is used to generally differentiate the naive from memory T cell populations. Thus, a "CD45RA-negative CD8 T cell" as provided herein is a CD8 T cell which lacks expression of detectable levels of CD45RA. In embodiments, the CD45RA- negative CD8 T cell is a memory T cell. A "CD45RA-negative CD4 T cell" as provided herein is a CD4 T cell which lacks expression of detectable amounts of CD45RA. In embodiments, the CD45RA-negative CD4 T cell is a memory T cell. In embodiments, the CD45RA-negative CD8 T cell is a memory T cell.
[0041] The term "CD19 protein" or "CD19" as used herein includes any of the recombinant or naturally-occurring forms of B-lymphocyte antigen CD 19, also known as CD 19 molecule (Cluster of Differentiation 19), B-Lymphocyte Surface Antigen B4, T-Cell Surface Antigen Leu- 12 and CVID3, or variants or homologs thereof that maintain CD 19 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to CD 19).
In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring CD 19 protein. In embodiments, the CD 19 protein is substantially identical to the protein identified by the UniProt reference number P15391 or a variant or homolog having substantial identity thereto.
[0042] The term "CD20 protein" or "CD20" as used herein includes any of the recombinant or naturally-occurring forms of B-lymphocyte antigen CD20 or Cluster of Differentiation 20 (CD20), or variants or homologs thereof that maintain CD20 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to CD20). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring CD20 protein. In embodiments, the CD20 protein is substantially identical to the protein identified by the UniProt reference number PI 1836 or a variant or homolog having substantial identity thereto.
[0043] CD20 is involved in regulating early steps in the activation and differentiation process of B cells (Tedder et al., Eur. J. Immunol. 16:881-887, 1986) and can function as a calcium ion channel (Tedder et al., J. Cell. Biochem.14D:195, 1990). Exemplary amino acid sequences for CD20 are provided in GENBANK® Accession Nos. NP 068769.2 (human), NP 690605.1 (human), and NP 031667.1 (mouse), which are incorporated by reference herein.
[0044] The term "PLC-y2", "PLCy2", "PLC-y2 protein" or "PLCy2 protein" as provided herein includes any of the recombinant or naturally-occurring forms of the 1- Phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2 (PLC-y2) enzyme or variants or homologs thereof that maintain PLC-y2 enzyme activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to PLC-y2). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring PLC- y2 polypeptide. In embodiments, PLC-y2 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference PI 6885, homolog or functional fragment thereof. In embodiments, PLC-y2 is the protein as identified by the NCBI Reference Sequence: NP_002652.2, homolog or functional fragment thereof. In embodiments, PLC-y2 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_002661.5, homolog or functional fragment thereof.
[0045] The term " AKT " or "AKT protein" as provided herein includes any of the recombinant or naturally-occurring forms of the Protein kinase B (PKB), also known as AKT or variants or homologs thereof that maintain AKT kinase activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to AKT). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring AKT polypeptide. In embodiments, AKT is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P31749, homolog or functional fragment thereof. In embodiments, AKT is encoded by a nucleic acid sequence identified by NCBI Reference Sequence:
NM_005163.2, homolog or functional fragment thereof.
[0046] The term "STAT5" or "STAT5 protein" as provided herein includes any of the recombinant or naturally-occurring forms of the Signal transducer and Activator of transcription 5 (STAT5) or variants or homologs thereof that maintain STAT5 protein activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to STAT5). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring STAT5 polypeptide. In embodiments, STAT5 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P42229, homolog or functional fragment thereof. In embodiments, STAT5 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_003152.4, homolog or functional fragment thereof.
[0047] The term "STAT1" or "STAT1 protein" as provided herein includes any of the recombinant or naturally-occurring forms of the Signal transducer and Activator of transcription 1 (STAT1) or variants or homologs thereof that maintain STAT1 protein activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to STAT1). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring STAT1 polypeptide. In embodiments, STAT1 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P42224.2, homolog or functional fragment thereof. In embodiments, STAT1 is the protein as identified by the NCBI Reference Sequence: NP 644671, homolog or functional fragment thereof. In embodiments, STAT1 is the protein as identified by the NCBI Reference Sequence: NP_009330, homolog or functional fragment thereof. In embodiments, STAT1 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_007315.4, homolog or functional fragment thereof. In embodiments, STAT1 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_139266.3, homolog or functional fragment thereof.
[0048] The term "interferon a", "interferon a", "IFN a" or "IFN-a" as provided herein includes any of the recombinant or naturally-occurring forms of the interferon alpha protein (IFN-a) or variants or homologs thereof that maintain IFN-a protein activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IFN-a). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring IFN-a polypeptide. In embodiments, IFN-a is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P01562, homolog or functional fragment thereof. In embodiments, IFN-a is the protein as identified by the NCBI Reference Sequence: NP_076918.1, homolog or functional fragment thereof. In embodiments, IFN-a is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_024013.3, homolog or functional fragment thereof. [0049] The term "interleukin-6", "interleukin 6" or "IL-6" as provided herein includes any of the recombinant or naturally-occurring forms of the cytokine interleukin 6 (IL-6) or variants or homologs thereof that maintain IL-6 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IL-6). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring IL-6 polypeptide.
In embodiments, IL-6 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P05231, homolog or functional fragment thereof. In embodiments, IL-6 is the protein as identified by the NCBI Reference Sequence: NP_001358025.1, homolog or functional fragment thereof. In embodiments, IL-6 is the protein as identified by the NCBI Reference Sequence: NP_001305024.1, homolog or functional fragment thereof. In embodiments, IL-6 is the protein as identified by the NCBI Reference Sequence:
NP 000591.1, homolog or functional fragment thereof. In embodiments, IL-6 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001371096.1, homolog or functional fragment thereof. In embodiments, IL-6 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001318095.2, homolog or functional fragment thereof. In embodiments, IL-6 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_000600.5, homolog or functional fragment thereof.
[0050] The term "interleukin-7", "interleukin 7" or "IL-7" as provided herein includes any of the recombinant or naturally-occurring forms of the cytokine interleukin 7 (IL-7) or variants or homologs thereof that maintain IL-7 activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IL-7). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring IL-7 polypeptide.
In embodiments, IL-7 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference PI 3232, homolog or functional fragment thereof. In embodiments, IL-7 is the protein as identified by the NCBI Reference Sequence: NP_001186817.1, homolog or functional fragment thereof. In embodiments, IL-7 is the protein as identified by the NCBI Reference Sequence: NP_001186816.1, homolog or functional fragment thereof. In embodiments, IL-7 is the protein as identified by the NCBI Reference Sequence:
NP 001186815.1, homolog or functional fragment thereof. In embodiments, IL-7 is the protein as identified by the NCBI Reference Sequence: NP_000871.1, homolog or functional fragment thereof. In embodiments, IL-7 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001199888.2, homolog or functional fragment thereof. In embodiments, IL-7 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001199887.2, homolog or functional fragment thereof. In embodiments, IL-7 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence:
NM_001199886.2, homolog or functional fragment thereof. In embodiments, IL-7 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_000880.4, homolog or functional fragment thereof.
[0051] The term "interleukin- 10", "interleukin 10" or "IL-10" as provided herein includes any of the recombinant or naturally-occurring forms of the cytokine interleukin 10 (IL-10) or variants or homologs thereof that maintain IL-10 activity (e.g. within at least 50%, 80%,
90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IL-10). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring IL-10 polypeptide. In embodiments, IL-10 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference P22301, homolog or functional fragment thereof. In embodiments, IL-10 is the protein as identified by the NCBI Reference Sequence: NP_000563.1, homolog or functional fragment thereof. In embodiments, IL-10 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_000572.3, homolog or functional fragment thereof.
[0052] The term "interleukin-21", "interleukin 21" or "IL-21" as provided herein includes any of the recombinant or naturally-occurring forms of the cytokine interleukin 21 (IL-21) or variants or homologs thereof that maintain IL-21 activity (e.g. within at least 50%, 80%,
90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to IL-21). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the whole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring IL-21 polypeptide. In embodiments, IL-21 is the protein as identified by the UniProtKB/Swiss-Prot sequence reference Q9HBE4, homolog or functional fragment thereof. In embodiments, IL-21 is the protein as identified by the NCBI Reference Sequence: NP_068575.1, homolog or functional fragment thereof. In embodiments, IL-21 is the protein as identified by the NCBI Reference Sequence: NP 001193935.1, homolog or functional fragment thereof. In embodiments, IL- 21 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_001207006.3, homolog or functional fragment thereof. In embodiments, IL-21 is encoded by a nucleic acid sequence identified by NCBI Reference Sequence: NM_021803.4, homolog or functional fragment thereof.
[0053] The term "LPS", "lipopoly saccharide" or "lipopolysaccharides" or "endotoxins" as provided herein includes any of the recombinant or naturally-occurring forms of lipopoly saccharide (LPS) or variants or homologs thereof that maintain LPS activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to LPS). In some aspects, the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% structural identity across the whole polymer or a portion thereof compared to a naturally occurring LPS. Lipopolysaccharides (LPSs) as provided herein are components of the cell wall of Gram-negative bacteria. In embodiments, LPS and its lipid A moiety stimulate cells of the innate immune system by the Toll-like receptor 4 (TLR4), a member of the Toll-like receptor protein family, which recognizes common pathogen-associated molecular-patterns (PAMPs).
[0054] The term "PMA" or "phorbol myristate acetate" as provided herein includes any of the recombinant or naturally-occurring forms of 12-O-Tetradecanoylphorbol-l 3-acetate (TP A), also commonly known as tetradecanoylphorbol acetate, tetradecanoyl phorbol acetate, and phorbol 12-myristate 13-acetate (PMA) or variants or homologs thereof that maintain PMA activity (e.g. within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to PMA). In embodiments, PMA is the compound identified by Cas Registry Number 16561-29-8.
[0055] A "cell" as used herein, refers to a cell carrying out metabolic or other function sufficient to preserve or replicate its genomic DNA. A cell can be identified by well-known methods in the art including, for example, presence of an intact membrane, staining by a particular dye, ability to produce progeny or, in the case of a gamete, ability to combine with a second gamete to produce a viable offspring. Cells may include prokaryotic and eukaroytic cells. Prokaryotic cells include but are not limited to bacteria. Eukaryotic cells include but are not limited to yeast cells and cells derived from plants and animals, for example mammalian, insect (e.g., spodoptera) and human cells. Cells may be useful when they are naturally nonadherent or have been treated not to adhere to surfaces, for example by trypsinization. [0056] "B Cells" or "B lymphocytes" refer to their standard use in the art. B cells are lymphocytes, a type of white blood cell (leukocyte), that develops into a plasma cell (a “mature B cell”), which produces antibodies. An “immature B cell” is a cell that can develop into a mature B cell. Generally, pro-B cells undergo immunoglobulin heavy chain rearrangement to become pro B pre B cells, and further undergo immunoglobulin light chain rearrangement to become an immature B cells. Immature B cells include T1 and T2 B cells.
[0057] "T cells" or "T lymphocytes" as used herein are a type of lymphocyte (a subtype of white blood cell) that plays a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells, by the presence of a T-cell receptor on the cell surface. T cells include, for example, natural killer T (NKT) cells, cytotoxic T lymphocytes (CTLs), regulatory T (Treg) cells, and T helper cells.
Different types of T cells can be distinguished by use of T cell detection agents.
[0058] A "CD4+ T lymphocyte" or "CD4 T cell" as referred to herein is lymphocyte that expresses the CD4 glycoprotein on its surface. CD4 T cells include helper T cells, which are T cells that help orchestrate the immune response, including antibody responses and killer T cell responses. CD4 T cell precursors differentiate into one of several subtypes, including TH1 (type 1 helper T cell), TH2 (type 2 helper T cell), TH3 (T helper 3 cells), TH17 (T helper 17 cells) or TFH (Follicular B helper T cells). These subtypes of helper T cells are characterized by their secretion of different cytokines to facilitate different types of immune responses. In embodiments, a CD4 T cell is an effector T cell. An "effector T cell" as referred to herein is a T cell that has been activated by its cognate antigen, and is actively involved in eliminating a pathogen. Thus, an effector T cell actively responds to a stimulus (a pathogen or a co-stimulation) and carries out a cell-mediated immune response. Non- limiting examples of effector T cells as referred to herein include helper T cells, killer T cells (cytotoxic T cells) and regulatory T cells.
[0059] A "CD8+ T lymphocyte" or "CD8 T cell" as referred to herein is a lymphocyte that expresses the CD8 glycoprotein on its surface. Examples of CD8 T cells include cytotoxic T cells and natural killer cells. In one embodiment, a CD8 T cell is a cytotoxic T cell. In embodiments, a CDS T cell is a suppressor T cell.
[0060] A "memory T cell" is a T cell that has previously encountered and responded to its cognate antigen during prior infection, encounter with cancer or previous vaccination. At a second encounter with its cognate antigen memory T cells can reproduce (divide) to mount a faster and stronger immune response than the first time the immune system responded to the pathogen. In embodiments, the memory T cell is a CD45RA-negative CD4 T cell. In embodiments, the memory T cell is a CD45RA-negative CD8 T cell.
[0061] A "regulatory T cell" or "suppressor T cell" is a lymphocyte which modulates the immune system, maintains tolerance to self-antigens, and prevents autoimmune disease. Regulatory T cells express the CD4, FOXP3, and CD25 and are thought to be derived from the same lineage as naive CD4 cells.
[0062] The terms "PBMC", "peripheral blood mononuclear cell" or "PBMCs" are used according to their conventional meaning in the biological arts. Thus, a peripheral blood mononuclear cell (PBMC) refers to any peripheral blood cell having a round nucleus.
PBMCs are a cell population including lymphocytes (T cells, B cells, NK cells) and monocytes. In embodiments, PBMCs do not include cells that lack a nucleus. In embodiments, PBMCs do not include erythrocytes or platelets. In embodiments, PBMCs do not include erythrocytes. In embodiments, PBMCs do not include platelets. In embodiments, PBMCs do not include cells with a multi-lobed nucleus. In embodiments, PBMCs do not include granulocytes. In embodiments, PBMCs do not include neutrophils. In embodiments, PBMCs do not include basophils. In embodiments, PBMCs do not include eosinophils. In embodiments, lymphocytes make up the majority of the PBMC population, followed by monocytes, and only a small percentage of dendritic cells.
[0063] A "natural killer T cell" or "NKT cell" is a heterogeneous group of T cells including T cells and natural killer cells (Nk cells). In embodiments, natural killer T cells bind to non- polymorphic CD Id molecule. Natural killer T cells constitute approximately 1% of all peripheral blood T cells. In embodiments, the natural killer T cell is aNKl.l+ T cell. In embodiments, the natural killer T cell is a NK1.1- In embodiments, the natural killer T cell is a CD4+ T cell. In embodiments, the natural killer T cell is a CD4- T cell. In embodiments, the natural killer T cell is a CD8+ T cell. In embodiments, the natural killer T cell is aNKl.l+ T cell CD8- T cell. In embodiments, the natural killer T cell is CD16+ and CD56+ T cell. In embodiments, the natural killer T cell is granzyme producing T cell.
[0064] A "natural killer cell", "NK cell" or "large granular lymphocyte (LGL)" is a type of cytotoxic lymphocyte critical to the innate immune system. In embodiments, a NK cell is identified by the expression of CD56 and the lack of expression of CD3 (CD56+, CD3-). In embodiments, a NK cell is a CD56+ and CD3- lymphocyte. In embodiments, a NK cell is a CD56bright or CD56dim lymphocyte. CD56bright NK cells are similar to T helper cells in exerting their influence by releasing cytokines. In embodiments, CD56bright NK cells constitute the majority of NK cells in bone marrow, secondary lymphoid tissue, liver, and skin. In embodiments, CD56dim NK cells form part of the peripheral blood and are capable of cell killing. In embodiments, CD56dim NK cells are CD 16 positive lymphocytes. In embodiments, CD56bright transition to CD56dim by expressing CD16. NK cells may eliminate virus -infected cells by CD16-mediated ADCC (antibody-dependent cellular cytotoxicity).
[0065] The term "monocyte" as provided herein is used according to its conventional meaning in the biological arts. Thus, a monocyte refers to a leukocyte that is able to differentiate in macrophages or dendritic cells. In embodiments, a monocyte is a CD14++ and CD 16- leukocyte. In embodiments, a monocyte is a CD 14+ and CD 16+ leukocyte. In embodiments, a monocyte is a CD14++ and CD 16+ leukocyte.
[0066] The term "basophil" as provided herein is used according to its conventional meaning in the biological arts. Thus, a basophil refers to a leukocyte that is responsible for inflammatory reactions during immune response, as well as in the formation of acute and chronic allergic diseases, including anaphylaxis, asthma, atopic dermatitis and hay fever. In embodiments, a basophil is aFcsRI+, CD123+, CD49b(DX-5)+, CD69+, Thy-1.2+, 2B4+, CDllbdull, CD117(c-kit)-, CD24-, CD19-, CD80-, CD14-, CD23-, Ly49c-, CD122- CDllc-, Gr-1-, NK1.1-, B220-, CD3-, gd TCR-, ajlTCR-. a4 and 4-integrin negative leukocyte. In embodiments, a monocyte is a CD14++ and CD16+ cell. In embodiments, a basophil is a cell positive for CD13, CD44, CD54, CD63, CD69, CD107a, CD123, CD164, CD 193/ CCR3, CD203c, TLR-4, and FcsRI. In embodiments, a basophil is an activated basophil and expresses increased levels of CD13, CD107a, CD164, or surface-exposed CD63 and the ectoenzyme CD203c relative to a non-activated basophil.
[0067] The term "dendritic cell" or "DC" as provided herein is used according to its conventional meaning in the biological arts. Thus, a dendritic cell refers to an antigen- presenting cells (APC) or accessory cell of the mammalian immune system. A dendritic cell processes antigen material and presents it on the cell surface to the T cells of the immune system. Dendritic cells act as messengers between the innate and the adaptive immune systems. The term dendritic cells include "myeloid DCs" (mDCs) and "plasmacytoid dendritic cells" (pDCs). In embodiments, the dendritic cell is an mDC. In embodiments, an mDC is a mDC-1 and is a major stimulator of T cells. In embodiments, an mDC is an mDC- 2 and involved in wound infection. In embodiments, the mDC expresses Interleukin 12 (IL- 12), Interleukin 6 (IL-6), TNF, and/or chemokines. In embodiments, the mDC expresses TLR2 and/or TLR4. In embodiments, the dendritic cell is a pDC. In embodiments, the pDC produces high amounts of IFN-a. In embodiments, the pDC expresses TLR7 and/or TLR9.
In embodiments, the dendritic cell is a CDlc+ myeloid DC. In embodiments, the dendritic cell is a CD141+ myeloid DC. In embodiments, the dendritic cell is a CD303+ plasmacytoid DC.
[0068] The term "plasmablast" as provided herein is used according to its conventional meaning in the biological arts. Thus, a plasmablast is a cell in a short-lived differentiation stage between a post germinal center B-cell and a mature plasma cell. Plasmablasts retain a proliferative capability together with an almost fully mature plasma cell phenotype. In embodiments, a plasmablast is a CD19+, CD20-, IG+/-, CD27++ and CD38++ cell. In embodiments, a plasma cell is a CD19+/-, CD20-, Ig-, CD27++, CD38+++ and CD138+ cell.
[0069] “T cell detection agents” refers to a chemical or molecular moiety capable of identifying T cells. In examples, a T cell detection agent can be an antibody to a T cell specific surface maker (e.g. an antibody against CD3, and antibody against CD4, or an antibody against CD8). T cell detection agents can be used alone or in combination. T cell detection agents can further be detected by fluorescence activated cell sorting (FACS).
[0070] The term “cell subset detection agent” refers to a chemical or molecule detection agent that can be used to identify and distinguish a specific subset of cells (e.g. senescent cells, naive cells, effector cells, memory cells etc.). Example cell subset detection agents include “naive cell detection agents”, “memory cell detection agents”, and “effector cell detection agent.” Cell subset detection agents can include antibodies against distinguishing cell surface markers. In embodiments, cell subset detection agents include antibodies against CD27 or antibodies against CD45RA.
[0071] “Selective” or “selectivity” or the like of a compound refers to the compound’s ability to discriminate between molecular targets (e.g. a compound having selectivity toward HMT SUV39H1 and/or HMT G9a). [0072] An “inhibitor” refers to a compound (e.g. compounds described herein) that reduces activity when compared to a control, such as absence of the compound or a compound with known inactivity.
[0073] “Contacting” is used in accordance with its plain ordinary meaning and refers to the process of allowing at least two distinct species (e.g. chemical compounds including biomolecules or cells) to become sufficiently proximal to react, interact or physically touch.
It should be appreciated; however, the resulting reaction product can be produced directly from a reaction between the added reagents or from an intermediate from one or more of the added reagents that can be produced in the reaction mixture.
[0074] The term “contacting” may include allowing two species to react, interact, or physically touch, wherein the two species may be a compound as described herein and a protein or enzyme. In some embodiments contacting includes allowing a compound described herein to interact with a protein or enzyme that is involved in a signaling pathway.
[0075] As defined herein, the term “activation”, “activate”, “activating”, “activator” and the like in reference to a protein-inhibitor interaction means positively affecting (e.g. increasing) the activity or function of the protein relative to the activity or function of the protein in the absence of the activator. In embodiments activation means positively affecting (e.g. increasing) the concentration or levels of the protein relative to the concentration or level of the protein in the absence of the activator. The terms may reference activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein decreased in a disease. Thus, activation may include, at least in part, partially or totally increasing stimulation, increasing or enabling activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein associated with a disease (e.g., a protein which is decreased in a disease relative to a non-diseased control). Activation may include, at least in part, partially or totally increasing stimulation, increasing or enabling activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein
[0076] The terms “agonist,” “activator,” “upregulator,” etc. refer to a substance capable of detectably increasing the expression or activity of a given gene or protein. The agonist can increase expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the agonist. In certain instances, expression or activity is 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold or higher than the expression or activity in the absence of the agonist.
[0077] As defined herein, the term “inhibition”, “inhibit”, “inhibiting” and the like in reference to a protein-inhibitor interaction means negatively affecting (e.g. decreasing) the activity or function of the protein relative to the activity or function of the protein in the absence of the inhibitor. In embodiments inhibition means negatively affecting (e.g. decreasing) the concentration or levels of the protein relative to the concentration or level of the protein in the absence of the inhibitor. In embodiments inhibition refers to reduction of a disease or symptoms of disease. In embodiments, inhibition refers to a reduction in the activity of a particular protein target. Thus, inhibition includes, at least in part, partially or totally blocking stimulation, decreasing, preventing, or delaying activation, or inactivating, desensitizing, or down-regulating signal transduction or enzymatic activity or the amount of a protein. In embodiments, inhibition refers to a reduction of activity of a target protein resulting from a direct interaction (e.g. an inhibitor binds to the target protein). In embodiments, inhibition refers to a reduction of activity of a target protein from an indirect interaction (e.g. an inhibitor binds to a protein that activates the target protein, thereby preventing target protein activation).
[0078] The terms “inhibitor,” “repressor” or “antagonist” or “downregulator” interchangeably refer to a substance capable of detectably decreasing the expression or activity of a given gene or protein. The antagonist can decrease expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the antagonist. In certain instances, expression or activity is 1.5-fold, 2-fold, 3- fold, 4-fold, 5-fold, 10-fold or lower than the expression or activity in the absence of the antagonist.
[0079] The term "expression" includes any step involved in the production of the polypeptide including, but not limited to, transcription, post-transcriptional modification, translation, post-translational modification, and secretion. Expression can be detected using conventional techniques for detecting protein (e.g., ELISA, Western blotting, flow cytometry, immunofluorescence, immunohistochemistry, etc.).
[0080] The term “recombinant” when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all. Transgenic cells and animals are those that express a heterologous gene or coding sequence, typically as a result of recombinant methods.
[0081] The term “exogenous” refers to a molecule or substance (e.g., a compound, nucleic acid or protein) that originates from outside a given cell or organism. Conversely, the term “endogenous” refers to a molecule or substance that is native to, or originates within, a given cell or organism.
[0082] The term “modulator” refers to a composition that increases or decreases the level of a target molecule or the function of a target molecule or the physical state of the target of the molecule relative to the absence of the modulator.
[0083] The term “modulate” is used in accordance with its plain ordinary meaning and refers to the act of changing or varying one or more properties. “Modulation” refers to the process of changing or varying one or more properties. For example, as applied to the effects of a modulator on a target protein, to modulate means to change by increasing or decreasing a property or function of the target molecule or the amount of the target molecule.
[0084] “Control” or “control experiment” is used in accordance with its plain ordinary meaning and refers to an experiment in which the subjects or reagents of the experiment are treated as in a parallel experiment except for omission of a procedure, reagent, or variable of the experiment. In some instances, the control is used as a standard of comparison in evaluating experimental effects. In some embodiments, a control is the measurement of the activity of a protein in the absence of a compound as described herein (including embodiments and examples).
[0085] A “standard control” as provided herein refers to a sample that serves as a reference, usually a known reference, for comparison to a test sample. For example, a test sample can be taken from a patient suspected of having a disease (e.g., Alzheimer’s disease, Parkinson’s disease) and compared to samples from a patient known to have the disease, or a known normal (non-disease) individual. A control can also represent an average value gathered from a population of similar individuals, e.g., disease patients or healthy individuals with a similar medical background, same age, weight, etc. A control value can also be obtained from the same individual, e.g., from an earlier-obtained sample, prior to disease, or prior to treatment. One of skill will recognize that controls can be designed for assessment of any number of parameters. One of skill in the art will understand which controls are valuable in a given situation and be able to analyze data based on comparisons to control values. Controls are also valuable for determining the significance of data. For example, if values for a given parameter are widely variant in controls, variation in test samples will not be considered as significant.
[0086] In embodiments, a standard control is a level of phosphorylation of an intracellular signaling molecule from a sample or subject lacking the disease, a sample or subject at a selected stage of the disease or disease state, or in the absence of a particular variable such as a therapeutic agent. Alternatively, the control includes a known level of phosphorylation of an intracellular signaling molecule. Such a known level correlates with an average level of subjects lacking the disease, at a selected stage of the disease or disease state, or in the absence of a particular variable such as a therapeutic agent. A control also includes the level of phosphorylation of an intracellular signaling molecule from one or more selected samples or subjects as described herein. For example, a control includes the level of phosphorylation of an intracellular signaling molecule in a sample from a subject that does not have the disease, is at a selected stage of disease or disease state, or has not received treatment for the disease. Another exemplary control level includes an assessment of the level of phosphorylation of an intracellular signaling molecule in samples taken from multiple subjects that do not have the disease, are at a selected stage of the disease, or have not received treatment for the disease.
[0087] When the control level of phosphorylation of an intracellular signaling molecule includes the level of phosphorylation of an intracellular signaling molecule in a sample or subject in the absence of a therapeutic agent, the control sample or subject is optionally the same sample or subject to be tested before or after treatment with a therapeutic agent or is a selected sample or subject in the absence of the therapeutic agent. Alternatively, a standard control is an average expression level calculated from a number of subjects without a particular disease. A control level also includes a known control level or value known in the art.
[0088] A “standard control” as used herein in reference to the level of phosphorylation of one or more intracellular signaling molecules (e.g., PLC-y2, AKT, STAT1 or STAT5) refers to the level measured in a control subject (e.g. in a sample from the control subject) or population of control subjects. In embodiments, the control subject is a healthy control subject relative to the subject being tested, wherein the healthy control subject does not have a neurological disease. In embodiments, the control subject is the test subject prior to treatment of the test subject, wherein the test subject and control subject have a neurological disease. For example, in embodiments, the test subject has been treated for a neurological disease with a neurological disease treatment and the control subject is the test subject prior to treatment. In embodiments, the population of control subjects is a diverse collection of healthy subjects and diseased subjects, wherein the level of phosphorylation of an intracellular signaling molecule in the test subject is compared to the levels of the population of control subjects (e.g. an average of phosphorylation levels of the population of control subjects). In embodiments, the population of control subjects is a collection of healthy subjects that do not have a neurological disease, wherein the level of the test subject is compared to the levels of the population of control subjects (e.g. an average of expression levels of the population of control subjects). In embodiments, the population of control subjects is a collection of subjects that have been treated for a neurological disease, wherein the level of the test subject is compared to the levels of the population of control subjects (e.g. an average of levels of the population of control subjects). In embodiments, the control subject and the test subject are the same. In further embodiments, the standard control is the level of phosphorylation of an intracellular signaling molecule in a sample from healthy tissue.
[0089] The term “sample,” as used herein, refers to a composition that is obtained or derived from a subject and/or individual of interest that contains a cellular and/or other molecular entity that is to be characterized and/or identified, for example based on physical, biochemical, chemical and/or physiological characteristics. For example, the phrase “disease sample” and variations thereof refers to any sample obtained from a subject of interest that would be expected or is known to contain the cellular and/or molecular entity that is to be characterized. Samples include, but are not limited to, tissue extracts such as homogenized tissue, tumor tissue, cellular extracts, primary or cultured cells or cell lines, cell supernatants, cell lysates, platelets, serum, plasma, vitreous fluid, lymph fluid, synovial fluid, follicular fluid, seminal fluid, amniotic fluid, milk, whole blood, blood-derived cells, urine, cerebro spinal fluid, saliva, sputum, tears, perspiration, mucus, tumor lysates, and tissue culture medium, and combinations thereof. [0090] By “tissue sample” or “cell sample” is meant a collection of similar cells obtained from a tissue of a subject or individual. The source of the tissue or cell sample may be solid tissue as from a FFPE, FF, fresh, frozen, and/or preserved organ, tissue sample, biopsy, and/or aspirate; blood or any blood constituents such as plasma; bodily fluids such as cerebral spinal fluid, amniotic fluid, peritoneal fluid, or interstitial fluid; cells from any time in gestation or development of the subject. The tissue sample may also be primary or cultured cells or cell lines. Optionally, the tissue or cell sample is obtained from a disease (e.g., cancer) tissue/organ. The tissue sample may contain compounds which are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like.
[0091] The term “associated” or “associated with” in the context of a substance or substance activity or function associated with a disease (e.g. a protein associated disease, a cancer (e.g., cancer, inflammatory disease, autoimmune disease, or infectious disease)) means that the disease (e.g. cancer, inflammatory disease, autoimmune disease, or infectious disease) is caused by (in whole or in part), or a symptom of the disease is caused by (in whole or in part) the substance or substance activity or function. As used herein, what is described as being associated with a disease, if a causative agent, could be a target for treatment of the disease.
[0092] The term “aberrant” as used herein refers to different from normal. When used to describe enzymatic activity or protein function, aberrant refers to activity or function that is greater or less than a normal control or the average of normal non-diseased control samples. Aberrant activity may refer to an amount of activity that results in a disease, wherein returning the aberrant activity to a normal or non-disease-associated amount (e.g. by administering a compound or using a method as described herein), results in reduction of the disease or one or more disease symptoms.
[0093] “Pathway” refers to a set of system components involved in two or more sequential molecular interactions that result in the production of a product or activity. A pathway can produce a variety of products or activities that can include, for example, intermolecular interactions, changes in expression of a nucleic acid or polypeptide, the formation or dissociation of a complex between two or more molecules, accumulation or destruction of a metabolic product, activation or deactivation of an enzyme or binding activity. Thus, the term "pathway" includes a variety of pathway types, such as, for example, a biochemical pathway, a gene expression pathway, a regulatory pathway, or a combination thereof.
[0094] The term “signaling pathway” as used herein refers to a series of interactions between cellular and optionally extra-cellular components (e.g. proteins, nucleic acids, small molecules, ions, lipids) that conveys a change in one component to one or more other components, which in turn may convey a change to additional components, which is optionally propagated to other signaling pathway components.
[0095] The term “diagnosis” refers to an identification or likelihood of the presence of a particular type of neurological disease (e.g., Parkinson’s disease, Alzheimer’s disease) or outcome in a subject. The term “prognosis” refers to the likelihood or risk of a subject developing a particular outcome or particular event (e.g., Parkinson’s disease, Alzheimer’s disease).
[0096] The term “biological sample” encompasses essentially any sample type obtained from a subject that can be used in a diagnostic or prognostic method described herein. The biological sample may be any bodily fluid, tissue or any other suitable sample. The definition encompasses blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom and the progeny thereof. The definition also includes samples that have been manipulated in any way after their procurement, such as by treatment with reagents, solubilization, or enrichment for certain components, such as cells (e.g., cancer cells), polypeptides, or proteins. The term "biological sample" encompasses a clinical sample, but also, includes cells in culture, cell supernatants, cell lysates, blood, serum, plasma, urine, cerebral spinal fluid, biological fluid, and tissue samples. The sample may be pretreated as necessary by dilution in an appropriate buffer solution or concentrated, if desired. Any of a number of standard aqueous buffer solutions, employing one of a variety of buffers, such as phosphate, Tris, or the like, preferably at physiological pH can be used. Biological samples can be derived from patients using well- known techniques such as venipuncture, lumbar puncture, fluid sample such as saliva or urine, or tissue biopsy and the like. In embodiments, the sample is a blood sample (e.g., containing PBMCs).
[0097] As used herein, the term “neurological disorder” or “neurological disease” refers to a disease or condition in which the function of a subject’s nervous system becomes impaired. Examples of neurological diseases that may be treated with a compound, pharmaceutical composition, or method described herein include Alexander's disease, Alper's disease, Alzheimer's disease, Amyotrophic lateral sclerosis, Ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), Bovine spongiform encephalopathy (BSE), Canavan disease, chronic fatigue syndrome, Cockayne syndrome, Corticobasal degeneration, Creutzfeldt-Jakob disease, frontotemporal dementia, Gerstmann- Straussler-Scheinker syndrome, Huntington's disease, HIV-associated dementia, Kennedy's disease, Krabbe's disease, kuru, Lewy body dementia, Machado-Joseph disease (Spinocerebellar ataxia type 3), Multiple sclerosis, Multiple System Atrophy, myalgic encephalomyelitis, Narcolepsy, Neuroborreliosis, Parkinson's disease, Pelizaeus-Merzbacher Disease, Pick's disease, Primary lateral sclerosis, Prion diseases, Refsum's disease, Sandhoff s disease, Schilder's disease, Subacute combined degeneration of spinal cord secondary to Pernicious Anaemia, Schizophrenia, Spinocerebellar ataxia (multiple types with varying characteristics), Spinal muscular atrophy, Steele-Richardson-Olszewski disease , progressive supranuclear palsy, or Tabes dorsalis.
[0098] The terms “treating”, or “treatment” refers to any indicia of success in the therapy or amelioration of an injury, disease, pathology or condition (e.g., Parkinson’s disease, Alzheimer’s disease) including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the injury, pathology or condition more tolerable to the patient; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; improving a patient’s physical or mental well-being. The treatment or amelioration of symptoms can be based on objective or subjective parameters; including the results of a physical examination, neuropsychiatric exams, and/or a psychiatric evaluation. The term "treating" and conjugations thereof, may include prevention of an injury, pathology, condition, or disease. In embodiments, treating is preventing. In embodiments, treating does not include preventing.
[0099] “Treating” or “treatment” as used herein (and as well-understood in the art) also broadly includes any approach for obtaining beneficial or desired results in a subject’s condition (e.g., Parkinson’s disease, Alzheimer’s disease), including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of the extent of a disease, stabilizing (i.e., not worsening) the state of disease, prevention of a disease’s transmission or spread, delay or slowing of disease progression, amelioration or palliation of the disease state, diminishment of the reoccurrence of disease, and remission, whether partial or total and whether detectable or undetectable. In other words, "treatment" as used herein includes any cure, amelioration, or prevention of a disease. Treatment may prevent the disease from occurring; inhibit the disease’s spread; relieve the disease’s symptoms, fully or partially remove the disease’s underlying cause, shorten a disease’s duration, or do a combination of these things.
[0100] "Treating" and "treatment" as used herein include prophylactic treatment.
Treatment methods include administering to a subject a therapeutically effective amount of an active agent. The administering step may consist of a single administration or may include a series of administrations. The length of the treatment period depends on a variety of factors, such as the severity of the condition, the age of the patient, the concentration of active agent, the activity of the compositions used in the treatment, or a combination thereof. It will also be appreciated that the effective dosage of an agent used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required. For example, the compositions are administered to the subject in an amount and for a duration sufficient to treat the patient. In embodiments, the treating or treatment is no prophylactic treatment.
[0101] The term “prevent” refers to a decrease in the occurrence of disease symptoms in a patient. As indicated above, the prevention may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment.
[0102] “Patient” or “subject in need thereof’ refers to a living organism suffering from or prone to a disease or condition that can be treated by administration of a pharmaceutical composition as provided herein. Non-limiting examples include humans, other mammals, bovines, rats, mice, dogs, monkeys, goat, sheep, cows, deer, and other non-mammalian animals. In some embodiments, a patient is human.
[0103] A “effective amount” is an amount sufficient for a compound to accomplish a stated purpose relative to the absence of the compound (e.g. achieve the effect for which it is administered, treat a disease, reduce enzyme activity, increase enzyme activity, reduce a signaling pathway, or reduce one or more symptoms of a disease or condition). An example of an “effective amount” is an amount sufficient to contribute to the treatment, prevention, or reduction of a symptom or symptoms of a disease, which could also be referred to as a “therapeutically effective amount.” A “reduction” of a symptom or symptoms (and grammatical equivalents of this phrase) means decreasing of the severity or frequency of the symptom(s), or elimination of the symptom(s). A “prophylactically effective amount” of a drug is an amount of a drug that, when administered to a subject, will have the intended prophylactic effect, e.g., preventing or delaying the onset (or reoccurrence) of an injury, disease, pathology or condition, or reducing the likelihood of the onset (or reoccurrence) of an injury, disease, pathology, or condition, or their symptoms. The full prophylactic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a prophylactically effective amount may be administered in one or more administrations. An “activity decreasing amount,” as used herein, refers to an amount of antagonist required to decrease the activity of an enzyme relative to the absence of the antagonist. A “function disrupting amount,” as used herein, refers to the amount of antagonist required to disrupt the function of an enzyme or protein relative to the absence of the antagonist. The exact amounts will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); and Remington: The Science and Practice of Pharmacy, 20th Edition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins).
[0104] For any compound described herein, the therapeutically effective amount can be initially determined from cell culture assays. Target concentrations will be those concentrations of active compound(s) that are capable of achieving the methods described herein, as measured using the methods described herein or known in the art.
[0105] As is well known in the art, therapeutically effective amounts for use in humans can also be determined from animal models. For example, a dose for humans can be formulated to achieve a concentration that has been found to be effective in animals. The dosage in humans can be adjusted by monitoring compounds effectiveness and adjusting the dosage upwards or downwards, as described above. Adjusting the dose to achieve maximal efficacy in humans based on the methods described above and other methods is well within the capabilities of the ordinarily skilled artisan.
[0106] The term “therapeutically effective amount,” as used herein, refers to that amount of the therapeutic agent (e.g., a Parkinson’s disease therapeutic agent, Alzheimer’s disease therapeutic agent) sufficient to ameliorate the disorder, as described above. For example, for the given parameter, a therapeutically effective amount will show an increase or decrease of at least 5%, 10%, 15%, 20%, 25%, 40%, 50%, 60%, 75%, 80%, 90%, or at least 100%. Therapeutic efficacy can also be expressed as “-fold” increase or decrease. For example, a therapeutically effective amount can have at least a 1.2-fold, 1.5-fold, 2-fold, 5-fold, or more effect over a control.
[0107] Dosages may be varied depending upon the requirements of the patient and the compound being employed. The dose administered to a patient, in the context of the present disclosure, should be sufficient to effect a beneficial therapeutic response in the patient over time. The size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached. Dosage amounts and intervals can be adjusted individually to provide levels of the administered compound effective for the particular clinical indication being treated. This will provide a therapeutic regimen that is commensurate with the severity of the individual's disease state.
[0108] As used herein, the term "administering" means oral administration, administration as a suppository, topical contact, intravenous, parenteral, intraperitoneal, intramuscular, intralesional, intrathecal, intranasal or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject. Administration is by any route, including parenteral and transmucosal (e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal). Parenteral administration includes, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial. Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc. In embodiments, the administering does not include administration of any active agent other than the recited active agent.
[0109] "Co-administer" it is meant that a composition described herein is administered at the same time, just prior to, or just after the administration of one or more additional therapies. The compounds provided herein can be administered alone or can be coadministered to the patient. Coadministration is meant to include simultaneous or sequential administration of the compounds individually or in combination (more than one compound). Thus, the preparations can also be combined, when desired, with other active substances (e.g. to reduce metabolic degradation). The compositions of the present disclosure can be delivered transdermally, by a topical route, or formulated as applicator sticks, solutions, suspensions, emulsions, gels, creams, ointments, pastes, jellies, paints, powders, and aerosols.
METHODS
[0110] The methods provided herein are, inter alia, useful for diagnosing neurological disease (e.g., Alzheimer’s Disease, Parkinson’s disease). The methods provided herein may be used to detect specific phosphorylation levels of an intracellular signaling molecules such as, for example, PLC-y2, AKT, STAT1 or STAT5, in a subject having or being at risk of developing a neurological disease (e.g., Alzheimers disease or Parkinson’s disease). The phosphorylation levels of intracellular signaling molecules are detected in peripheral blood mononuclear cells (PBMCs) and subpopulations thereof, which have been stimulated with a stimulatory agent ex vivo (in, for example, a cell culture container after the PBMCs have been isolated from a subject). Stimulatory agents include, for example, interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA).
[0111] A “level of phosphorylation” as provided herein and in reference to intracellular signaling molecules is a detectable amount of phosphorylated intracellular signaling molecules or a detectable amount of phosphorylation of intracellular signaling molecules that are present in a PBMC after said PBMC has been contacted with a stimulatory agent. Thus, a level of phosphorylation may be increased relative to a standard control or it may be decreased relative to a standard control. In embodiments, the level of phosphorylation of an intracellular signaling molecule is increased relative to a standard control. In embodiments, the level of phosphorylation of an intracellular signaling molecule is decreased relative to a standard control. In embodiments, the standard control is a level of phosphorylation of an intracellular signaling molecule in the absence of a stimulatory agent. In embodiments, the level of phosphorylation of an intracellular signaling molecule is increased subsequent to the contacting of the PBMC with the stimulatory agent. In embodiments, the level of phosphorylation of an intracellular signaling molecule is decreased subsequent to the contacting of the PBMC with the stimulatory agent. [0112] An “intracellular signaling molecule” as provided herein refers to a protein inside a cell that forms part of a cellular signaling pathway. In embodiments, the intracellular signaling molecule is PLC-y2. AKT, STAT1 or STAT5. In embodiments, the intracellular signaling molecule is PLC-y2. In embodiments, the intracellular signaling molecule is AKT. In embodiments, the intracellular signaling molecule is STAT1. In embodiments, the intracellular signaling molecule is STAT5. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2, phosphorylated AKT, phosphorylated STAT1 or phosphorylated STAT5. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2. In embodiments, the intracellular signaling molecule is phosphorylated AKT. In embodiments, the intracellular signaling molecule is phosphorylated STAT1. In embodiments, the intracellular signaling molecule is phosphorylated STAT5.
[0113] The “expression level,” “amount,” or “level,” are used herein interchangeably, of a biomarker is a detectable level in a biological sample. “Expression” generally refers to the process by which information (e.g., gene-encoded and/or epigenetic) is converted into the structures present and operating in the cell. Therefore, as used herein, “expression” may refer to transcription into a polynucleotide, translation into a polypeptide, or even polynucleotide and/or polypeptide modifications (e.g., posttranslational modification of a polypeptide). Fragments of the transcribed polynucleotide, the translated polypeptide, or polynucleotide and/or polypeptide modifications (e.g., posttranslational modification of a polypeptide) shall also be regarded as expressed whether they originate from a transcript generated by alternative splicing or a degraded transcript, or from a post-translational processing of the polypeptide, e.g., by proteolysis. “Expressed genes” include those that are transcribed into a polynucleotide as mRNA and then translated into a polypeptide, and also those that are transcribed into RNA but not translated into a polypeptide (for example, transfer and ribosomal RNAs). Expression levels can be measured by methods known to one skilled in the art and also disclosed herein. The expression level or amount of a biomarker can be used to identify/characterize a subject having a neurological disease. The expression level or amount of a biomarker provided herein in a subject having a neurological disease described herein can also be used to determine and/or track the benefit of an administered neurological therapy over time. In embodiments, the biomarker is a level of phosphorylation of an intracellular signaling molecule. [0114] The term “biomarker” as used herein refers to an indicator, e.g., a predictive, prognostic, and/or a pharmacodynamic indicator which can be detected in a sample (e.g., a tissue sample, e.g., a peripheral blood sample). The biomarker may serve as an indicator of a particular type of a neurological disease or disorder (e.g., Parkinson’s disease, Alzheimer’s disease) characterized by certain molecular, pathological, histological, and/or clinical features. In some embodiments, a biomarker is a level of phosphorylation of an intracellular signaling molecule. Exemplary sets of biomarkers are found in Table 7.
[0115] A “stimulatory agent” as provided herein refers to an agent (e.g., interferon a (IFN- a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA) that positively affects (e.g. increases) the level of phosphorylation of an intracellular signaling molecule (e.g., PLC-y2, AKT, STAT1 or STAT5) relative to the level of phosphorylation of the intracellular signaling molecule in the absence of the agent. In embodiments, stimulating means positively affecting (e.g. increasing) the levels of phosphorylation of the intracellular signaling molecule relative to the levels of phosphorylation of the intracellular signaling molecule in the absence of the agent. In embodiments, stimulating means positively affecting (e.g. increasing) the amount of phosphorylated intracellular signaling molecules relative to the amount of phosphorylated intracellular signaling molecules in the absence of the agent. The stimulatory agent can increase the level of phosphorylation of an intracellular signaling molecule or the amount of phosphorylated intracellular signaling molecule 10%, 20%, 30%, 40%, 50%, 60%, 70%,
80%, 90% or more in comparison to a control level or control amount in the absence of the stimulatory agent. In certain instances, level or amount is 1.5-fold, 2-fold, 3-fold, 4-fold, 5- fold, 10-fold or higher than the level or amount in the absence of the stimulatory agent.
[0116] In embodiments, the stimulatory agent is any one of the agents listed in Table 5. In embodiments, the stimulatory agent is any one of the agents listed in Table 5 and is administered at a concentration as set forth in Table 5.
[0117] In embodiments, the stimulatory agent is IFN-a. In embodiments, IFN-a is administered at a concentration of about 10,000 units/ ml. In embodiments, IFN-a is administered at a concentration of 10,000 units/ ml. In embodiments, IFN-a is administered at a concentration of about 5,000 units/ ml. In embodiments, IFN-a is administered at a concentration of 5,000 units/ ml. In embodiments, IFN-a is administered at a concentration of about 15,000 units/ ml. In embodiments, IFN-a is administered at a concentration of 15,000 units/ ml. In embodiments, IFN-a is administered at a concentration of about 20,000 units/ ml. In embodiments, IFN-a is administered at a concentration of 20,000 units/ ml. In embodiments, IFN-a is administered at a concentration from about 5,000 units/ ml to about 20,000 units/ ml. In embodiments, IFN-a is administered at a concentration from 5,000 units/ ml to 20,000 units/ ml.
[0118] In embodiments, the stimulatory agent is IL-6. In embodiments, IL-6 is administered at a concentration of about 50 ng/ml. In embodiments, IL-6 is administered at a concentration of 50 ng/ml. In embodiments, IL-6 is administered at a concentration of about 10 ng/ml. In embodiments, IL-6 is administered at a concentration of 10 ng/ml. In embodiments, IL-6 is administered at a concentration of about 15 ng/ml. In embodiments, IL-6 is administered at a concentration of 15 ng/ml. In embodiments, IL-6 is administered at a concentration of about 20 ng/ml. In embodiments, IL-6 is administered at a concentration of 20 ng/ml. In embodiments, IL-6 is administered at a concentration of about 25 ng/ml. In embodiments, IL-6 is administered at a concentration of 25 ng/ml. In embodiments, IL-6 is administered at a concentration of about 30 ng/ml. In embodiments, IL-6 is administered at a concentration of 30 ng/ml. In embodiments, IL-6 is administered at a concentration of about 35 ng/ml. In embodiments, IL-6 is administered at a concentration of 35 ng/ml. In embodiments, IL-6 is administered at a concentration of about 40 ng/ml. In embodiments, IL-6 is administered at a concentration of 40 ng/ml. In embodiments, IL-6 is administered at a concentration of about 45 ng/ml. In embodiments, IL-6 is administered at a concentration of 45 ng/ml. In embodiments, IL-6 is administered at a concentration of about 55 ng/ml. In embodiments, IL-6 is administered at a concentration of 55 ng/ml. In embodiments, IL-6 is administered at a concentration of about 60 ng/ml. In embodiments, IL-6 is administered at a concentration of 60 ng/ml. In embodiments, IL-6 is administered at a concentration of about 65 ng/ml. In embodiments, IL-6 is administered at a concentration of 65 ng/ml. In embodiments, IL-6 is administered at a concentration of about 70 ng/ml. In embodiments, IL-6 is administered at a concentration of 70 ng/ml. In embodiments, IL-6 is administered at a concentration of about 80 ng/ml. In embodiments, IL-6 is administered at a concentration of 80 ng/ml. In embodiments, IL-6 is administered at a concentration of about 90 ng/ml. In embodiments, IL-6 is administered at a concentration of 90 ng/ml. In embodiments, IL-6 is administered at a concentration of about 100 ng/ml. In embodiments, IL-6 is administered at a concentration of 100 ng/ml. In embodiments, IL-6 is administered at a concentration from about 10 ng/ml to about 100 ng/ml. In embodiments, IL-6 is administered at a concentration from about 10 ng/ml to 100 ng/ml.
[0119] In embodiments, the stimulatory agent is IL-7. In embodiments, IL-7 is administered at a concentration of about 50 ng/ml. In embodiments, IL-7 is administered at a concentration of 50 ng/ml. In embodiments, IL-7 is administered at a concentration of about 10 ng/ml. In embodiments, IL-7 is administered at a concentration of 10 ng/ml. In embodiments, IL-7 is administered at a concentration of about 15 ng/ml. In embodiments, IL-7 is administered at a concentration of 15 ng/ml. In embodiments, IL-7 is administered at a concentration of about 20 ng/ml. In embodiments, IL-7 is administered at a concentration of 20 ng/ml. In embodiments, IL-7 is administered at a concentration of about 25 ng/ml. In embodiments, IL-7 is administered at a concentration of 25 ng/ml. In embodiments, IL-7 is administered at a concentration of about 30 ng/ml. In embodiments, IL-7 is administered at a concentration of 30 ng/ml. In embodiments, IL-7 is administered at a concentration of about 35 ng/ml. In embodiments, IL-7 is administered at a concentration of 35 ng/ml. In embodiments, IL-7 is administered at a concentration of about 40 ng/ml. In embodiments, IL-7 is administered at a concentration of 40 ng/ml. In embodiments, IL-7 is administered at a concentration of about 45 ng/ml. In embodiments, IL-7 is administered at a concentration of 45 ng/ml. In embodiments, IL-7 is administered at a concentration of about 55 ng/ml. In embodiments, IL-7 is administered at a concentration of 55 ng/ml. In embodiments, IL-7 is administered at a concentration of about 60 ng/ml. In embodiments, IL-7 is administered at a concentration of 60 ng/ml. In embodiments, IL-7 is administered at a concentration of about 65 ng/ml. In embodiments, IL-7 is administered at a concentration of 65 ng/ml. In embodiments, IL-7 is administered at a concentration of about 70 ng/ml. In embodiments, IL-7 is administered at a concentration of 70 ng/ml. In embodiments, IL-7 is administered at a concentration of about 80 ng/ml. In embodiments, IL-7 is administered at a concentration of 80 ng/ml. In embodiments, IL-7 is administered at a concentration of about 90 ng/ml. In embodiments, IL-7 is administered at a concentration of 90 ng/ml. In embodiments, IL-7 is administered at a concentration of about 100 ng/ml. In embodiments, IL-7 is administered at a concentration of 100 ng/ml. In embodiments, IL-7 is administered at a concentration from about 10 ng/ml to about 100 ng/ml. In embodiments, IL-7 is administered at a concentration from about 10 ng/ml to 100 ng/ml.
[0120] In embodiments, the stimulatory agent is IL-10. In embodiments, IL-10 is administered at a concentration of about 50 ng/ml. In embodiments, IL-10 is administered at a concentration of 50 ng/ml. In embodiments, IL-10 is administered at a concentration of about 10 ng/ml. In embodiments, IL-10 is administered at a concentration of 10 ng/ml. In embodiments, IL-10 is administered at a concentration of about 15 ng/ml. In embodiments, IL-10 is administered at a concentration of 15 ng/ml. In embodiments, IL-10 is administered at a concentration of about 20 ng/ml. In embodiments, IL-10 is administered at a concentration of 20 ng/ml. In embodiments, IL-10 is administered at a concentration of about 25 ng/ml. In embodiments, IL-10 is administered at a concentration of 25 ng/ml. In embodiments, IL-10 is administered at a concentration of about 30 ng/ml. In embodiments, IL-10 is administered at a concentration of 30 ng/ml. In embodiments, IL-10 is administered at a concentration of about 35 ng/ml. In embodiments, IL-10 is administered at a concentration of 35 ng/ml. In embodiments, IL-10 is administered at a concentration of about 40 ng/ml. In embodiments, IL-10 is administered at a concentration of 40 ng/ml. In embodiments, IL-10 is administered at a concentration of about 45 ng/ml. In embodiments, IL-10 is administered at a concentration of 45 ng/ml. In embodiments, IL-10 is administered at a concentration of about 55 ng/ml. In embodiments, IL-10 is administered at a concentration of 55 ng/ml. In embodiments, IL-10 is administered at a concentration of about 60 ng/ml. In embodiments, IL-10 is administered at a concentration of 60 ng/ml. In embodiments, IL-10 is administered at a concentration of about 65 ng/ml. In embodiments, IL-10 is administered at a concentration of 65 ng/ml. In embodiments, IL-10 is administered at a concentration of about 70 ng/ml. In embodiments, IL-10 is administered at a concentration of 70 ng/ml. In embodiments, IL-10 is administered at a concentration of about 80 ng/ml. In embodiments, IL-10 is administered at a concentration of 80 ng/ml. In embodiments, IL-10 is administered at a concentration of about 90 ng/ml. In embodiments, IL-10 is administered at a concentration of 90 ng/ml. In embodiments, IL-10 is administered at a concentration of about 100 ng/ml. In embodiments, IL-10 is administered at a concentration of 100 ng/ml. In embodiments, IL-10 is administered at a concentration from about 10 ng/ml to about 100 ng/ml. In embodiments, IL-10 is administered at a concentration from about 10 ng/ml to 100 ng/ml.
[0121] In embodiments, the stimulatory agent is IL-21. In embodiments, IL-21 is administered at a concentration of about 50 ng/ml. In embodiments, IL-21 is administered at a concentration of 50 ng/ml. In embodiments, IL-21 is administered at a concentration of about 10 ng/ml. In embodiments, IL-21 is administered at a concentration of 10 ng/ml. In embodiments, IL-21 is administered at a concentration of about 15 ng/ml. In embodiments, IL-21 is administered at a concentration of 15 ng/ml. In embodiments, IL-21 is administered at a concentration of about 20 ng/ml. In embodiments, IL-21 is administered at a concentration of 20 ng/ml. In embodiments, IL-21 is administered at a concentration of about 25 ng/ml. In embodiments, IL-21 is administered at a concentration of 25 ng/ml. In embodiments, IL-21 is administered at a concentration of about 30 ng/ml. In embodiments, IL-21 is administered at a concentration of 30 ng/ml. In embodiments, IL-21 is administered at a concentration of about 35 ng/ml. In embodiments, IL-21 is administered at a concentration of 35 ng/ml. In embodiments, IL-21 is administered at a concentration of about 40 ng/ml. In embodiments, IL-21 is administered at a concentration of 40 ng/ml. In embodiments, IL-21 is administered at a concentration of about 45 ng/ml. In embodiments, IL-21 is administered at a concentration of 45 ng/ml. In embodiments, IL-21 is administered at a concentration of about 55 ng/ml. In embodiments, IL-21 is administered at a concentration of 55 ng/ml. In embodiments, IL-21 is administered at a concentration of about 60 ng/ml. In embodiments, IL-21 is administered at a concentration of 60 ng/ml. In embodiments, IL-21 is administered at a concentration of about 65 ng/ml. In embodiments, IL-21 is administered at a concentration of 65 ng/ml. In embodiments, IL-21 is administered at a concentration of about 70 ng/ml. In embodiments, IL-21 is administered at a concentration of 70 ng/ml. In embodiments, IL-21 is administered at a concentration of about 80 ng/ml. In embodiments, IL-21 is administered at a concentration of 80 ng/ml. In embodiments, IL-21 is administered at a concentration of about 90 ng/ml. In embodiments, IL-21 is administered at a concentration of 90 ng/ml. In embodiments, IL-21 is administered at a concentration of about 100 ng/ml. In embodiments, IL-21 is administered at a concentration of 100 ng/ml. In embodiments, IL-21 is administered at a concentration from about 10 ng/ml to about 100 ng/ml. In embodiments, IL-21 is administered at a concentration from about 10 ng/ml to 100 ng/ml.
[0122] In embodiments, the stimulatory agent is LPS. In embodiments, LPS is administered at a concentration of about 1 ug/ml. In embodiments, LPS is administered at a concentration of 1 ug/ml. In embodiments, LPS is administered at a concentration of about 2 ug/ml. In embodiments, LPS is administered at a concentration of 2 ug/ml. In embodiments, LPS is administered at a concentration of about 3 ug/ml. In embodiments, LPS is administered at a concentration of 3 ug/ml. In embodiments, LPS is administered at a concentration of about 4 ug/ml. In embodiments, LPS is administered at a concentration of 4 ug/ml. In embodiments, LPS is administered at a concentration of about 5 ug/ml. In embodiments, LPS is administered at a concentration of 5 ug/ml. In embodiments, LPS is administered at a concentration of about 10 ug/ml. In embodiments, LPS is administered at a concentration of 10 ug/ml. In embodiments, LPS is administered at a concentration from about 1 ug/ml to about 10 ug/ml. In embodiments, LPS is administered at a concentration from 1 ug/ml to 10 ug/ml.
[0123] In embodiments, the stimulatory agent is PMA. In embodiments, PMA is administered at a concentration of about 100 ng/ml. In embodiments, PMA is administered at a concentration of 100 ng/ml. In embodiments, PMA is administered at a concentration of about 10 ng/ml. In embodiments, PMA is administered at a concentration of 10 ng/ml. In embodiments, PMA is administered at a concentration of about 50 ng/ml. In embodiments, PMA is administered at a concentration of 50 ng/ml. In embodiments, PMA is administered at a concentration of about 150 ng/ml. In embodiments, PMA is administered at a concentration of 150 ng/ml. In embodiments, PMA is administered at a concentration from about 10 ng/ml to about 150 ng/ml. In embodiments, PMA is administered at a concentration from 10 ng/ml to 150 ng/ml.
[0124] In one aspect, a method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease is provided. The method includes (i) obtaining or having obtained a sample from a subject having or being at risk of developing a neurological disease; (ii) isolating a peripheral blood mononuclear cell (PBMC) from the sample; wherein the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC); (iii) contacting the PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopolysaccharides (LPS) or phorbol myristate acetate (PMA); and (iv) detecting a level of phosphorylation of an intracellular signaling molecule in the ex vivo stimulated PBMC, wherein the intracellular signaling molecule is PLC-y2, AKT, STAT1 or STAT5.
[0125] In one aspect, a method of treating a neurological disease in a subject in need thereof is provided. The method includes (i) obtaining or having obtained a sample from a subject having a neurological disease; (ii) isolating a peripheral blood mononuclear cell (PBMC) from the sample; wherein the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC); (iii) contacting the PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA); (iv) detecting a level of phosphorylation of an intracellular signaling molecule in the ex vivo stimulated PBMC, wherein the intracellular signaling molecule is PLC-y2, AKT, STAT1 or STAT5; and (v) administering to the subject a therapeutically effective amount of a neurological treatment.
[0126] In one aspect, a method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject undergoing treatment for a neurological disease is provided. The method includes (i) obtaining or having obtained a sample from a subject undergoing treatment for a neurological disease; (ii) isolating a peripheral blood mononuclear cell (PBMC) from the sample; wherein the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC); (iii) contacting the PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA); and (iv) detecting a level of phosphorylation of an intracellular signaling molecule in the ex vivo stimulated PBMC, wherein the intracellular signaling molecule is PLC-y2, AKT, STAT1 or STAT5.
[0127] In one aspect, a method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease, the method including: (i) obtaining or having obtained a sample from a subject having or being at risk of developing a neurological disease; (ii) isolating a peripheral blood mononuclear cell (PBMC) from the sample; wherein the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC); (iii) contacting the PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopolysaccharides (LPS) or phorbol myristate acetate (PMA); (iv) detecting a level of phosphorylation of an intracellular signaling molecule in the ex vivo stimulated PBMC, wherein the intracellular signaling molecule is PLC-y2, AKT, STAT1 or STAT5; and (v) comparing the level of phosphorylation to a standard control, thereby detecting a level of phosphorylation of an intracellular signaling molecule in a subject.
[0128] In embodiments, the PBMC is a CD4+ T cell. In embodiments, the PBMC is a CD8+ T cell. In embodiments, the PBMC is a natural killer T cell. In embodiments, the PBMC is a natural killer (NK) cell. In embodiments, the PBMC is a B cell. In embodiments, the PBMC is a monocyte. In embodiments, the PBMC is a basophil. In embodiments, the PBMC is a plasmablast. In embodiments, the PBMC is a dendritic cell (DC). In embodiments, the PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast and a dendritic cell (DC).
[0129] In embodiments, the stimulatory agent is interferon a (IFN-a). In embodiments, the stimulatory agent is interleukin-6 (IL-6). In embodiments, the stimulatory agent is interleukin-7 (IL-7). In embodiments, the stimulatory agent is interleukin- 10 (IL-10). In embodiments, the stimulatory agent is interleukin-21 (IL-21). In embodiments, the stimulatory agent is lipopolysaccharides (LPS). In embodiments, the stimulatory agent is phorbol myristate acetate (PMA). In embodiments, the stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL- 21), lipopolysaccharides (LPS) and phorbol myristate acetate (PMA). For the methods provided herein the PBMCs are contacted with the stimulatory agent in a non-natural environment, such as an in vitro cell culture container. In embodiments, the cell culture container (e.g., petri dish) forms part of a detection device.
[0130] In embodiments, the intracellular signaling molecule is PLC-y2. In embodiments, the intracellular signaling molecule is AKT. In embodiments, the intracellular signaling molecule is STAT1. In embodiments, the intracellular signaling molecule is STAT5. In embodiments, the intracellular signaling molecule is PLC-y2, AKT, STAT1 and STAT5. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2. In embodiments, the intracellular signaling molecule is phosphorylated AKT. In embodiments, the intracellular signaling molecule is phosphorylated STAT1. In embodiments, the intracellular signaling molecule is phosphorylated STAT5. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2, phosphorylated AKT, phosphorylated STAT1 and phosphorylated STAT5. [0131] In embodiments, an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In embodiments, a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In embodiments, the method includes based at least in part on the level of phosphorylation in step (v) administering a neurological disease treatment to the subject. In embodiments, the neurological disease treatment is an Alzheimer’s disease treatment. In embodiments, the neurological disease treatment is a dementia treatment. In embodiments, the neurological disease treatment is a Parkinson’s disease treatment.
[0132] When the stimulatory agent is “unstimulated”, the PBMCs are contacted with a non stimulatory (inert, non-active) agent (e.g., a cell culture buffer). A non-stimulatory agent as provided herein is an agent that upon contacting with the PBMC does not detectably increase or decrease the level of phosphorylation of an intracellular signaling molecule provided herein. In other words, the level of phosphorylation of the intracellular signaling molecule in presence of a non-stimulatory agent is the same as the level of phosphorylation of the intracellular signaling molecule in the absence of the non-stimulatory agent.
[0133] In embodiments, the intracellular signaling molecule is PLC-y2. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2.
[0134] In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell. In embodiments, the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL- 10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0135] In embodiments, the stimulatory agent is unstimulated and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell. In embodiments, the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0136] In embodiments, the stimulatory agent is IL-10 and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell. In embodiments, the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0137] In embodiments, the stimulatory agent is IL-21 and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell. In embodiments, the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0138] In embodiments, the stimulatory agent is LPS and the PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell or a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell. In embodiments, the stimulatory agent is IL-21 or LPS and the PBMC is a CD16high monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0139] In embodiments, the stimulatory agent is IL-6 and the PBMC is a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell. In embodiments, the stimulatory agent is IL-6 and the PBMC is a CD8+ activated T cell. In embodiments, the stimulatory agent is IL- 6 and the PBMC is a CD4+ CD8+ T cell or a natural killer T cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0140] In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD56bright NK cell, or a CD16high monocyte. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a natural killer T cell. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD8+ activated T cell or a CD16high monocyte. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, or a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0141] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD56bright NK cell, or a CD16high monocyte. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a natural killer T cell. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD8+ activated T cell or a CD16high monocyte. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, or a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0142] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD56bright NK cell, or a CD16high monocyte. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a natural killer T cell. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD8+ activated T cell or a CD16high monocyte. In embodiments, the stimulatory agent is IFN-a or IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, or a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one further embodiment, the standard control is a healthy subject.
[0143] In embodiments, the intracellular signaling molecule is STAT1. In embodiments, the intracellular signaling molecule is phosphorylated STAT1.
[0144] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD56bright NK cell, a myeloid dendritic cell (mDC) or a plasmacytoid dendritic cell (pDC). In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject.
[0145] In embodiments, the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject.
[0146] In embodiments, the stimulatory agent is LPS and the PBMC is a CD8+ central memory T cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. [0147] In embodiments, the intracellular signaling molecule is STAT5. In embodiments, the intracellular signaling molecule is phosphorylated STAT5.
[0148] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, a CD16high monocyte or a CD161ow monocyte. In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD16high monocyte or a CD161ow monocyte. In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, or a CD4- CD8- T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject.
[0149] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, or a CD8+ effector memory T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject.
[0150] In embodiments, the stimulatory agent is LPS and the PBMC is a CD4+ CD8+ T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject.
[0151] In embodiments, the subject is a male subject.
[0152] In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dimNK cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0153] In embodiments, the stimulatory agent is unstimulated and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0154] In embodiments, the stimulatory agent is IL-10 and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dimNK cell, a CD16high monocyte or a CD161ow monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0155] In embodiments, the stimulatory agent is IL-21 and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dimNK cell, a CD16high monocyte or a CD161ow monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0156] In embodiments, the stimulatory agent is LPS and the PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell. In embodiments, the stimulatory agent is unstimulated, IL-10, IL-21 or LPS and the PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0157] In embodiments, the stimulatory agent is IFN-a, IL-6 or IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0158] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0159] In embodiments, the stimulatory agent is IL-6 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0160] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0161] In embodiments, the stimulatory agent is PMA and the PBMC is a CD4+ activated T cell, a CD4+ naive T cell, a CD8+ activated T cell, a natural killer T cell, a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a B switched memory cell, or a B translational cell. In embodiments, the stimulatory agent is PMA and the PBMC is a CD4+ activated T cell, a CD4+ naive T cell, or a natural killer T cell. In embodiments, the stimulatory agent is PMA and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a B switched memory cell, or a B translational cell. In embodiments, the stimulatory agent is PMA and the PBMC is a CD8+ activated T cell or a CD4+ CD8+ T cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0162] In embodiments, the intracellular signaling molecule is AKT. In embodiments, the intracellular signaling molecule is phosphorylated AKT.
[0163] In embodiments, the subject is a male subject.
[0164] In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0165] In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0166] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0167] In embodiments, the stimulatory agent is IL-6 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0168] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0169] In embodiments, the stimulatory agent is IL-10 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0170] In embodiments, the stimulatory agent is IL-21 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0171] In embodiments, the stimulatory agent is LPS and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0172] In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell. In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0173] In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell. In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0174] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell. In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0175] In embodiments, the stimulatory agent is IL-6 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell. In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0176] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell. In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0177] In embodiments, the stimulatory agent is IL-10 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell. In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0178] In embodiments, the stimulatory agent is IL-21 and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell. In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0179] In embodiments, the stimulatory agent is LPS and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In embodiments, the stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and the PBMC is a CD4+ effector memory T cell. In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0180] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD4+ activated T cell. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0181] In embodiments, the stimulatory agent is PMA and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a mDC, a pDC, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is AKT and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is AKT and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy male subject.
[0182] In embodiments, the subject is a female subject.
[0183] In embodiments, the stimulatory agent is unstimulated, IL-7, IL-10 or LPS and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0184] In embodiments, the stimulatory agent is unstimulated and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell.
In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0185] In embodiments, the stimulatory agent is IL-7 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0186] In embodiments, the stimulatory agent is IL-10 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0187] In embodiments, the stimulatory agent is LPS and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0188] In embodiments, the stimulatory agent is unstimulated and the PBMC is a B naive cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0189] In embodiments, the stimulatory agent is unstimulated, IL-7, IL-10 or LPS and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0190] In embodiments, the stimulatory agent is unstimulated and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0191] In embodiments, the stimulatory agent is IL-7 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0192] In embodiments, the stimulatory agent is IL-10 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0193] In embodiments, the stimulatory agent is LPS and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0194] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a CD4- CD8- T cell, a natural killer T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, a mDC, a pDC, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0195] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, or a pDC. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0196] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD4+ CD8+ T cell, a CD4- CD8- T cell, a natural killer T cell, a plasmablast cell, or a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0197] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ naive T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a mDC, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0198] In embodiments, the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject. [0199] In embodiments, the stimulatory agent is IL-6 and the PBMC is an IgD- CD27- B cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0200] In embodiments, the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, or a B naive cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0201] In embodiments, the stimulatory agent is IL-21 and the PBMC is an IgD- CD27- B cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0202] In embodiments, the subject is a female subject. In embodiments, the intracellular signaling molecule is STAT5. In embodiments, the intracellular signaling molecule is phosphorylated STAT5.
[0203] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0204] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD8+ effector memory T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0205] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ effector T cell, a CD8+ central memory T cell, a CD8+ naive T cell, or a natural killer T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0206] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD4+ activated T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ effector T cell, a CD4- CD8- T cell, a CD4+ CD8+ T cell, a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0207] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD4- CD8- T cell, a CD4+ CD8+ T cell or a natural killer T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0208] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD8+ central memory T cell, or a CD8+ effector memory T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject. [0209] In embodiments, the stimulatory agent is IL-7 and the PBMC is a natural killer T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0210] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD8+ effector T cell, a CD4- CD8- T cell, or a CD4+ CD8+ T cell. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the standard control is a healthy female subject.
[0211] In embodiments, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0212] In embodiments, the intracellular signaling molecule is PLC-y2. In embodiments, the intracellular signaling molecule is phosphorylated PLC-y2. [0213] In embodiments, the stimulatory agent is unstimulated and the PBMC is a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0214] In embodiments, the stimulatory agent is IL-10, IL-21 or LPS and the PBMC is a basophil. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0215] In embodiments, the stimulatory agent is IL-10 and the PBMC is a basophil. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0216] In embodiments, the stimulatory agent is IL-21 and the PBMC is a basophil. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0217] In embodiments, the stimulatory agent is LPS and the PBMC is a basophil. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0218] In embodiments, the intracellular signaling molecule is STAT1.
[0219] In embodiments, the stimulatory agent is IFN-a and the PBMC is a plasmablast or CD56bright NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0220] In embodiments, the intracellular signaling molecule is STAT5.
[0221] In embodiments, the stimulatory agent is IFN-a and the PBMC is a CD16high monocyte or a CD161ow monocyte. In one embodiment, the intracellular signaling molecule is STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT5 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0222] In embodiments, the neurological disease is Alzheimer’s disease.
[0223] In embodiments, the intracellular signaling molecule is PLC-y2.
[0224] In embodiments, the stimulatory agent is IL-10, IL-21, LPS or PMA and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0225] In embodiments, the stimulatory agent is IL-10 and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0226] In embodiments, the stimulatory agent is IL-21 and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0227] In embodiments, the stimulatory agent is LPS and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0228] In embodiments, the stimulatory agent is PMA and the PBMC is a CD4+ CD8+ T cell, an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0229] In embodiments, the stimulatory agent is IL-10, IL-21, LPS or PMA and the PBMC is an IgA- B cell or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease. [0230] In embodiments, the stimulatory agent is IL-10 and the PBMC is an IgA- B cell or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0231] In embodiments, the stimulatory agent is IL-21 and the PBMC is an IgA- B cell or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0232] In embodiments, the stimulatory agent is LPS and the PBMC is an IgA- B cell or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0233] In embodiments, the stimulatory agent is PMA and the PBMC is an IgA- B cell or a B translational cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0234] In embodiments, the stimulatory agent is IL-10, IL-21, LPS or PMA and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0235] In embodiments, the stimulatory agent is IL-10 and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0236] In embodiments, the stimulatory agent is IL-21 and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0237] In embodiments, the stimulatory agent is LPS and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0238] In embodiments, the stimulatory agent is PMA and the PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, or a B switched memory cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0239] In embodiments, the stimulatory agent is IL-7, IL-10, or IL-21 and the PBMC is a CD8+ activated T cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0240] In embodiments, the stimulatory agent is IL-7 and the PBMC is a CD8+ activated T cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0241] In embodiments, the stimulatory agent is IL-10 and the PBMC is a CD8+ activated T cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0242] In embodiments, the stimulatory agent is IL-21 and the PBMC is a CD8+ activated T cell. In one embodiment, the intracellular signaling molecule is PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated PLC-y2 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0243] In embodiments, the intracellular signaling molecule is STAT1.
[0244] In embodiments, the stimulatory agent is unstimulated, and the PBMC is an IgD+ B memory cell, an IgD- CD27- B cell or a B naive cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0245] In embodiments, the stimulatory agent is IL-6 or IL-7 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0246] In embodiments, the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0247] In embodiments, the stimulatory agent is IL-7 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0248] In embodiments, the stimulatory agent is IL-6 and the PBMC is an IgA- B cell, an IgD+ B memory cell, an IgD- CD27- B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0249] In embodiments, the stimulatory agent is LPS or PMA and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0250] In embodiments, the stimulatory agent is LPS and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0251] In embodiments, the stimulatory agent is PMA and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0252] In embodiments, the stimulatory agent is LPS or PMA and the PBMC is a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0253] In embodiments, the stimulatory agent is LPS and the PBMC is a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0254] In embodiments, the stimulatory agent is PMA and the PBMC is a CD56dimCD16dim NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease. [0255] In embodiments, the stimulatory agent is LPS or PMA and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, or a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0256] In embodiments, the stimulatory agent is PMA and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, or a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0257] In embodiments, the stimulatory agent is LPS and the PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD- CD27- B cell, a B naive cell, a CD16high NK cell, or a CD56bright NK cell. In one embodiment, the intracellular signaling molecule is STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and an increased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the intracellular signaling molecule is phosphorylated STAT1 and a decreased level of phosphorylation relative to the standard control indicates that the subject has or is at risk of developing a neurological disease. In one embodiment, the standard control is a healthy subject. In one embodiment, the neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0258] In embodiments, the sample from the subject is a blood sample or a plasma sample. In embodiments, the sample from the subject is a blood sample. In embodiments, the sample from the subject is a plasma sample. In embodiments, the detecting includes contacting the ex vivo stimulated PBMC with one or more immune cell specific antibodies, thereby forming a labeled ex vivo stimulated PBMC. In embodiments, the one or more immune cell-specific antibodies includes a detectable moiety.
[0259] In embodiments, the one or more immune cell-specific antibodies are an anti-CD3, an anti-CD4 antibody, an anti-CD7 antibody, an anti-CD8 antibody, an anti-CDllb antibody, an anti-CDllc antibody, an anti-CD14 antibody, an anti-CD16 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti-CD24 antibody, an anti-CD25 antibody, an anti- CD27 antibody, an anti-CD38 antibody, an anti-CD45RA antibody, an anti-CD56 antibody, an anti-CD 123 antibody, an anti-CD 127 antibody, an anti -IgA antibody, an anti-IgD antibody or an anti-HLA-Dr antibody. In embodiments, the one or more immune cell-specific antibodies are selected from a group of antibodies consisting of an anti-CD3, an anti-CD4 antibody, an anti-CD7 antibody, an anti-CD8 antibody, an anti-CDllb antibody, an anti- CDllc antibody, an anti-CD14 antibody, an anti-CD16 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti-CD24 antibody, an anti-CD25 antibody, an anti-CD27 antibody, an anti-CD38 antibody, an anti-CD45RA antibody, an anti-CD56 antibody, an anti-CD123 antibody, an anti-CD 127 antibody, an anti-IgA antibody, an anti-IgD antibody and an anti- HLA-Dr antibody.
[0260] In embodiments, the detecting further includes contacting the labeled ex vivo stimulated PBMC with one or more anti-intracellular signaling molecule antibodies, thereby forming a intracellularly labeled ex vivo stimulated PBMC. In embodiments, the one or more anti-intracellular signaling molecule antibodies are an anti-pERKl/2 antibody, an anti-IkBa antibody, an anti-NF-kB antibody, an anti-p38 antibody, an anti-pAKT antibody, an anti- pCREB antibody, an anti-pLCK antibody, an anti-pPLC-y2 antibody, an anti-pS6 antibody, an anti-pSTATl antibody, an anti-pSTAT3 antibody, an anti-pSTAT5 antibody, an anti- Lamp2 antibody, an anti-EEAl antibody, or an anti-Rab5 antibody. In embodiments, the one or more anti-intracellular signaling molecule antibodies are selected from a group of antibodies consisting of an anti-pERKl/2 antibody, an anti-IkBa antibody, an anti-NF-kB antibody, an anti-p38 antibody, an anti-pAKT antibody, an anti-pCREB antibody, an anti- pLCK antibody, an anti-pPLC-y2 antibody, an anti-pS6 antibody, an anti-pSTATl antibody, an anti-pSTAT3 antibody, an anti-pSTAT5 antibody, an anti-Lamp2 antibody, an anti-EEAl antibody, and an anti-Rab5 antibody.
[0261] In embodiments, the detecting a level of phosphorylation includes detecting binding of the one or more anti-intracellular signaling molecule antibodies to the labeled ex vivo stimulated PBMC. In embodiments, the ex vivo stimulated PBMC is a permeabilized PBMC. In embodiments, the ex vivo stimulated PBMC is in a detection device.
[0262] In embodiments, the neurological disease treatment is a PLC-y2 pathway activator. In embodiments, the neurological disease treatment is galantamine, rivastigmine, donepezil, memantine, levodopa, carbidopa, amantadine, a dopamine agonist, a monoamine oxidase B inhibitor, a Catechol-O-methyltransferase inhibitor, or an anticholinergic. In embodiments, the neurological disease treatment is galantamine. In embodiments, the neurological disease treatment is rivastigmine. In embodiments, the neurological disease treatment is donepezil.
In embodiments, the neurological disease treatment is memantine. In embodiments, the neurological disease treatment is levodopa. In embodiments, the neurological disease treatment is carbidopa. In embodiments, the neurological disease treatment is amantadine. In embodiments, the neurological disease treatment is a dopamine agonist. In embodiments, the neurological disease treatment is a monoamine oxidase B inhibitor. In embodiments, the neurological disease treatment is a Catechol-O-methyltransferase inhibitor. In embodiments, the neurological disease treatment is an anticholinergic.
EXAMPLES
[0263] Provided are, inter alia, methods for measuring the propensity of a patient to be diagnosed with Alzheimer’s disease that include: a) conducting a measurement on a patient sample to determine frequency and functional signals of pPLC-gamma;2 responses from unstimulated and after LPS -stimulated in most peripheral blood mononuclear cells but especially in T cell subsets, B cells, NK cells, and monocytes, pSTATl responses in IFN- alpha; stimulated peripheral plasmablast, CD4+CD8+ T cells, and CD4-CD8+ T cells, and IL-6 stimulated basophils and CD8+ central memory T cells, and pSTAT5 responses in IFN- alpha; stimulated CD4+ activated T cells, CD4-CD8+ T cells, CD4+ activated T cells, and monocytes b) feeding the measurement to the developed multivariate machine learning model and statistical analyses to achieve diagnosis likelihood. The methods provided herein including embodiments thereof may be used to develop tools for diagnosis and early detection of Alzheimer’s disease. The methods provided herein including embodiments thereof may be also be used to select patients for drug development and clinical trial of an intervention in Alzheimer’s disease.
[0264] Applicants have built a novel data set that comprises a large sample set (for discovery of biomarkers) from well-matched participants with research quality annotation. Applicants have employed cutting-edge high-throughput mass cytometry technique to screen over four thousand cell signals. The selected biomarkers, with the developed models, can also be transferred to separate Parkinson’s patients from healthy cohort. They retain their capability to distinguish Alzheimer’s patients from Parkinson’s as well as dominantly inherited Alzheimer’s patients.
[0265] Peripheral blood mononuclear cells (PBMCs) might provide insight into the pathogenesis of Alzheimer’s disease (AD) or Parkinson’s disease (PD). We investigated samples from 132 well-characterized research participants using 7 canonical immune stimulants, mass cytometric identification of 35 PBMC subsets, and single-cell quantification of 15 intracellular signaling markers followed by machine learning model development to increase predictive power. From these, three main intracellular signaling pathways assayed were identified specifically in PBMC subsets from people with AD vs. match controls: reduced activation of PLCy2 across many cell types and stimulations, and more selectively variable activation of STAT1 and STAT5 depending on stimulant and cell type. Our findings functionally buttress the now multiple validated observations that a rare coding variant in PLCG2 is associated with a decreased risk of AD. Together, these data point to enhancing PLCy2 activity as a potential new therapeutic target for AD that has a readily accessible pharmacodynamic biomarker.
[0266] Introduction
[0267] Many laboratories have tested the hypothesis that peripheral blood mononuclear cells (PBMCs) can provide a window into the pathogenesis of neurodegenerative diseases because some subset of cells, such as T cell subsets or monocytes, might traffic into the brain and directly participate in disease mechanisms. Alternatively, it could be because of some inherited or acquired traits shared between PBMCs and brain cells might indirectly provide biomarkers of neuroinflammation (1, 2). Indeed, PubMed lists over 1200 citations for human leukocytes and Alzheimer’s disease (AD) and over 550 citations for human leukocytes and Parkinson’s disease (PD) (3-11). There has been some success in using this approach as quantitative traits in leukocytes to validate genetic risk loci for AD or PD (12, 13); however, none of these studies yet has led to an outcome that has been broadly reproduced across research laboratories. This could be due to diagnostic misclassification or lack of validation cohort or cell specific resolution. Because of this, none has transitioned to a blood-based clinical biomarker of AD or PD (14).
[0268] Although it is difficult to generalize across such a large number of studies, a few themes emerge. Most studies assembled PBMCs from relatively small groups of individuals with variable clinical characterization. Most used unstimulated PBMCs, which means that the leukocytes likely were in variable states due to site-specific processes of collection and enrichment. A few studies used ex vivo PBMC stimulants to investigate immune response, but the repertoire of stimulants has been limited, using only indiscriminate stimuli like PMA/ionomycin or non-canonical stimuli like amyloid beta or alpha-synuclein that activate through incompletely understood mechanisms (15). Most studies have made bulk measurements of PBMCs with relatively few using conventional flow cytometry to isolate specific subsets of PBMCs, and even those that did identified only a limited number of cell types (16). Of those that measured secreted factors, most measured only one or a small number of cellular or secreted molecules, and those that did measure a larger number of secreted factors were irreproducible (17, 18). Finally, only a few previous studies (14) have included multiple neurodegenerative diseases to control for features of being chronically ill.
[0269] To systematically test this hypothesis, we assembled discovery cohorts of research participants with sporadic (lacking monogenetic cause) AD dementia, sporadic PD, matched healthy controls, younger healthy controls. We also assembled separate validation cohorts of AD dementia and matched healthy controls. Importantly, the diagnosis of all participants was performed by a consensus panel of experts. We then broadly characterized the immune response following stimulation by an array of canonical activators using cytometry by time- of-flight (CyTOF), a proteomics technology that assesses the abundance of cell subsets, protein expression, and activation of signaling pathways with single-cell resolution (19-23). Using 7 different canonical stimulants (plus unstimulated), we assayed 15 different intracellular signaling responses in 35 different cell types within PBMCs (4200 total intracellular signaling responses) in more than 1.2 million individual cells per sample (except for a few samples) from each of the 132 individuals who had undergone extensive research quality clinical assessment. Machine learning algorithms and statistical analyses were employed to identify patterns of each diagnosis group from these extensive single cell data and examine the generalizability and predictive power of the findings.
[0270] Results
[0271] Single-cell profiling of the immune response in AD patients, PD patients, and healthy controls
[0272] The goal of our study was to determine which components of the peripheral immune response correlated with diagnoses of AD or PD. To achieve this goal, we performed deep single-cell profiling of PBMCs from our discovery cohort (AD, PD, HC-I, and HC-II; FIG.s 1A-1C) in response to a panel of ex vivo stimulants that activate through known cell signaling mechanisms. Specifically, HC-I refers to matched healthy controls for AD, and HC- Isub refers to a subset of HC-I selected as matched healthy controls for PD (Table 1). Given the results from our discovery work, we then further tested the developed model, without any retraining, on a separate validation cohort including AD patients (AD-V) and matched healthy controls (HC-V). [0273] Before investigating response to stimulants, we first assessed the relative abundance of different unstimulated PBMC subsets using a panel of cell surface markers to subtype the populations of monocytes, CD8+ T cells, CD4+ T cells, B cells, NK cells, DCs. Relative abundance was characterized by significance analysis of microarrays (24) with equal events sampled (minimal cluster size 1%, and false discovery rate 1%). We used CITRUS to compare PBMC subtype abundance for AD/HC-I, and for PD/HC-Isub· Unsupervised hierarchical clustering of age and sex-matched groups identified no significant differences among these well-established PBMC subtypes in paired comparison of AD or PD with its appropriate healthy controls. Although less well-matched for age and sex, we also compared and found significant difference between AD/PD, and present these exploratory data in FIG.s. 2A-2F.
[0274] Stimulant response was detected with additional probes directed at intracellular signaling molecules. Signals obtained from AD, PD, and HC-I PBMCs in unstimulated state or in response to IFN-a, IL-6, and IL-7 were strongly correlated to each other as indicated by proximity and the communities formed by these features in the correlation network (FIG.
3 A). Similarly, responses to IL-10, IL-21, LPS, and PMA also were strongly correlated with each other. To obtain a more meaningful description of each community, dimension reduction (UMAP) followed by unsupervised clustering (Umeans) segmented the network into 24 annotated communities (FIG. 3B). The annotation was based on PBMC subtype, stimulant, and/or intracellular signals that appeared most frequently in each community. Immune features with the same intracellular signals were likely to belong to the same community. In some communities, features could be further subdivided by cell type and stimulant.
[0275] iEN model developed from immune responses predicted AD diagnosis
[0276] Given the highly correlated immune features, we constructed a multivariate model using immunological Elastic Net (iEN), a recently developed regression algorithm designed for immune signals (25), and examined its predictive power on unseen data instead of using multiple univariate tests. In the discovery cohort, the cross-validated results indicated satisfactory classification of AD/HC-I (P = 1.03 c 103, AUC = 0.72; FIG.s 4A and 4B), but poor performance for PD/HC-Isub ( » 0.05). The final model developed from AD/HC-I classification was further tested using completely separate validation cohorts: HC-V and AD- V. The model showed its generalizability by achieving accurate AD-V/HC-V classification (P = 5.42 x 103, AUC = 0.84; FIG.s 4A and 4B). Note that one of the inputs to the iEN model was a list of immune features that represent literature-based canonical signaling pathways to prioritize during model optimization. Additionally, we have tried using signals that were experimentally present in this study (-10% discrepancy compared to canonical features; FIG.s 5 A and 5B), and it showed no significant effect on the model performance. Other conventional algorithms were also attempted but obtained less accuracy. Next, we proceeded to focus on the iEN model’s components developed from AD and HC-I.
[0277] Analyses of the model components revealed broader cellular differences between
Figure imgf000099_0001
[0278] The coefficients assigned to the immune features were components of the iEN model that could be used as a proxy to examine biological plausibility. These coefficients were displayed on a correlation network with red color indicating immune features that increased with the likelihood of carrying the AD diagnosis, and vice versa for blue (FIG. 4C). Piecewise regression analysis of the model revealed that only about 14 of the features with the highest coefficient magnitudes were necessary, and 111 components were needed to get comparable performance to using the entire set of immune features (4200 components; FIG. 4D). These top 14 and 111 features were depicted on the correlation networks (FIG. 4E and FIG. 6A). We next explored the four communities that contained the top 14 features, since features within the same community were highly correlated, and exhibited high univariate P value of the immune feature to diagnosis: community 18, community 12, community 17, and community 20 (FIG. 4C).
[0279] Community 18 comprised features with pPLCy2 intracellular signal. Visualizing the differences in pPLCy2 signal between the HC-I and the AD participants by PBMC subtypes and stimulant highlighted strong differences from diverse cell types (with the exception of most CD4+ T cells and DCs) in unstimulated and various stimulated conditions (FIG. 7A and FIG. 6C), where all of the responses tended to be lower in the AD group (FIG. 6B), particularly in NKT cells (FIG.s 7D and 7E). Among these are the most informative components of the iEN model, including pPLCy2 response in unstimulated NKT,
CD4+CD8+ T, and CD56bri§ht NK cells” (FIG.s 7D and 7E; FIG. 6C). These findings provide functional data that buttress the previous association that a variant in the gene that encodes PLCy2 lowers the risk of AD (26). [0280] Community 12, 17, and 20 were primarily a mix of pSTATl and pSTAT5 responses, which were highly correlated with each other. Examining the strength of these signal differences between HC-I and AD by cell type and stimulant using univariate analysis revealed that for both pSTATl and pSTAT5 intracellular responses, the strongest signals were mostly from IFN-a stimulated cells (FIG.s 7B and 7C). However, unlike pPLCy2, the trends of both intracellular responses were not monotonic across cell types and stimulations ; the responses tended to be higher in HC-I only in IFN-a stimulated cells or certain types of CD4+ T cells, while other conditions mostly led to an opposite response (FIG. 6B). All of the pSTATs features among the top 11 components of the iEN model with high univariate P value were IFN-a stimulated cells, including “pSTATl response of IFN-a stimulated plasmablast and CD4+CD8+ T cells” and “pSTAT5 response of IFN-a stimulated CD4+ TActivated” (FIG.s 7G and 7H; FIG.s 6D and 6D). These reflected differential JAK/STAT signaling after IFN-a stimulation between the two diagnostic groups. Additionally, other pSTATs signals that were expected, such as pSTATl and pSTAT5 from IL-7 stimulated CD4+ and CD8+ cells also were observed. On the other hand, those that were not expected, such as pSTAT signaling from LPS and PMA stimulated cells, were not observed (FIG.s 8A- 8G) (27, 28). These alignments to the existing signaling knowledge further supported the strength of our experimental approach.
Figure imgf000100_0001
[0282] The change in univariate P values depicted on the correlation network when separated by sex strongly suggested that the majority of the differences in pPLCy2 responses stemmed from male participants, whereas pSTATs were mostly contributed by female participants in the discovery cohorts (FIG.s 9A and 9B). The pPLCy2 response was very similar even if only AD patients were separated by sex, and compared to the entire HC-I (FIG.s 9C and 9D), suggesting sex specific immune responses in AD patients. Indeed, sex specific immune response are not uncommon and are being studied actively (28). Additionally, for participants with known APOE genotype, separating the AD patients into those with or without an APOE e4 allele and then comparing both with HC-I with no APOE s4, implied relevance of pPLCy2 in both APOE subgroups of AD. In contrast, the significance of pSTATs signals were reduced in both APOE subgroups. This was potentially due to fewer female participants in AD. The results were similar if the entire HC-I cohort was used regardless of APOE e4 status (results not shown). Others have shown repeatedly that macrophage and microglial innate immune responses have apoE isoform-dependent components in experimental settings (29-31); however, we are aware of only one report from a small number of participants that observed a modest apoE isoform-specific effect on human PBMC response to PMA stimulation (32). However, the number of observations in each of these subanalyses is limited and consequently more susceptible to potential unknown confounding factors.
[0283] Another possibility is that the pPLCy2 and pSTATs differences observed between AD and HC-I might be due to changes of aging that are accentuated in the AD group. However, correlation networks between HC-II/HC-I and HC-II/AD implied that, for both pairs, only a small number of signal differences were contributed by pSTATs and no significant contribution from pPLCy2 region (FIG.s 10A and 10B). Additionally, the specific pSTATs regions were different from those highlighted by HC-I/ AD comparison. These results further increase confidence that differential PBMC responses in pPLCy2 and some pSTATs are specific to AD.
Figure imgf000101_0001
[0285] The prediction accuracy of the developed iEN models from AD/HC-I could be used as a proxy to determine the similarities between the diagnostic groups. As expected, the developed models can perfectly classify the AD diagnostic group on which it was trained on (P = 2.59 x 10 13, AUC = 1; FIG.s 11A and 11B). Disease cross-prediction to PD, i.e. using AD/HC-I trained model to classify PD/HC-Isub participants, resulted in a satisfactory performance (P = 1.75 c 106, AUC = 0.88), although not as high as in the original domain. The correlation network of PD/HC-Isub diagnostic groups illustrated potential regions containing features associated with the developed iEN components from AD/HC-I (strong colors) that were also significantly different between PD and HC-Isub (sizable nodes) such as those in community 20 (FIG. 11C). The accurate PD/HC-Isub prediction may indicate co morbidity, shared risk factors, or shared mechanisms between the two neurodegenerative diseases. However, other communities whose features were important in the AD/HC-I iEN model, particularly communities 12, 17 (pSTATs region), and 18 (pPLCy2 region), did not overlap with signal differences in PD/HC-Isub. Hence, the model also can separate AD and PD patients (P = 3.99 c 103, AUC = 0.75), although the accuracy decreased when combined with AD-V predictions (P = 1.70 c 102, AUC = 0.70), suggesting that it did not merely classify between HC and those with an unspecified neurodegenerative disease. [0286] Discussion
[0287] Our work joins a range of previous works that have tested whether PBMCs might provide insight into the pathogenesis of neurodegenerative disease, either because some subset of PBMCs traffic into the brain and directly participate in disease mechanisms (1, 2), or because of inherited or acquired traits shared between some PBMCs and some brain- resident immune-competent cells. Our robust study design is distinguished by a large sample set from well-matched participants with research quality annotation, multiple canonical immune stimulants, flow cytometric identification of 35 PBMC subsets, single-cell quantification of 15 intracellular signaling markers, and inclusion of a neurodegenerative disease control in addition to matched HC, and validation of major findings with a completely independent cohort. Our data analysis is distinguished by the application of machine learning, instead of univariate testing, to demonstrate the accuracy and generalizability of the findings from this complex set of diagnostic phenotypes and single-cell quantitative molecular data. Mainly, three of the 15 intracellular signaling pathways assayed were differentially activated in PBMC subsets from people with AD compared to age and sex-matched HC: muted activation of PLCy2 across many cell types and stimulations, and more selective activation of STAT1 and STAT5 depending on stimulant and cell type. As far as we are aware, none of the previous studies that focused on PBMCs in AD have highlighted altered responses by PLCy2, STAT1, or STAT5.
[0288] Reduced pPLCy2 activation (community 18) was a strong feature of AD in all major subclasses of cells except dendritic cells and most CD4+ T cells, and under all stimulation conditions except IL-6 (FIG. 7A). Although pPLCy2 differential response was observed across most stimulants, it appeared strongest with LPS. pSTATl (community 20) and pSTAT5 (community 17) differential responses were more selective and largely restricted to IFN-a stimulation. pSTAT5 differential response was focused strongly in monocytes (FIG. 7C), while pSTATl differential response was weaker and more broadly observed in CD8+ T cells, B cells, and NK cells but not in monocytes (FIG. 7B). These differential immune responses have potential pathogenetic relevance to AD. Ab peptides directly and indirectly activate toll-like receptor (TLR) 4, the pattern recognition receptor for LPS (33, 34). STAT1 tissue concentration is increased in diseased regions of AD brain (35) and can regulate expression of beta secretase 1, one of the endoproteases that sequentially catalyzes the hydrolysis of amyloid precursor protein to generate Ab peptides (36). STAT5 activation regulates microglial activation and is required for monocyte-mediated synaptic degradation (37). Indeed, a recent pathway analysis that combined multiple GWAS data with clinical annotation nominated JAK-STAT signaling abnormalities as prominent contributors to AD etiology or pathogenesis (38). Finally, both T cells and monocytes can traffic from peripheral blood into brain, at least raising the possibility that the differential responses observed in these subsets of PBMCs might access brain, rather than simply being peripheral biomarkers of brain immune responses.
[0289] A rare variant in the gene that encodes pPLCy2 ( PCLG2 ; rs72824905, P522R) recently has been linked to decreased risk of AD (26); a finding that was recently validated in three other diverse cohorts (39-41). One of these validation studies also demonstrated that rs72824905 is associated with decreased risk of other forms of dementia but showed no risk modification for PD despite adequate sample size (40). None of these four genetic association studies evaluated the influence of sex or APOE genotype on PLCG2 risk modification for AD. Three groups have published results from human and mouse brain showing that expression of PLCG2 is limited to microglia among the major cell types in the brain, and that the protective P522R polymorphism modestly increases enzyme activity (39, 40, 42). These findings from brains of individuals with a rare variant of PLCG2 are consonant with our results, and suggest that reduced activation of PLCy2 in some T cell and monocyte subsets was a molecular feature of AD but not PD. Together, these data raise the possibility that increasing PLCy2 activity may be a new therapeutic target for AD and perhaps other forms of dementia. Moreover, our data suggest that a PBMC biomarker may be developed to aid in screening for therapeutics that enhance PLCy2 activity. However, other variants in PLCG2 have been associated with immune dysregulation (43), warranting caution in attempting to modulate its activity.
[0290] The role of microglia and the immune response in AD remains unclear but is an area of very active investigation highlighted by single cell approaches (44). Based on epidemiologic observations and results from transgenic mouse models, initial hypotheses focused on increased immune response in AD brain; however, recent observational and experimental data support a more nuanced view of both pro- and anti-inflammatory facets to AD progression (45). Our work fits well within this contemporary construct by revealing reduced and increased peripheral immune responses as characteristic of AD.
[0291] The iEN algorithm was unable to develop a predictive model for PD despite a similarly sized sample set as AD. Although disappointing, these results enhance our confidence in the predictive model for AD by controlling for non-specific features of age- related neurodegenerative disease, like reduced physical activity. Our disease cross prediction analysis showed that the AD/HC-I model much more weakly, but significantly, predicted PD/HC-Isub. There are several possible explanations for this outcome including partially shared genetic or environmental risk factors, or partially shared neurodegenerative mechanisms. The latter is supported by the now well-established observation that the neuropathological hallmark of PD (Lewy body disease) is present in about one-third to one- half of people diagnosed clinically with AD despite using the most rigorous research criteria. Alternatively, this disease cross-prediction may indicate partially shared alterations in the peripheral immune response between people with AD or PD. In this respect, it is interesting to note that disease cross-prediction of PD was driven largely by community 20, and not by community 18 (pPLCy2 region; FIG. 11C); consonant with the finding that the genetic variant in PLCG2 that lowers risk for AD does not modulate risk of PD (40).
[0292] Our extensive and unbiased investigation and robust analysis of the peripheral immune response strongly implicates reduced activation of PLCy2 as a molecular characteristic of AD but not PD or advancing age. Our experimental data from patient samples functionally buttress the now multiply validated observations that a rare coding variant in PLCG2 is associated with decreased risk of AD (40). Finally, our analyses suggest that PLCy2 activity is more commonly a feature of men with AD, and that it is not strongly influenced by APOE genotype or aging signals in PBMC. Together, these data point to enhancing PLCy2 activity as a potentially new therapeutic target for AD that has a readily accessible pharmacodynamic biomarker.
[0293] Materials and Methods
[0294] Study design
[0295] The aim of this study was to determine whether differences in peripheral immune responses between healthy participants and participants with neurodegenerative diseases are detectable from the CyTOF analysis of PBMCs. Participants were research volunteers at Stanford University in the Alzheimer’s Disease Research Center (ADRC) or the Pacific Udall Center (PUC). The work was approved by the IRB. Clinical diagnosis was made by consensus criteria (46, 47, 48), see Supplemental Materials for details. [0296] Blood was collected from volunteers after informed consent. We assembled a discovery cohort (n = 108) and a completely separate validation cohort (n = 24; summary statistics in Table 1, individual information in Tables 2 and 3). The discovery cohort consisted of four groups: AD, PD, and two different HC groups. The first healthy controls (HC-I) were older and were matched for AD with a subgroup of these people (HC-Isub; age between 63-80 for male and 67-73 for female) matched for PD. The second healthy controls (HC-II) were younger. A separate validation cohort also was assembled: AD-V and HC-V.
No validation cohort was assembled for PD due to no generalizable model identified from the discovery cohort. A separate technical control of PBMCs was prepared from a single healthy individual (70-year-old man) not included in the discovery or validation cohorts, and frozen into multiple aliquots and included in every CyTOF run.
[0297] Metal-tagged monoclonal antibodies
[0298] A panel of 37 metal -tagged monoclonal antibodies was used to probe PBMCs (Table 4). All pre-conjugated antibodies were purchased from Fluidigm. All other antibodies were purchased in carrier protein free PBS and conjugated in house with the respective metal isotope using the MaxPar antibody conjugation kit (Fluidigm). Metal labelled antibodies were diluted to 0.5 mg/ml in Candor PBS Antibody Stabilization solution (Candor Bioscience GmbH) for storage at 4 °C.
[0299] PBMC processing and stimulations
[0300] PBMCs were isolated from freshly drawn whole blood using density gradient centrifugation (Ficoll-Paque PLUS; GE Healthcare) in Sepmate tubes (49). The isolated and washed whole blood PBMCs were resuspended in 10% DMSO, 90% FBS and cryopreserved in liquid nitrogen. Frozen aliquots of PBMCs (batch of ~10 samples and an aliquot of technical control) were washed twice in RPMI at 37 °C. The samples were brought up in 1 ml RPMI and viability checked. One hundred pi of each sample were aliquoted and rested for 1 hr at 37 °C. PBMCs were then incubated in RPMI (unstimulated) or one of the seven specific stimulants, either a chemokine/cytokine (IFN-a, IL-6, IL-7, IL-10, IL-21), LPS, or PMA plus Ionomycin, for 15 min at 37 °C as shown in Table 5 and described previously (50). Following incubation under these eight conditions, PBMCs were fixed for 15 min with 4% PFA at room temperature, washed, permeabilized and barcoded exactly according to published methods to facilitate processing and minimize batch effects (51). [0301] All 8 stimulation conditions from a barcoded sample were pooled and incubated with titrated metal-labelled antibodies directed at cell surface markers designed to identify 35 immune cell subsets (52, 53). Cell types were identified by surface antibody signal for the following lineage markers: CD3, CD4, CD7, CD8, CDllb, CDllc, CD14, CD16, CD19, CD20, CD24, CD25, CD27, CD38, CD45RA, CD56, CD123, CD127, IgA, IgD and HLA- Dr. Pooled sample was then permeabilized with methanol and stored at -80 °C. Frozen samples were washed and incubated with metal-labelled antibodies directed at 15 intracellular signaling markers, followed by Ir-intercalator staining and resuspended in 0.1 EQ Four Element Calibration Beads (54). Intracellular signaling markers were pERKl/2, IkBa, NF-kB, p38, pAKT, pCREB, pLCK, pPLCy2, pS6, pSTATl, pSTAT3, pSTAT5, and endosomal proteins, Lamp2, EEA1, and Rab5 (55-57). Single cell data was acquired by CyTOF (Model: Helios, software version 6.5.358, Fluidigm, South San Francisco, CA) at 300 to 400 events per second. Initiation and tuning were performed according to manufacturer’s recommendation. Post-acquisition normalization and debarcoding was done with CyTOF software version 6.7.1014. Gating was performed using Flowjo-10 (FIG. 12) and Cytobank. More details on data processing and gating can be found in the Supplemental Materials.
[0302] Statistical Analysis
[0303] Application and evaluation of immunological Elastic Net (iEN) for multivariate modeling of mass cytometry data
[0304] The iEN model was employed as a multivariate model to examine generalizability and predictive power. The iEN added on to the commonly used EN algorithm the capability to incorporate knowledge of intracellular signal transduction on the generation of the mass cytometry data and was shown to yield superior performance (25). Briefly, the iEN algorithm optimized the coefficient (b) for each associated feature by minimizing the cost function:
Figure imgf000106_0001
Here, X is a matrix of size m x n, where m is the number of samples and n is the number of all immune features (intracellular signals from cells under different stimulating conditions), Y is a vector of ground truths (diagnostic groups) with length n, b is a vector of the model’s coefficients with length n, l and a together control the magnitude of the model regularization. Lastly, F is a diagonal matrix of size n x n containing a prioritization value of 1 for elements associated with canonical immune feature or e-ip for other lower priority (non- canonical) features. The list of canonical signals that were prioritized was tabulated in Table 6
[0305] To objectively determine the value of the hyperparameters as well as the generalizability of the iEN model from the discovery cohort, 250 iterations of a two-layered cross validation scheme were employed. In each iteration, the outer layer randomly held out one-third of the samples for performance evaluation on unseen data, and the inner layer used the rest of the samples for parameter optimization (grid search of l, a, and cp). At the completion of the 250 iterations, the mean values of the predictions were used for model evaluation by Wilcoxon rank-sum test P value and area under receiver operating characteristic curve (AUC), and the weights and cp determined from all iterations were averaged to obtain a final model. The final model was used for feature component analyses, prediction of validation cohort, and disease cross-prediction.
[0306] Supplementary methods
[0307] 1. Clinical diagnosis
[0308] A consensus panel consisting of one board-certified movement disorders neurologist or behavioral neurologist, one board-certified neuropsychologist, and other study personnel adjudicated the diagnosis for each participant. PD diagnosis was based on UK PD Society Brain Bank clinical diagnostic criteria (46), as previously reported (47). AD included patients with dementia likely due to AD pathology based on the NIH Alzheimer’s Disease Diagnostic Guidelines (48). Healthy controls were determined to have no neurodegenerative disease or concerning cognitive decline by history, a normal neurological exam, and cognitive test scores within 1.5 standard deviations from normative values.
[0309] 2. Data Processing
[0310] The fcs files acquired on the CyTOF (HELIOS, software version 6.5.358 by Fluidigm, S. San Francisco) was bead normalized using Fluidigm software. The normalized files were then debarcoded using Fluidigm software and the unassigned events was removed. The debarcoded fcs files was gated on Flow-jo Version 10.4.1 to remove beads, doublets and then gated to DNA+ events. The data was then transformed and percentile normalized. The percentile normalized fcs files were then gated by Flow-jo to characterize a broad range of cell types and signaling (FIG. 12). The median values of the gated signals were used for any further analysis.
[0311] Six samples that resulted in less than 45% DNA hi of total cells in any kind of stimulation condition were excluded from the study. Missing values after gating (-0.1% of the signals in the discovery cohorts) were due to insufficient cell counts, and were addressed by mean imputation. A batch correction method, ComBat (58), was applied to reduce the staining batch effect. The batch effect was eliminated as measured by principal variance component analysis (59).
[0312] 3. Model reduction [0313] A one-hundred iterations bootstrapping procedure with replacement, each with a subset of patients equal to the size of the full dataset, was employed to identify the number of the most significant components of the iEN model. A piecewise regression analysis (60) was then performed on the median P value of the model from all iterations as a function of number of features to identify the number of features. [0314] 4. Correlation networks
[0315] The layout of the correlation network was obtained from applying dimension reduction algorithm, t-SNE (61), on the Spearman correlation matrix of all available immune features. The edge of the graph represents those with Spearman’s P value < 0.05 after Bonferroni adjustment. For visualization of the group of highly correlated immune features, the communities were identified by using uniform manifold approximation and projection (UMAP) to reduce the dimension to 10% of the original immune features (62), followed by unsupervised clustering using A-means algorithm. The optimized number of clusters (24 clusters) was determined based on optimized C-index and Baker-Hubert gamma index.
TABLES
[0316] Table 1. Summary of participants in the discovery («=108) and validation cohorts («=24) diagnosed with Alzheimer’s disease (AD), Parkinson’s disease (PD), or healthy controls (HC).
Figure imgf000109_0001
[0317] Table 2. Demographics (age, sex, and counts of APOE e4 gene) of participants in discovery cohort by diagnostic groups.
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
[0318] Table 3. Demographics (age, sex, and counts of APOE e4 gene) of participants in validation cohort by diagnostic groups.
Figure imgf000112_0002
Figure imgf000113_0001
[0319] Table 4. Target, clone, conjugation information, and product identifier.
Figure imgf000113_0002
Il l
Figure imgf000114_0001
Figure imgf000115_0001
[0320] Table 5. Stimulants used to activate PBMCs
Figure imgf000115_0002
PATENT
Attorney Docket No. 041243-554001WO Client Ref No. 19-495
[0321] Table 6. Prioritization matrix used for iEN model (1 indicated canonical response and vice versa for 0).
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
PATENT
Attorney Docket No. 041243-554001WO Client Ref. No. 19-495
[0322] Table 7. Exemplary embodiments of intracellular signaling molecules, stimulatory agents and PBMCs contemplated for the methods provided herein including embodiments thereof.
Figure imgf000129_0001
Figure imgf000129_0002
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000135_0002
Figure imgf000136_0001
Figure imgf000137_0001
CeS fes^yes¾c¥ from ali cohorts jseparatifto ¾PD1
2. CS4' sells
3. iS' T^ceSs
4 GOSS* CDtS4 ' creseytes
REFERENCES
[0323] 1. O. Butovsky, M. Koronyo-Hamaoui, G. Kunis, E. Ophir, G. Landa, H. Cohen,
M. Schwartz, Glatiramer acetate fights against Alzheimer’s disease by inducing dendritic-like microglia expressing insulin-like growth factor 1, Proc. Natl. Acad. Sci. U.S.A. 103, 11784— 11789 (2006).
[0324] 2. M. Fiala, Q. N. Liu, J. Sayre, V. Pop, V. Brahmandam, M. C. Graves, H. V.
Vinters, Cyclooxygenase-2-positive macrophages infiltrate the Alzheimer’s disease brain and damage the blood-brain barrier, Pur. J. Clin. Invest. 32, 360-371 (2002).
[0325] 3. T. J. Lewis, C. L. Trempe, The End of Alzheimer’s: The Brain and Beyond
(Academic Press, Cambridge, MA, 2017).
[0326] 4. R. Pemeczky, Biomarkers for Alzheimer ’s Disease Drug Development
(Humana Press, Totowa, NJ, 2018).
[0327] 5. K. Rezai-Zadeh, D. Gate, C. A. Szekely, T. Town, Can peripheral leukocytes be used as Alzheimer’s disease biomarkers?, Expert Rev. Neurother. 9, 1623-1633 (2009).
[0328] 6. R. E. Akins, K. G. Robinson, Biomarker blood tests for cerebral palsy,
Cerebral Palsy, 1-8 (2019).
[0329] 7. G. Schmitz, K. Leuthauser-Jaschinski, E. Orso, Are circulating monocytes as microglia orthologues appropriate biomarker targets for neuronal diseases? (Supplementry Table), Central Nervous System Agents in Medicinal Chemistry 9, 307-330 (2009).
[0330] 8. M. M. Gonzalez, Atlas of Biomarkers for Alzheimer ’s Disease (Springer, New
York, 2014).
[0331] 9. C. Henchcbffe, Blood and cerebrospinal fluid markers in Parkinson’s disease: current biomarker findings, Current Biomarker Findings 5, 1-11 (2014).
[0332] 10. S. Lehmann, C. E. Teunissen, Editorial: Biomarkers of Alzheimer’s disease:
The Present and the future, Front. Neurol. 7 (2016).
[0333] 11. K.-L. Huang, E. Marcora, A. A. Pimenova, A. F. Di Narzo, M. Kapoor, S. C.
Jin, O. Harari, S. Bertelsen, B. P. Fairfax, J. Czajkowski, V. Chouraki, B. Grenier-Boley, C. Bellenguez, Y. Deming, A. McKenzie, T. Raj, A. E. Renton, J. Budde, A. Smith, A. Fitzpatrick, J. C. Bis, A. DeStefano, H. H. H. Adams, M. Arfan Ikram, S. van der Lee, J. L. Del-Aguila, M. V. Fernandez, L. Ibanez, R. Sims, V. Escott-Price, R. Mayeux, J. L. Haines, L. A. Farrer, M. A. Pericak-Vance, J. C. Lambert, C. van Duijn, L. Launer, S. Seshadri, J. Williams, P. Amouyel, G. D. Schellenberg, B. Zhang, I. Borecki, J. S. K. Kauwe, C. Cruchaga, K. Hao, A. M. Goate, The International Genomics of Alzheimer’s Project, The Alzheimer’s Disease Neuroimaging Initiative, A common haplotype lowers PU.l expression in myeloid cells and delays onset of Alzheimer’s disease, Nat. Neurosci. 20, 1052-1061 (2017).
[0334] 12. T. Raj, K. Rothamel, S. Mostafavi, C. Ye, M. N. Lee, J. M. Replogle, T. Feng,
M. Lee, N. Asinovski, I. Frohlich, S. Imboywa, A. Von Korff, Y. Okada, N. A. Patsopoulos, S. Davis, C. McCabe, H.-I. Paik, G. P. Srivastava, S. Raychaudhuri, D. A. Hafler, D. Roller, A. Regev, N. Hacohen, D. Mathis, C. Benoist, B. E. Stranger, P. L. De Jager, Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes, Science 344, 519-523 (2014).
[0335] 13. K. J. Ryan, C. C. White, K. Patel, J. Xu, M. Olah, J. M. Replogle, M.
Frangieh, M. Cimpean, P. Winn, A. McHenry, B. J. Kaskow, G. Chan, N. Cuerdon, D. A. Bennett, J. D. Boyd, J. Imitola, W. Elyaman, P. L. De Jager, E. M. Bradshaw, A human microglia-like cellular model for assessing the effects of neurodegenerative disease gene variants, Sci. Transl. Med. 9 (2017).
[0336] 14. E. Delvaux, D. Mastroeni, J. Nolz, N. Chow, M. Sabbagh, R. J. Caselli, E. M.
Reiman, F. J. Marshall, P. D. Coleman, Multivariate analyses of peripheral blood leukocyte transcripts distinguish Alzheimer’s, Parkinson's, control, and those at risk for developing Alzheimer's, Neurobiol. Aging 58, 225-237 (2017).
[0337] 15. G. Schmidbauer, W. W. Hancock, B. Wasowska, A. M. Badger, J. W. Kupiec-
Weglinski, Abrogation by rapamycin of accelerated rejection in sensitized rats by inhibition of alloantibody responses and selective suppression of intragraft mononuclear and endothelial cell activation, cytokine production, and cell adhesion, Transplantation 57, 933-941 (1994).
[0338] 16. R. Davies, P. Vogelsang, R. Jonsson, S. Appel, An optimized multiplex flow cytometry protocol for the analysis of intracellular signaling in peripheral blood mononuclear cells, J. Immunol. Methods 436, 58-63 (2016).
[0339] 17. S. Ray, M. Britschgi, C. Herbert, Y. Takeda-Uchimura, A. Boxer, K.
Blennow, L. F. Friedman, D. R. Galasko, M. Jutel, A. Karydas, J. A. Kaye, J. Leszek, B. L. Miller, L. Minthon, J. F. Quinn, G. D. Rabinovici, W. H. Robinson, M. N. Sabbagh, Y. T. So, D. L. Sparks, M. Tabaton, J. Tinklenberg, J. A. Yesavage, R. Tibshirani, T. Wyss-Coray, Classification and prediction of clinical Alzheimer’s diagnosis based on plasma signaling proteins, Nat. Med. 13, 1359-1362 (2007).
[0340] 18. H. D. Soares, Y. Chen, M. Sabbagh, A. Rohrer, E. Schrijvers, M. Breteler,
Identifying early markers of Alzheimer’s disease using quantitative multiplex proteomic immunoassay panels, Ann. NY Acad. Sci. 1180, 56-67 (2009).
[0341] 19. A. Cosma, G. Nolan, B. Gaudilliere, Mass cytometry: The time to settle down,
Cytom. Part A 91, 12-13 (2017).
[0342] 20. Q. Baca, A. Cosma, G. Nolan, B. Gaudilliere, The road ahead: Implementing mass cytometry in clinical studies, one cell at a time, Cytom. Part B-Clin. Cy. 92, 10-11 (2017).
[0343] 21. P. Kvistborg, C. Gouttefangeas, N. Aghaeepour, A. Cazaly, P. K.
Chattopadhyay, C. Chan, J. Eckl, G. Finak, S. R. Hadrup, H. T. Maecker, D. Maurer, T. Mosmann, P. Qiu, R. H. Scheuermann, M. J. P. Welters, G. Ferrari, R. R. Brinkman, C. M. Britten, Thinking outside the gate: single-cell assessments in multiple dimensions, Immunity 42, 591-592 (2015).
[0344] 22. S. C. Bendall, G. P. Nolan, M. Roederer, P. K. Chattopadhyay, A deep profiler’s guide to cytometry, Trends Immunol. 33, 323-332 (2012).
[0345] 23. N. Aghaeepour, P. Chattopadhyay, M. Chikina, T. Dhaene, S. Van Gassen, M.
Kursa, B. N. Lambrecht, M. Malek, G. J. McLachlan, Y. Qian, P. Qiu, Y. Saeys, R. Stanton, D. Tong, C. Vens, S. Walkowiak, K. Wang, G. Finak, R. Gottardo, T. Mosmann, G. P.
Nolan, R. H. Scheuermann, R. R. Brinkman, A benchmark for evaluation of algorithms for identification of cellular correlates of clinical outcomes, Cytom. Part A 89, 16-21 (2016).
[0346] 24. V. G. Tusher, R. Tibshirani, G. Chu, Significance analysis of microarrays applied to the ionizing radiation response, Proc. Natl. Acad. Sci. U.S.A. 98, 5116-5121 (2001).
[0347] 25. N. Aghaeepour, E. A. Ganio, D. Mcilwain, A. S. Tsai, M. Tingle, S. Van
Gassen, D. K. Gaudilliere, Q. Baca, L. McNeil, R. Okada, M. S. Ghaemi, D. Furman, R. J. Wong, V. D. Winn, M. L. Druzin, Y. Y. El-Sayed, C. Quaintance, R. Gibbs, G. L. Darmstadt, G. M. Shaw, D. K. Stevenson, R. Tibshirani, G. P. Nolan, D. B. Lewis, M. S. Angst, B. Gaudilliere, An immune clock of human pregnancy, Sci. Immunol. 2 (2017).
[0348] 26. R. Sims, S. J. van der Lee, A. C. Naj, C. Bellenguez, N. Badarinarayan, J.
Jakobsdottir, B. W. Kunkle, A. Boland, R. Raybould, J. C. Bis, E. R. Martin, B. Grenier- Boley, S. Heilmann-Heimbach, V. Chouraki, A. B. Kuzma, K. Sleegers, M. Vronskaya, A. Ruiz, R. R. Graham, R. Olaso, P. Hoffmann, M. L. Grove, B. N. Vardarajan, M. Hiltunen, M. M. Nothen, C. C. White, K. L. Hamilton-Nelson, J. Epelbaum, W. Maier, S.-H. Choi, G. W. Beecham, C. Dulary, S. Herms, A. V. Smith, C. C. Funk, C. Derbois, A. J. Forstner, S. Ahmad, H. Li, D. Bacq, D. Harold, C. L. Satizabal, O. Valladares, A. Squassina, R. Thomas, J. A. Brody, L. Qu, P. Sanchez-Juan, T. Morgan, F. J. Wolters, Y. Zhao, F. S. Garcia, N. Denning, M. Fomage, J. Malamon, M. C. D. Naranjo, E. Majounie, T. H. Mosley, B. Dombroski, D. Wallon, M. K. Lupton, J. Dupuis, P. Whitehead, L. Fratiglioni, C. Medway, X. Jian, S. Mukheqee, L. Keller, K. Brown, H. Lin, L. B. Cantwell, F. Panza, B.
McGuinness, S. Moreno-Grau, J. D. Burgess, V. Solfrizzi, P. Proitsi, H. H. Adams, M. Allen, D. Seripa, P. Pastor, L. A. Cupples, N. D. Price, D. Hannequin, A. Frank-Garcia, D. Levy, P. Chakrabarty, P. Caffarra, I. Giegling, A. S. Beiser, V. Giedraitis, H. Hampel, M. E. Garcia,
X. Wang, L. Lannfelt, P. Mecocci, G. Eiriksdottir, P. K. Crane, F. Pasquier, V. Boccardi, I. Henandez, R. C. Barber, M. Scherer, L. Tarraga, P. M. Adams, M. Leber, Y. Chen, M. S. Albert, S. Riedel-Heller, V. Emilsson, D. Beekly, A. Braae, R. Schmidt, D. Blacker, C. Masullo, H. Schmidt, R. S. Doody, G. Spalletta, W. T. Longstreth Jr, T. J. Fairchild, P.
Bossu, O. L. Lopez, M. P. Frosch, E. Sacchinelli, B. Ghetti, Q. Yang, R. M. Huebinger, F. Jessen, S. Li, M. I. Kamboh, J. Morris, O. Sotolongo-Grau, M. J. Katz, C. Corcoran, M. Dunstan, A. Braddel, C. Thomas, A. Meggy, R. Marshall, A. Gerrish, J. Chapman, M. Aguilar, S. Taylor, M. Hill, M. D. Fairen, A. Hodges, B. Vellas, H. Soininen, I. Kloszewska, M. Daniilidou, J. Uphill, Y. Patel, J. T. Hughes, J. Lord, J. Turton, A. M. Hartmann, R. Cecchetti, C. Fenoglio, M. Serpente, M. Arcaro, C. Caltagirone, M. D. Orfei, A. Ciaramella, S. Pichler, M. Mayhaus, W. Gu, A. Lleo, J. Fortea, R. Blesa, I. S. Barber, K. Brookes, C. Cupidi, R. G. Maletta, D. Carrell, S. Sorbi, S. Moebus, M. Urbano, A. Pilotto, J. Komhuber, P. Bosco, S. Todd, D. Craig, J. Johnston, M. Gill, B. Lawlor, A. Lynch, N. C. Fox, J. Hardy, ARUK Consortium, R. L. Albin, L. G. Apostolova, S. E. Arnold, S. Asthana, C. S. Atwood, C. T. Baldwin, L. L. Barnes, S. Barral, T. G. Beach, J. T. Becker, E. H. Bigio, T. D. Bird, B. F. Boeve, J. D. Bowen, A. Boxer, J. R. Burke, J. M. Bums, J. D. Buxbaum, N. J. Cairns, C. Cao, C. S. Carlson, C. M. Carlsson, R. M. Camey, M. M. Carrasquillo, S. L. Carroll, C. C. Diaz, H. C. Chui, D. G. Clark, D. H. Cribbs, E. A. Crocco, C. DeCarli, M. Dick, R. Duara, D.
A. Evans, K. M. Faber, K. B. Fallon, D. W. Fardo, M. R. Farlow, S. Ferris, T. M. Foroud, D. R. Galasko, M. Gearing, D. H. Geschwind, J. R. Gilbert, N. R. Graff-Radford, R. C. Green, J. H. Growdon, R. L. Hamilton, L. E. Harrell, L. S. Honig, M. J. Huentelman, C. M. Hulette, B. T. Hyman, G. P. Jarvik, E. Abner, L.-W. Jin, G. Jun, A. Karydas, J. A. Kaye, R. Kim, N. W. Kowall, J. H. Kramer, F. M. LaFerla, J. J. Lah, J. B. Leverenz, A. I. Levey, G. Li, A. P. Lieberman, K. L. Lunetta, C. G. Lyketsos, D. C. Marson, F. Martiniuk, D. C. Mash, E. Masliah, W. C. McCormick, S. M. McCurry, A. N. McDavid, A. C. McKee, M. Mesulam, B.
L. Miller, C. A. Miller, J. W. Miller, J. C. Morris, J. R. Murrell, A. J. Myers, S. O’Bryant, J.
M. Olichney, V. S. Pankratz, J. E. Parisi, H. L. Paulson, W. Perry, E. Peskind, A. Pierce, W. W. Poon, H. Potter, J. F. Quinn, A. Raj, M. Raskind, B. Reisberg, C. Reitz, J. M. Ringman,
E. D. Roberson, E. Rogaeva, H. J. Rosen, R. N. Rosenberg, M. A. Sager, A. J. Saykin, J. A. Schneider, L. S. Schneider, W. W. Seeley, A. G. Smith, J. A. Sonnen, S. Spina, R. A. Stem,
R. H. Swerdlow, R. E. Tanzi, T. A. Thomton-Wells, J. Q. Trojanowski, J. C. Troncoso, V. M. Van Deerlin, L. J. Van Eldik, H. V. Vinters, J. P. Vonsattel, S. Weintraub, K. A. Welsh- Bohmer, K. C. Wilhelmsen, J. Williamson, T. S. Wingo, R. L. Woltjer, C. B. Wright, C.-E. Yu, L. Yu, F. Garzia, F. Golamaully, G. Septier, S. Engelborghs, R. Vandenberghe, P. P. De Deyn, C. M. Femadez, Y. A. Benito, H. Thonberg, C. Forsell, L. Lilius, A. Kinhult- Stahlbom, L. Kilander, R. Brundin, L. Concari, S. Helisalmi, A. M. Koivisto, A. Haapasalo, V. Dermecourt, N. Fievet, O. Hanon, C. Dufouil, A. Brice, K. Ritchie, B. Dubois, J. J.
Himali, C. D. Keene, J. Tschanz, A. L. Fitzpatrick, W. A. Kukull, M. Norton, T. Aspelund, E.
B. Larson, R. Munger, J. I. Rotter, R. B. Lipton, M. J. Bullido, A. Hofman, T. J. Montine, E. Coto, E. Boerwinkle, R. C. Petersen, V. Alvarez, F. Rivadeneira, E. M. Reiman, M. Gallo, C. J. O’Donnell, J. S. Reisch, A. C. Bruni, D. R. Royall, M. Dichgans, M. Sano, D. Galimberti, P. St George-Hyslop, E. Scarpini, D. W. Tsuang, M. Mancuso, U. Bonuccelli, A. R.
Winslow, A. Daniele, C.-K Wu, GERAD/PERADES, CHARGE, ADGC, EADI, O. Peters, B. Nacmias, M. Riemenschneider, R. Heun, C. Brayne, D. C. Rubinsztein, J. Bras, R. Guerreiro, A. Al-Chalabi, C. E. Shaw, J. Collinge, D. Mann, M. Tsolaki, J. Clarimon, R. Sussams, S. Lovestone, M. C. O’Donovan, M. J. Owen, T. W. Behrens, S. Mead, A. M. Goate, A. G. Uitterlinden, C. Holmes, C. Cruchaga, M. Ingelsson, D. A. Bennett, J. Powell,
T. E. Golde, C. Graff, P. L. De Jager, K. Morgan, N. Ertekin-Taner, O. Combarros, B. M. Psaty, P. Passmore, S. G. Younkin, C. Berr, V. Gudnason, D. Rujescu, D. W. Dickson, J.-F. Dartigues, A. L. DeStefano, S. Ortega-Cubero, H. Hakonarson, D. Campion, M. Boada, J. K. Kauwe, L. A. Farrer, C. Van Broeckhoven, M. A. Ikram, L. Jones, J. L. Haines, C. Tzourio,
L. J. Launer, V. Escott-Price, R. Mayeux, J.-F. Deleuze, N. Amin, P. A. Holmans, M. A. Pericak-Vance, P. Amouyel, C. M. van Duijn, A. Ramirez, L.-S. Wang, J.-C. Lambert, S. Seshadri, J. Williams, G. D. Schellenberg, Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease, Nat. Genet. 49, 1373-1384 (2017).
[0349] 27. C. Le Saout, M. A. Luckey, A. V. Villarino, M. Smith, R. B. Hasley, T. G.
Myers, H. Imamichi, J.-H. Park, J. J. O’Shea, H. C. Lane, M. Catalfamo, IL-7-dependent STAT1 activation limits homeostatic CD4+ T cell expansion, JCI Insight 2 (2017).
[0350] 28. G. K. Fragiadakis, Z. B. Bjomson-Hooper, D. Madhireddy, K. Sachs, M. H.
Spitzer, S. C. Bendall, G. P. Nolan, Variation of immune cell responses in humans reveals sex-specific coordinated signaling across cell types, bioRxiv, 567784 (2019).
[0351] 29. J. R. Lynch, W. Tang, H. Wang, M. P. Vitek, E. R. Bennett, P. M. Sullivan, D.
S. Warner, D. T. Laskowitz, APOE genotype and an ApoE-mimetic peptide modify the systemic and central nervous system inflammatory response, J. Biol. Chem. 278, 48529- 48533 (2003).
[0352] 30. M. P. Vitek, C. M. Brown, C. A. Colton, APOE genotype-specific differences in the innate immune response, Neurobiol Aging 30, 1350-1360 (2009).
[0353] 31. R. W. Mahley, S. C. Rail Jr, Apolipoprotein E: far more than a lipid transport protein, Annu. Rev. Genomics Hum. Genet. 1, 507-537 (2000).
[0354] 32. P. Olgiati, A. Politis, P. Malitas, D. Albani, S. Dusi, L. Polito, S. De Mauro,
A. Zisaki, C. Piperi, E. Stamouli, A. Mailis, S. Batelli, G. Forloni, D. De Ronchi, A. Kalofoutis, I. Liappas, A. Serretti, APOE epsilon-4 allele and cytokine production in Alzheimer’s disease, Int. J. Geriatr. Psych. 25, 338-344 (2010).
[0355] 33. C. R. Stewart, L. M. Stuart, K. Wilkinson, J. M. van Gils, J. Deng, A. Halle,
K. J. Rayner, L. Boyer, R. Zhong, W. A. Frazier, A. Lacy-Hulbert, J. El Khoury, D. T. Golenbock, K. J. Moore, CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer, Nat. Immunol. 11, 155-161 (2010).
[0356] 34. K. Fujita, K. Motoki, K. Tagawa, X. Chen, H. Hama, K. Nakajima, H.
Homma, T. Tamura, H. Watanabe, M. Katsuno, C. Matsumi, M. Kajikawa, T. Saito, T. Saido, G. Sobue, A. Miyawaki, H. Okazawa, HMGB1, a pathogenic molecule that induces neurite degeneration via TLR4-MARCKS, is a potential therapeutic target for Alzheimer’s disease, Sci. Rep. 6, 31895 (2016).
[0357] 35. Y. Kitamura, S. Shimohama, T. Ota, Y. Matsuoka, Y. Nomura, T. Taniguchi,
Alteration of transcription factors NF-kappaB and STAT1 in Alzheimer’s disease brains, Neurosci. Lett. 237, 17-20 (1997).
[0358] 36. W. J. Lee, S. A. Ham, G. H. Lee, M. Choi, H. Yoo, K. S. Paek, D. Lim, K.
Hong, J. S. Hwang, H. G. Seo, Activation of peroxisome proliferator-activated receptor delta suppresses BACE 1 expression by up-regulating SOCS 1 in a JAK2/ STAT1 -dependent manner, J. Neurochem. 151, 370-385 (2019).
[0359] 37. G. Di Liberto, S. Pantelyushin, M. Kreutzfeldt, N. Page, S. Musardo, R. Coras,
K. Steinbach, I. Vincenti, B. Klimek, T. Lingner, G. Salinas, N. Lin-Marq, O. Staszewski, M. J. Costa Jordao, I. Wagner, K. Egervari, M. Mack, C. Bellone, I. Bliimcke, M. Prinz, D. D. Pinschewer, D. Merkler, Neurons under T cell attack coordinate phagocyte-mediated synaptic stripping, Cell 175, 458-471 (2018).
[0360] 38. A. J. Nevado-Holgado, E. Ribe, L. Thei, L. Furlong, M.-A. Mayer, J. Quan, J.
C. Richardson, J. Cavanagh, N. Consortium, S. Lovestone, Genetic and real-world clinical data, combined with empirical validation, nominate JAK-STAT Signaling as a target for Alzheimer’s disease therapeutic development, Cells 8, 425 (2019).
[0361] 39. O. J. Conway, M. M. Carrasquillo, X. Wang, J. M. Bredenberg, J. S. Reddy, S.
L. Strickland, C. S. Younkin, J. D. Burgess, M. Allen, S. J. Lincoln, T. Nguyen, K. G. Malphrus, A. I. Soto, R. L. Walton, B. F. Boeve, R. C. Petersen, J. A. Lucas, T. J. Ferman,
W. P. Cheshire, J. A. van Gerpen, R. J. Uitti, Z. K. Wszolek, O. A. Ross, D. W. Dickson, N. R. Graff-Radford, N. Ertekin-Taner, ABI3 and PLCG2 missense variants as risk factors for neurodegenerative diseases in Caucasians and African Americans, Mol. Neurodegener. 13, 53 (2018).
[0362] 40. S. J. van der Lee, O. J. Conway, I. Jansen, M. M. Carrasquillo, L. Kleineidam,
E. van den Akker, I. Hernandez, K. R. van Eijk, N. Stringa, J. A. Chen, A. Zettergren, T. F.
M. Andlauer, M. Diez-Fairen, J. Simon-Sanchez, A. Lleo, H. Zetterberg, M. Nygaard, C. Blauwendraat, J. E. Savage, J. Mengel-From, S. Moreno-Grau, M. Wagner, J. Fortea, M. J. Keogh, K. Blennow, I. Skoog, M. A. Friese, O. Pletnikova, M. Zulaica, C. Lage, I. de Rojas, S. Riedel-Heller, I. Illan-Gala, W. Wei, B. Jeune, A. Orellana, F. Then Bergh, X. Wang, M. Hulsman, N. Beker, N. Tesi, C. M. Morris, B. Indakoetxea, L. E. Collij, M. Scherer, E. Morenas-Rodriguez, J. W. Ironside, B. N. M. van Berckel, D. Alcolea, H. Wiendl, S. L. Strickland, P. Pastor, E. Rodriguez Rodriguez, DESGESCO (Dementia Genetics Spanish Consortium), EADB (Alzheimer Disease European DNA biobank), EADB (Alzheimer Disease European DNA biobank), IFGC (International FTD-Genomics Consortium), IPDGC (The International Parkinson Disease Genomics Consortium), IPDGC (The International Parkinson Disease Genomics Consortium), RiMod-FTD (Risk and Modifying factors in Fronto-Temporal Dementia), Netherlands Brain Bank (NBB), B. F. Boeve, R. C. Petersen, T. J. Ferman, J. A. van Gerpen, M. J. T. Reinders, R. J. Uitti, L. Tarraga, W. Maier, O. Dols- Icardo, A. Kawalia, M. C. Dalmasso, M. Boada, U. K. Zettl, N. M. van Schoor, M. Beekman, M. Allen, E. Masliah, A. L. de Munain, A. Pantelyat, Z. K. Wszolek, O. A. Ross, D. W. Dickson, N. R. Graff-Radford, D. Knopman, R. Rademakers, A. W. Lemstra, Y. A. L. Pijnenburg, P. Scheltens, T. Gasser, P. F. Chinnery, B. Hemmer, M. A. Huisman, J.
Troncoso, F. Moreno, E. A. Nohr, T. I. A. Sorensen, P. Heutink, P. Sanchez- Juan, D. Posthuma, GIFT (Genetic Investigation in Frontotemporal Dementia and Alzheimer’s Disease) Study Group, J. Clarimon, K. Christensen, N. Ertekin-Taner, S. W. Scholz, A. Ramirez, A. Ruiz, E. Slagboom, W. M. van der Flier, H. Holstege, A nonsynonymous mutation in PLCG2 reduces the risk of Alzheimer’s disease, dementia with Lewy bodies and frontotemporal dementia, and increases the likelihood of longevity, Acta Neuropathol. 138, 237-250 (2019).
[0363] 41. M. C. Dalmasso, L. I. Brusco, N. Olivar, C. Muchnik, C. Hanses, E. Milz, J.
Becker, S. Heilmann-Heimbach, P. Hoffmann, F. A. Prestia, P. Galeano, M. S. S. Avalos, L. E. Martinez, M. E. Carulla, P. J. Azurmendi, C. Liberczuk, C. Fezza, M. Sampano, M. Fierens, G. Jemar, P. Solis, N. Medel, J. Lisso, Z. Sevillano, P. Bosco, P. Bossu, G. Spalletta, D. Galimberti, M. Mancuso, B. Nacmias, S. Sorbi, P. Mecocci, A. Pilotto, P. Caffarra, F. Panza, M. Bullido, J. Clarimon, P. Sanchez-Juan, E. Coto, F. Sanchez-Garcia, C. Graff, M. Ingelsson, C. Bellenguez, E. M. Castano, C. Kairiyama, D. G. Politis, S. Kochen, H. Scaro, W. Maier, F. Jessen, C. A. Mangone, J.-C. Lambert, L. Morelli, A. Ramirez, Transethnic meta-analysis of rare coding variants in PLCG2, ABI3, and TREM2 supports their general contribution to Alzheimer’s disease, Transl. Psychiatry 9, 55 (2019).
[0364] 42. L. Magno, C. B. Lessard, M. Martins, P. Cruz, M. Katan, J. Bilsland, P.
Chakrabaty, T. E. Golde, P. Whiting, Alzheimer’s disease Phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph, Alzheimer's Research & Therapy 11,
1 (2019).
[0365] 43. J. D. Milner, PLAID: a Syndrome of Complex Patterns of Disease and Unique
Phenotypes, ./ Clin. Immunol. 35, 527-530 (2015).
[0366] 44. A. Nott, I. R. Holtman, N. G. Coufal, J. C. M. Schlachetzki, M. Yu, R. Hu, C.
Z. Han, M. Pena, J. Xiao, Y. Wu, Z. Keulen, M. P. Pasillas, C. O’Connor, C. K. Nickl, S. T. Schafer, Z. Shen, R. A. Rissman, J. B. Brewer, D. Gosselin, D. D. Gonda, M. L. Levy, M. G. Rosenfeld, G. McVicker, F. H. Gage, B. Ren, C. K. Glass, Brain cell type-specific enhancer- promoter interactome maps and disease risk association, Science 366, 1134-1139 (2019).
[0367] 45. T. E. Golde, Harnessing immunoproteostasis to treat neurodegenerative disorders, Neuron 101, 1003-1015 (2019).
[0368] 46. I. Litvan, K. P. Bhatia, D. J. Bum, C. G. Goetz, A. E. Lang, I. McKeith, N.
Quinn, K. D. Sethi, C. Shults, G. K. Wenning, SIC Task Force appraisal of clinical diagnostic criteria for parkinsonian disorders, Movement Disord. 18, 467-486 (2003).
[0369] 47. B.A. Cholerton, C.P. Zabetian, J.F. Quinn, K.A. Chung, A. Peterson, A.J.
Espay, F.J. Revilla, J. Devoto, G. Watson, S.C. Hu, K.L. Edwards, Pacific Northwest Udall Center of excellence clinical consortium: study design and baseline cohort characteristics, J. Parkinson dis., 3, 205-214 (2013).
[0370] 48. G. M. McKhann, D. S. Knopman, H. Chertkow, B. T. Hyman, C. R. Jack, C.
H. Kawas, W. E. Klunk, W. J. Koroshetz, J. J. Manly, R. Mayeux, R. C. Mohs, J. C. Morris, M. N. Rossor, P. Scheltens, M. C. Carrillo, B. Thies, S. Weintraub, C. H. Phelps, The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging- Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease, Alzheimer ’s & Dementia 7, 263-269 (2011).
[0371] 49. H. W. Grievink, T. Luisman, C. Kluft, M. Moerland, K. E. Malone,
Comparison of three isolation techniques for human peripheral blood mononuclear cells: Cell recovery and viability, population composition, and cell functionality, Biopreserv. Biobank. 14, 410-415 (2016).
[0372] 50. R. Fernandez, H. Maecker, Cytokine-stimulated phosphoflow of PBMC using
CyTOF mass cytometry, Bio Protoc. 5 (2015). [0373] 51. E. R. Zunder, R. Finck, G. K. Behbehani, E.-A. D. Amir, S. Krishnaswamy, V.
D. Gonzalez, C. G. Lorang, Z. Bjomson, M. H. Spitzer, B. Bodenmiller, W. J. Fantl, D.
Pe’er, G. P. Nolan, Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm, Nat. Protoc. 10, 316-333 (2015).
[0374] 52. J. Choi, R. Fernandez, H. T. Maecker, M. J. Butte, Systems approach to uncover signaling networks in primary immunodeficiency diseases, J. Allergy Clin. Immunol. 140, 881-884 (2017).
[0375] 53. S. C. Bendall, E. F. Simonds, P. Qiu, E.-A. D. Amir, P. O. Krutzik, R. Finck,
R. V. Bruggner, R. Melamed, A. Trejo, O. I. Omatsky, R. S. Balderas, S. K. Plevritis, K. Sachs, D. Pe’er, S. D. Tanner, G. P. Nolan, Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum, Science 332, 687-696 (2011).
[0376] 54. R. Finck, E. F. Simonds, A. Jager, S. Krishnaswamy, K. Sachs, W. Fantl, D.
Pe’er, G. P. Nolan, S. C. Bendall, Normalization of mass cytometry data with bead standards, Cytom. Part A 83, 483-494 (2013).
[0377] 55. W. E. O’Gorman, H. Huang, Y.-L. Wei, K. L. Davis, M. D. Leipold, S. C.
Bendall, B. A. Kidd, C. L. Dekker, H. T. Maecker, Y.-H. Chien, M. M. Davis, The Split Virus Influenza Vaccine rapidly activates immune cells through Fey receptors, Vaccine 32, 5989-5997 (2014).
[0378] 56. C. L. Galligan, J. C. Siebert, K. A. Siminovitch, E. C. Keystone, V. Bykerk,
O. D. Perez, E. N. Fish, Multiparameter phospho-flow analysis of lymphocytes in early rheumatoid arthritis: implications for diagnosis and monitoring drug therapy, PLoS One 4, e6703 (2009).
[0379] 57. W. Xu, F. Fang, J. Ding, C. Wu, Dysregulation of Rab5 -mediated endocytic pathways in Alzheimer’s disease, Traffic 19, 253-262 (2018).
[0380] 58. W. E. Johnson, W. Evan Johnson, C. Li, A. Rabinovic, Adjusting batch effects in microarray expression data using empirical Bayes methods, Biostatistics 8, 118-127 (2007).
[0381] 59. A. Scherer, Batch Effects and Noise in Microarray Experiments: Sources and
Solutions (John Wiley & Sons, Hoboken, NJ, 2009). [0382] 60. V. M. R. Muggeo, Estimating regression models with unknown break-points,
Stat. Med. 22, 3055-3071 (2003).
[0383] 61. G. H. Laurens van der Maaten, Visualizing data using t-SNE, J. Mach. Learn.
Res. 9, 2579-2605 (2008). [0384] 62. L. Mclnnes, J. Healy, N. Saul, L. GroBberger. UMAP: Uniform manifold approximation and projection, Journal of Open Source Software 3, 861 (2018).
EMBODIMENTS
[0385] Embodiment 1. A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease, said method comprising:
(i) obtaining or having obtained a sample from a subject having or being at risk of developing a neurological disease;
(ii) isolating a peripheral blood mononuclear cell (PBMC) from said sample; wherein said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
(iii) contacting said PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein said stimulatory agent is interferon a (IFN-a), interleukin-6 (IL- 6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopolysaccharides (LPS) or phorbol myristate acetate (PMA); and
(iv) detecting a level of phosphorylation of an intracellular signaling molecule in said ex vivo stimulated PBMC, wherein said intracellular signaling molecule is PLC-y2, AKT, STAT1 or STAT5.
[0386] Embodiment 2. A method of treating a neurological disease in a subject in need thereof, said method comprising:
(i) obtaining or having obtained a sample from a subject having a neurological disease;
(ii) isolating a peripheral blood mononuclear cell (PBMC) from said sample; wherein said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
(iii) contacting said PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein said stimulatory agent is interferon a (IFN-a), interleukin-6 (IL- 6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopolysaccharides (LPS) or phorbol myristate acetate (PMA);
(iv) detecting a level of phosphorylation of an intracellular signaling molecule in said ex vivo stimulated PBMC, wherein said intracellular signaling molecule is PLC-D2, AKT, STAT1 or STAT5; and (v) administering to said subject a therapeutically effective amount of a neurological treatment.
[0387] Embodiment 3. A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject undergoing treatment for a neurological disease, said method comprising:
(i) obtaining or having obtained a sample from a subject undergoing treatment for a neurological disease;
(ii) isolating a peripheral blood mononuclear cell (PBMC) from said sample; wherein said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
(iii) contacting said PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein said stimulatory agent is interferon a (IFN-a), interleukin-6 (IL- 6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopolysaccharides (LPS) or phorbol myristate acetate (PMA); and
(iv) detecting a level of phosphorylation of an intracellular signaling molecule in said ex vivo stimulated PBMC, wherein said intracellular signaling molecule is PLC-D2, AKT, STAT1 or STAT5.
[0388] Embodiment 4. A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease, said method comprising:
(i) obtaining or having obtained a sample from a subject having or being at risk of developing a neurological disease;
(ii) isolating a peripheral blood mononuclear cell (PBMC) from said sample; wherein said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
(iii) contacting said PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein said stimulatory agent is interferon a (IFN-a), interleukin-6 (IL- 6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopolysaccharides (LPS) or phorbol myristate acetate (PMA); (iv) detecting a level of phosphorylation of an intracellular signaling molecule in said ex vivo stimulated PBMC, wherein said intracellular signaling molecule is PLC-D2, AKT, STAT1 or STAT5; and
(v) comparing said level of phosphorylation to a standard control, thereby detecting a level of phosphorylation of an intracellular signaling molecule in a subject.
[0389] Embodiment 5. The method of embodiment 4, wherein an increased level of phosphorylation relative to said standard control indicates that said subject has or is at risk of developing a neurological disease.
[0390] Embodiment 6. The method of embodiment 4, wherein a decreased level of phosphorylation relative to said standard control indicates that said subject has or is at risk of developing a neurological disease.
[0391] Embodiment 7. The method of any one of embodiments 4-6, comprising based at least in part on said level of phosphorylation in step (v) administering a neurological disease treatment to said subject.
[0392] Embodiment 8. The method of any one of embodiments 1-7, wherein said intracellular signaling molecule is PLC-y2.
[0393] Embodiment 9. The method of embodiment 8, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD 16h'"h NK cell, a CD56bnght NK cell, a CD56dimCD16dim NK cell or a CD16hl"h monocyte.
[0394] Embodiment 10. The method of embodiment 8 or 9, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell.
[0395] Embodiment 11. The method of embodiment 8 or 9, wherein said stimulatory agent is IL-21 or LPS and said PBMC is a CDie1^11 monocyte.
[0396] Embodiment 12. The method of embodiment 8 or 9, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD 16high NK cell, a CD56bnght NK cell, or a CD56dimCD16dim NK cell.
[0397] Embodiment 13. The method of embodiment 8, wherein said stimulatory agent is IL-6 and said PBMC is a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
[0398] Embodiment 14. The method of embodiment 8 or 13, wherein said stimulatory agent is IL-6 and said PBMC is a CD8+ activated T cell.
[0399] Embodiment 15. The method of embodiment 8 or 13, wherein said stimulatory agent is IL-6 and said PBMC is a CD4+ CD8+ T cell or a natural killer T cell.
[0400] Embodiment 16. The method of embodiment 8, wherein said stimulatory agent is IFN-a or IL-7 and said PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD56bnght NK cell, or a CDie1^11 monocyte.
[0401] Embodiment 17. The method of embodiment 8 or 16, wherein said stimulatory agent is IFN-a or IL-7 and said PBMC is a natural killer T cell.
[0402] Embodiment 18. The method of embodiment 8 or 16, wherein said stimulatory agent is IFN-a or IL-7 and said PBMC is a CD8+ activated T cell or a CDlh^811 monocyte.
[0403] Embodiment 19. The method of embodiment 8 or 16, wherein said stimulatory agent is IFN-a or IL-7 and said PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, or a CD56bright NK cell.
[0404] Embodiment 20. The method of embodiment 8, wherein said stimulatory agent is PMA and said PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
[0405] Embodiment 21. The method of embodiment 8 or 20, wherein said stimulatory agent is PMA and said PBMC is a CD4+ activated T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell. [0406] Embodiment 22. The method of embodiment 8 or 20, wherein said stimulatory agent is PMA and said PBMC is a CD8+ activated T cell.
[0407] Embodiment 23. The method of any one of embodiments 1 -7, wherein said intracellular signaling molecule is STAT1.
[0408] Embodiment 24. The method of embodiment 23, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8 T cell, a CD4+ CD8+ T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD56bnghtNK cell, a myeloid dendritic cell (mDC) or a plasmacytoid dendritic cell (pDC).
[0409] Embodiment 25. The method of embodiment 23, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
[0410] Embodiment 26. The method of embodiment 23, wherein said stimulatory agent is LPS and said PBMC is a CD8+ central memory T cell.
[0411] Embodiment 27. The method of any one of embodiments 1-7, wherein said intracellular signaling molecule is STAT5.
[0412] Embodiment 28. The method of embodiment 27, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8 T cell, a CD16high monocyte or a CD16low monocyte.
[0413] Embodiment 29. The method of embodiment 27 or 28, wherein said stimulatory agent is IFN-a and said PBMC is a CD 16hlgh monocyte or a CD16low monocyte.
[0414] Embodiment 30. The method of embodiment 27 or 28, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, or a CD4 CD8 T cell.
[0415] Embodiment 31. The method of embodiment 27, wherein said stimulatory agent is IL-7 and said PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, or a CD8+ effector memory T cell. [0416] Embodiment 32. The method of embodiment 27, wherein said stimulatory agent is LPS and said PBMC is a CD4+ CD8+ T cell.
[0417] Embodiment 33. The method of embodiment 8, wherein said subject is a male subject.
[0418] Embodiment 34. The method of embodiment 33, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD1611"1’ NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD161'1"1' monocyte or a CD16low monocyte.
[0419] Embodiment 35. The method of embodiment 33 or 34, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
[0420] Embodiment 36. The method of embodiment 33 or 34, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CDie^NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16hlgh monocyte or a CD16low monocyte.
[0421] Embodiment 37. The method of embodiment 33, wherein said stimulatory agent is IFN-a, IL-6 or IL-7 and said PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD16high monocyte or a CD16low monocyte.
[0422] Embodiment 38. The method of embodiment 33, wherein said stimulatory agent is PMA and said PBMC is a CD4+ activated T cell, a CD4+ naive T cell, a CD8+ activated T cell, a natural killer T cell, a CD4+ CD8+ T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell. [0423] Embodiment 39. The method of embodiment 33 or 38, wherein said stimulatory agent is PMA and said PBMC is a CD4+ activated T cell, a CD4+ naive T cell, or a natural killer T cell.
[0424] Embodiment 40. The method of embodiment 33 or 38, wherein said stimulatory agent is PMA and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
[0425] Embodiment 41. The method of embodiment 33 or 38, wherein said stimulatory agent is PMA and said PBMC is a CD8+ activated T cell or a CD4+ CD8+ T cell.
[0426] Embodiment 42. The method of any one of embodiments 1 -7, wherein said intracellular signaling molecule is AKT.
[0427] Embodiment 43. The method of embodiment 42, wherein said subject is a male subject.
[0428] Embodiment 44. The method of embodiment 43, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a
CD4+ naive T cell, or a regulatory T cell.
[0429] Embodiment 45. The method of embodiment 43 or 44, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
[0430] Embodiment 46. The method of embodiment 43 or 44, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4+ effector memory T cell.
[0431] Embodiment 47. The method of embodiment 43, wherein said stimulatory agent is unstimulated and said PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
[0432] Embodiment 48. The method of embodiment 43, wherein said stimulatory agent is IL-7 and said PBMC is a CD4+ activated T cell. [0433] Embodiment 49. The method of embodiment 43, wherein said stimulatory agent is PMA and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a mDC, a pDC, a CD 161""1' monocyte or a CD16low monocyte.
[0434] Embodiment 50. The method of embodiment 23, wherein said subject is a female subject.
[0435] Embodiment 51. The method of embodiment 50, wherein said stimulatory agent is unstimulated, IL-7, IL-10 or LPS and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, or a B translational cell.
[0436] Embodiment 52. The method of embodiment 50 or 51, wherein said stimulatory agent is unstimulated and said PBMC is a B naive cell.
[0437] Embodiment 53. The method of embodiment 50 or 51, wherein said stimulatory agent is unstimulated, IL-7, IL-10 or LPS and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, or a B translational cell.
[0438] Embodiment 54. The method of embodiment 50, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a CD4 CD8 T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CDie^NK cell, a CD56bnght NK cell, a mDC, a pDC, a CD 161""1' monocyte or a CD16low monocyte.
[0439] Embodiment 55. The method of embodiment 50 or 51, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, or a pDC.
[0440] Embodiment 56. The method of embodiment 50 or 51, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD4+ CD8+ T cell, a CD4 CD8 T cell, a natural killer T cell, a plasmablast cell, or a CD56bnght NK cell.
[0441] Embodiment 57. The method of embodiment 50 or 51, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ naive T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, a B translational cell, a CD 16h'"h NK cell, a mDC, a CD 16h'"h monocyte or a CD16low monocyte.
[0442] Embodiment 58. The method of embodiment 50, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
[0443] Embodiment 59. The method of embodiment 50 or 58, wherein said stimulatory agent is IL-6 and said PBMC is an IgD CD27 B cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
[0444] Embodiment 60. The method of embodiment 50 or 58, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD+ B memory cell, or a B naive cell.
[0445] Embodiment 61. The method of embodiment 50, wherein said stimulatory agent is IL-21 and said PBMC is an IgD CD27 B cell.
[0446] Embodiment 62. The method of embodiment 27, wherein said subject is a female subject. [0447] Embodiment 63. The method of embodiment 62, wherein said stimulatory agent is
IFN-a and said PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8 T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD 16h'"h monocyte or a CD16low monocyte.
[0448] Embodiment 64. The method of embodiment 62 or 63, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ effector memory T cell.
[0449] Embodiment 65. The method of embodiment 62 or 63, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ effector T cell, a CD8+ central memory T cell, a CD8+ naive T cell, or a natural killer T cell.
[0450] Embodiment 66. The method of embodiment 62 or 63, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ activated T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ effector T cell, a CD4 CD8 T cell, a CD4+ CD8+ T cell, a CD 16h'"h monocyte or a CD16low monocyte. [0451] Embodiment 67. The method of embodiment 62, wherein said stimulatory agent is IL-7 and said PBMC is a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD4 CD8 T cell, a CD4+ CD8+ T cell or a natural killer T cell.
[0452] Embodiment 68. The method of embodiment 62 or 66, wherein said stimulatory agent is IL-7 and said PBMC is a CD8+ central memory T cell, or a CD8+ effector memory T cell.
[0453] Embodiment 69. The method of embodiment 62 or 66, wherein said stimulatory agent is IL-7 and said PBMC is a natural killer T cell.
[0454] Embodiment 70. The method of embodiment 62 or 66, wherein said stimulatory agent is IL-7 and said PBMC is a CD8+ effector T cell, a CD4 CD8 T cell, or a CD4+ CD8+ T cell.
[0455] Embodiment 71. The method of any one of embodiments 1-70, wherein said neurological disease is Alzheimer’s disease or Parkinson’s disease.
[0456] Embodiment 72. The method of embodiment 71, wherein said intracellular signaling molecule is PLC-y2.
[0457] Embodiment 73. The method of embodiment 71 or 72, wherein said stimulatory agent is unstimulated and said PBMC is a CD56bnght NK cell.
[0458] Embodiment 74. The method of embodiment 71 or 72, wherein said stimulatory agent is IL-10, IL-21 or LPS and said PBMC is a basophil. [0459] Embodiment 75. The method of embodiment 71, wherein said intracellular signaling molecule is STAT1.
[0460] Embodiment 76. The method of embodiment 71 or 75, wherein said stimulatory agent is IFN-a and said PBMC is a plasmablast or CD56bnght NK cell.
[0461] Embodiment 77. The method of embodiment 71, wherein said intracellular signaling molecule is STAT5.
[0462] Embodiment 78. The method of embodiment 71 or 77, wherein said stimulatory agent is IFN-a and said PBMC is a CD 16h'"h monocyte or a CD16low monocyte.
[0463] Embodiment 79. The method of any one of embodiments 1-71, wherein said neurological disease is Alzheimer’s disease. [0464] Embodiment 80. The method of embodiment 79, wherein said intracellular signaling molecule is PLC-y2.
[0465] Embodiment 81. The method of embodiment 80, wherein said stimulatory agent is IL-10, IL-21, LPS or PMA and said PBMC is a CD4+ CD8+ T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
[0466] Embodiment 82. The method of embodiment 80 or 81, wherein said stimulatory agent is IL-10, IL-21, LPS or PMA and said PBMC is an IgA B cell or a B translational cell.
[0467] Embodiment 83. The method of embodiment 80 or 81, wherein said stimulatory agent is IL-10, IL-21, LPS or PMA and said PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, or a B switched memory cell.
[0468] Embodiment 84. The method of embodiment 80, wherein said stimulatory agent is IL-7, IL-10, or IL-21 and said PBMC is a CD8+ activated T cell.
[0469] Embodiment 85. The method of embodiment 79, wherein said intracellular signaling molecule is STAT1.
[0470] Embodiment 86. The method of embodiment 85, wherein said stimulatory agent is unstimulated, and said PBMC is an IgD+ B memory cell, an IgD CD27 B cell or a B naive cell.
[0471] Embodiment 87. The method of embodiment 85, wherein said stimulatory agent is IL-6 or IL-7 and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD161""1' NK cell, a CDSe^^ NK cell, or a CD56dimCD16dim NK cell.
[0472] Embodiment 88. The method of embodiment 85, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
[0473] Embodiment 89. The method of embodiment 85, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8- T cell, an IgD CD27 B cell, a B naive cell, a Oϋΐό^11 NK cell, a 0056^^ NK cell, or a CD56dimCD16dim NK cell.
[0474] Embodiment 90. The method of embodiment 85 or 89, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD56dimCD16dim NK cell.
[0475] Embodiment 91. The method of embodiment 85 or 89, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4- CD8- T cell, an IgD CD27 B cell, a B naive cell, a CD 16^ NK cell, or a CD56bri§ht NK cell.
[0476] Embodiment 92. The method of any one of embodiments 1-91, wherein said sample from said subject is a blood sample or a plasma sample.
[0477] Embodiment 93. The method of any one of embodiments 1-92, wherein said detecting comprises contacting said ex vivo stimulated PBMC with one or more immune cell specific antibodies, thereby forming a labeled ex vivo stimulated PBMC.
[0478] Embodiment 94. The method of embodiment 93, wherein said one or more immune cell-specific antibodies comprises a detectable moiety.
[0479] Embodiment 95. The method of embodiment 93 or 94, wherein said one or more immune cell-specific antibodies are an anti-CD3, an anti-CD4 antibody, an anti-CD7 antibody, an anti-CD8 antibody, an anti-CDllb antibody, an anti-CDllc antibody, an anti- CD 14 antibody, an anti-CD 16 antibody, an anti-CD 19 antibody, an anti-CD20 antibody, an anti-CD24 antibody, an anti-CD25 antibody, an anti-CD27 antibody, an anti-CD38 antibody, an anti-CD45RA antibody, an anti-CD56 antibody, an anti-CD123 antibody, an anti-CD127 antibody, an anti-IgA antibody, an anti-IgD antibody or an anti-HLA-Dr antibody.
[0480] Embodiment 96. The method of any one of embodiments 93-95, wherein said detecting further comprises contacting said labeled ex vivo stimulated PBMC with one or more anti-intracellular signaling molecule antibodies, thereby forming a intracellularly labeled ex vivo stimulated PBMC.
[0481] Embodiment 97. The method of embodiment 96, wherein said one or more anti- intracellular signaling molecule antibodies are an anti-pERKl/2 antibody, an anti-IkBa antibody, an anti-NF-kB antibody, an anti-p38 antibody, an anti-pAKT antibody, an anti- pCREB antibody, an anti-pLCK antibody, an anti-pPLC-y2 antibody, an anti-pS6 antibody, an anti-pSTATl antibody, an anti-pSTAT3 antibody, an anti-pSTAT5 antibody, an anti- Lamp2 antibody, an anti-EEAl antibody, or an anti-Rab5 antibody. [0482] Embodiment 98. The method of any one of embodiments 96-97, wherein said detecting a level of phosphorylation comprises detecting binding of said one or more anti- intracellular signaling molecule antibodies to said labeled ex vivo stimulated PBMC.
[0483] Embodiment 99. The method of any one of embodiments 1-98, wherein said ex vivo stimulated PBMC is a permeabilized PBMC. [0484] Embodiment 100. The method of any one of embodiments 1-99, wherein said ex vivo stimulated PBMC is in a detection device.
[0485] Embodiment 101. The method of any one of embodiments 2 or 7-100, wherein said neurological disease treatment is a PLC-y2 pathway activator.
[0486] Embodiment 102. The method of any one of embodiments 2 or 7-100, wherein said neurological disease treatment is galantamine, rivastigmine, donepezil, memantine, levodopa, carbidopa, amantadine, a dopamine agonist, a monoamine oxidase B inhibitor, a Catechol-O- methyltransferase inhibitor, or an anticholinergic.

Claims

WHAT IS CLAIMED IS:
1. A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease, said method comprising:
(i) obtaining or having obtained a sample from a subject having or being at risk of developing a neurological disease;
(ii) isolating a peripheral blood mononuclear cell (PBMC) from said sample; wherein said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
(iii) contacting said PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein said stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA); and
(iv) detecting a level of phosphorylation of an intracellular signaling molecule in said ex vivo stimulated PBMC, wherein said intracellular signaling molecule is PLC-y2,
AKT, STAT1 or STAT5.
2. A method of treating a neurological disease in a subject in need thereof, said method comprising:
(i) obtaining or having obtained a sample from a subject having a neurological disease;
(ii) isolating a peripheral blood mononuclear cell (PBMC) from said sample; wherein said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
(iii) contacting said PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein said stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA);
(iv) detecting a level of phosphorylation of an intracellular signaling molecule in said ex vivo stimulated PBMC, wherein said intracellular signaling molecule is PLC-y2,
AKT, STAT1 or STAT5; and
(v) administering to said subject a therapeutically effective amount of a neurological treatment.
3. A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject undergoing treatment for a neurological disease, said method comprising:
(i) obtaining or having obtained a sample from a subject undergoing treatment for a neurological disease;
(ii) isolating a peripheral blood mononuclear cell (PBMC) from said sample; wherein said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
(iii) contacting said PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein said stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA); and
(iv) detecting a level of phosphorylation of an intracellular signaling molecule in said ex vivo stimulated PBMC, wherein said intracellular signaling molecule is PLC-y2, AKT, STAT1 or STAT5.
4. A method of detecting a level of phosphorylation of an intracellular signaling molecule in a subject having or being at risk of developing a neurological disease, said method comprising:
(i) obtaining or having obtained a sample from a subject having or being at risk of developing a neurological disease;
(ii) isolating a peripheral blood mononuclear cell (PBMC) from said sample; wherein said PBMC is a CD4+ T cell, a CD8+ T cell, a natural killer T cell, a natural killer (NK) cell, a B cell, a monocyte, a basophil, a plasmablast or a dendritic cell (DC);
(iii) contacting said PBMC with a stimulatory agent ex vivo, thereby forming an ex vivo stimulated PBMC, wherein said stimulatory agent is interferon a (IFN-a), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin- 10 (IL-10), interleukin-21 (IL-21), lipopoly saccharides (LPS) or phorbol myristate acetate (PMA);
(iv) detecting a level of phosphorylation of an intracellular signaling molecule in said ex vivo stimulated PBMC, wherein said intracellular signaling molecule is PLC-y2, AKT, STAT1 or STAT5; and
(v) comparing said level of phosphorylation to a standard control, thereby detecting a level of phosphorylation of an intracellular signaling molecule in a subject.
5. The method of claim 4, wherein an increased level of phosphorylation relative to said standard control indicates that said subject has or is at risk of developing a neurological disease.
6. The method of claim 4, wherein a decreased level of phosphorylation relative to said standard control indicates that said subject has or is at risk of developing a neurological disease.
7. The method of claim 4, comprising based at least in part on said level of phosphorylation in step (v) administering a neurological disease treatment to said subject.
8. The method of claim 1, wherein said intracellular signaling molecule is
PLC-Y2.
9. The method of claim 8, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD4+ activated T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16h'"h NK cell, a CD56bn"lu NK cell, a CD56dimCD16dim NK cell or a CDie1^11 monocyte.
10. The method of claim 8, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, or a natural killer T cell.
11. The method of claim 8, wherein said stimulatory agent is IL-21 or LPS and said PBMC is a Oϋΐό^11 monocyte.
12. The method of claim 8, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA B cell, an IgD+
B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD 16h'"h NK cell, a CD56bnght NK cell, or a CD56dimCD16dim NK cell.
13. The method of claim 8, wherein said stimulatory agent is IL-6 and said PBMC is a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
14. The method of claim 8, wherein said stimulatory agent is IL-6 and said PBMC is a CD8+ activated T cell.
15. The method of claim 8, wherein said stimulatory agent is IL-6 and said PBMC is a CD4+ CD8+ T cell or a natural killer T cell.
16. The method of claim 8, wherein said stimulatory agent is IFN-a or IL- 7 and said PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD56bnght NK cell, or a CD 161""1' monocyte.
17. The method of claim 8, wherein said stimulatory agent is IFN-a or IL- 7 and said PBMC is a natural killer T cell.
18. The method of claim 8, wherein said stimulatory agent is IFN-a or IL- 7 and said PBMC is a CD8+ activated T cell or a CDie1^11 monocyte.
19. The method of claim 8, wherein said stimulatory agent is IFN-a or IL- 7 and said PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, or a CD56bright NK cell.
20. The method of claim 8, wherein said stimulatory agent is PMA and said PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
21. The method of claim 8, wherein said stimulatory agent is PMA and said PBMC is a CD4+ activated T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
22. The method of claim 8, wherein said stimulatory agent is PMA and said PBMC is a CD8+ activated T cell.
23. The method of claim 1, wherein said intracellular signaling molecule is STAT1.
24. The method of claim 23, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8 T cell, a CD4+ CD8+ T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD56bnght NK cell, a myeloid dendritic cell (mDC) or a plasmacytoid dendritic cell (pDC).
25. The method of claim 23, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
26. The method of claim 23, wherein said stimulatory agent is LPS and said PBMC is a CD8+ central memory T cell.
27. The method of claim 1, wherein said intracellular signaling molecule is STAT5.
28. The method of claim 27, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8 T cell, a CDie1^11 monocyte or a CD16low monocyte.
29. The method of claim 27, wherein said stimulatory agent is IFN-a and said PBMC is a CD 16h'"h monocyte or a CD16low monocyte.
30. The method of claim 27, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, or a CD4 CD8 T cell.
31. The method of claim 27, wherein said stimulatory agent is IL-7 and said PBMC is a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, or a CD8+ effector memory T cell.
32. The method of claim 27, wherein said stimulatory agent is LPS and said PBMC is a CD4+ CD8+ T cell.
33. The method of claim 8, wherein said subject is a male subject.
34. The method of claim 33, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a CD4+ activated T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD1 high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16hlgh monocyte or a CD16low monocyte.
35. The method of claim 33, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a CD4+ activated T cell, a CD8+ activated T cell, a CD4+ CD8+ T cell or a natural killer T cell.
36. The method of claim 33, wherein said stimulatory agent is unstimulated, IL-10, IL-21 or LPS and said PBMC is a basophil, a regulatory T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD1 high NK cell, a CD56bright NK cell, a CD56dimCD16dim NK cell, a CD16hl"h monocyte or a CD16low monocyte.
37. The method of claim 33, wherein said stimulatory agent is IFN-a, IL-6 or IL-7 and said PBMC is a CD4+ activated T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD8+ activated T cell, a CD 16hlgh monocyte or a CD16low monocyte.
38. The method of claim 33, wherein said stimulatory agent is PMA and said PBMC is a CD4+ activated T cell, a CD4+ naive T cell, a CD8+ activated T cell, a natural killer T cell, a CD4+ CD8+ T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
39. The method of claim 33, wherein said stimulatory agent is PMA and said PBMC is a CD4+ activated T cell, a CD4+ naive T cell, or a natural killer T cell.
40. The method of claim 33, wherein said stimulatory agent is PMA and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, or a B translational cell.
41. The method of claim 33, wherein said stimulatory agent is PMA and said PBMC is a CD8+ activated T cell or a CD4+ CD8+ T cell.
42. The method of claim 1, wherein said intracellular signaling molecule is AKT.
43. The method of claim 42, wherein said subject is a male subject.
44. The method of claim 43, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, or a regulatory T cell.
45. The method of claim 43, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
46. The method of claim 43, wherein said stimulatory agent is unstimulated, IFN-a, IL-6, IL-7, IL-10, IL-21 or LPS and said PBMC is a CD4+ effector memory T cell.
47. The method of claim 43, wherein said stimulatory agent is unstimulated and said PBMC is a CD4+ activated T cell, a CD4+ central memory T cell, a CD4+ effector T cell, a CD4+ naive T cell, or a regulatory T cell.
48. The method of claim 43, wherein said stimulatory agent is IL-7 and said PBMC is a CD4+ activated T cell.
49. The method of claim 43, wherein said stimulatory agent is PMA and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a mDC, a pDC, a CD16high monocyte or a CD16low monocyte.
50. The method of claim 23, wherein said subject is a female subject.
51. The method of claim 50, wherein said stimulatory agent is unstimulated, IL-7, IL-10 or LPS and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, or a B translational cell.
52. The method of claim 50, wherein said stimulatory agent is unstimulated and said PBMC is a B naive cell.
53. The method of claim 50, wherein said stimulatory agent is unstimulated, IL-7, IL-10 or LPS and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, or a B translational cell.
54. The method of claim 50, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4+ CD8+ T cell, a CD4 CD8 T cell, a natural killer T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD16high NK cell, a CD56bnght NK cell, a mDC, a pDC, a CD 16high monocyte or a CD16low monocyte.
55. The method of claim 50, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, or a pDC.
56. The method of claim 50, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD4+ CD8+ T cell, a CD4 CD8 T cell, a natural killer T cell, a plasmablast cell, or a CD56bnght NK cell.
57. The method of claim 50, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ naive T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a B switched memory cell, a B translational cell, a CD1611"1’ NK cell, a mDC, a CD 161""1' monocyte or a CD16low monocyte.
58. The method of claim 50, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
59. The method of claim 50, wherein said stimulatory agent is IL-6 and said PBMC is an IgD CD27 B cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
60. The method of claim 50, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD+ B memory cell, or a B naive cell.
61. The method of claim 50, wherein said stimulatory agent is IL-21 and said PBMC is an IgD CD27 B cell.
62. The method of claim 27, wherein said subject is a female subject.
63. The method of claim 62, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ activated T cell, a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8 T cell, a CD4+ CD8+ T cell, a natural killer T cell, a CD 16h'"h monocyte or a CD16low monocyte.
64. The method of claim 62, wherein said stimulatory agent is IFN-a and said PBMC is a CD8+ effector memory T cell.
65. The method of claim 62, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ effector T cell, a CD8+ central memory T cell, a CD8+ naive T cell, or a natural killer T cell.
66. The method of claim 62, wherein said stimulatory agent is IFN-a and said PBMC is a CD4+ activated T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a CD8+ activated T cell, a CD8+ effector T cell, a CD4 CD8 T cell, a CD4+ CD8+ T cell, a CD 16h'"h monocyte or a CD 16low monocyte.
67. The method of claim 62, wherein said stimulatory agent is IL-7 and said PBMC is a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD4 CD8 T cell, a CD4+ CD8+ T cell or a natural killer T cell.
68. The method of claim 62, wherein said stimulatory agent is IL-7 and said PBMC is a CD8+ central memory T cell, or a CD8+ effector memory T cell.
69. The method of claim 62, wherein said stimulatory agent is IL-7 and said PBMC is a natural killer T cell.
70. The method of claim 62, wherein said stimulatory agent is IL-7 and said PBMC is a CD8+ effector T cell, a CD4 CD8 T cell, or a CD4+ CD8+ T cell.
71. The method of claim 1, wherein said neurological disease is Alzheimer’s disease or Parkinson’s disease.
72. The method of claim 71, wherein said intracellular signaling molecule is PLC-Y2.
73. The method of claim 71, wherein said stimulatory agent is unstimulated and said PBMC is a CD56bnght NK cell.
74. The method of claim 71, wherein said stimulatory agent is IL- 10, IL- 21 or LPS and said PBMC is a basophil.
75. The method of claim 71, wherein said intracellular signaling molecule is STAT1.
76. The method of claim 71, wherein said stimulatory agent is IFN-a and said PBMC is a plasmablast or CD56bnght NK cell.
77. The method of claim 71, wherein said intracellular signaling molecule is STAT5.
78. The method of claim 71, wherein said stimulatory agent is IFN-a and said PBMC is a CD 16h'"h monocyte or a CD16low monocyte.
79. The method of claim 1, wherein said neurological disease is Alzheimer’s disease.
80. The method of claim 79, wherein said intracellular signaling molecule is PLC-Y2.
81. The method of claim 80, wherein said stimulatory agent is IL-10, IL- 21, LPS or PMA and said PBMC is a CD4+ CD8+ T cell, an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
82. The method of claim 80, wherein said stimulatory agent is IL-10, IL- 21, LPS or PMA and said PBMC is an IgA B cell or a B translational cell.
83. The method of claim 80, wherein said stimulatory agent is IL-10, IL- 21, LPS or PMA and said PBMC is a CD4+ CD8+ T cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, or a B switched memory cell.
84. The method of claim 80, wherein said stimulatory agent is IL-7, IL-10, or IL-21 and said PBMC is a CD8+ activated T cell.
85. The method of claim 79, wherein said intracellular signaling molecule is STAT1.
86. The method of claim 85, wherein said stimulatory agent is unstimulated, and said PBMC is an IgD+ B memory cell, an IgD CD27 B cell or a B naive cell.
87. The method of claim 85, wherein said stimulatory agent is IL-6 or IL-7 and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, a B translational cell, a CD161""1' NK cell, a CD56bright NK cell, or a CD56dimCD16dim NK cell.
88. The method of claim 85, wherein said stimulatory agent is IL-6 and said PBMC is an IgA B cell, an IgD+ B memory cell, an IgD CD27 B cell, a B naive cell, a plasmablast cell, a B switched memory cell, or a B translational cell.
89. The method of claim 85, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8 T cell, an IgD CD27 B cell, a B naive cell, a Oϋΐό^11 NK cell, a CD56bri§ht NK cell, or a CD56dimCD16dim NK cell.
90. The method of claim 85, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD56dimCD16dimNK cell.
91. The method of claim 85, wherein said stimulatory agent is LPS or PMA and said PBMC is a CD4+ effector T cell, a CD4+ effector memory T cell, a CD4+ naive T cell, a regulatory T cell, a CD8+ activated T cell, a CD8+ central memory T cell, a CD8+ effector T cell, a CD8+ effector memory T cell, a CD8+ naive T cell, a CD4 CD8 T cell, an IgD CD27 B cell, a B naive cell, a CD^11 NK cell, or a CD56bri§ht NK cell.
92. The method of claim 1, wherein said sample from said subject is a blood sample or a plasma sample.
93. The method of claim 1, wherein said detecting comprises contacting said ex vivo stimulated PBMC with one or more immune cell specific antibodies, thereby forming a labeled ex vivo stimulated PBMC.
94. The method of claim 93, wherein said one or more immune cell- specific antibodies comprises a detectable moiety.
95. The method of claim 93, wherein said one or more immune cell- specific antibodies are an anti-CD3, an anti-CD4 antibody, an anti-CD7 antibody, an anti- CD8 antibody, an anti-CDllb antibody, an anti-CDllc antibody, an anti-CD14 antibody, an anti-CD 16 antibody, an anti-CD 19 antibody, an anti-CD20 antibody, an anti-CD24 antibody, an anti-CD25 antibody, an anti-CD27 antibody, an anti-CD38 antibody, an anti-CD45RA antibody, an anti-CD56 antibody, an anti-CD 123 antibody, an anti-CD 127 antibody, an anti- IgA antibody, an anti-IgD antibody or an anti-HLA-Dr antibody.
96. The method of claim 93, wherein said detecting further comprises contacting said labeled ex vivo stimulated PBMC with one or more anti-intracellular signaling molecule antibodies, thereby forming a intracellularly labeled ex vivo stimulated PBMC.
97. The method of claim 96, wherein said one or more anti-intracellular signaling molecule antibodies are an anti-pERKl/2 antibody, an anti-IkBa antibody, an anti- NF-KB antibody, an anti-p38 antibody, an anti-pAKT antibody, an anti-pCREB antibody, an anti-pLCK antibody, an anti-pPLC-y2 antibody, an anti-pS6 antibody, an anti-pSTATl antibody, an anti-pSTAT3 antibody, an anti-pSTAT5 antibody, an anti-Lamp2 antibody, an anti-EEAl antibody, or an anti-Rab5 antibody.
98. The method of claim 96, wherein said detecting a level of phosphorylation comprises detecting binding of said one or more anti-intracellular signaling molecule antibodies to said labeled ex vivo stimulated PBMC.
99. The method of claim 1, wherein said ex vivo stimulated PBMC is a permeabilized PBMC.
100. The method of claim 1, wherein said ex vivo stimulated PBMC is in a detection device.
101. The method of any one of claims 2 or 7, wherein said neurological disease treatment is a PLC-y2 pathway activator.
102. The method of any one of claims 2 or 7, wherein said neurological disease treatment is galantamine, rivastigmine, donepezil, memantine, levodopa, carbidopa, amantadine, a dopamine agonist, a monoamine oxidase B inhibitor, a Catechol-O- methyltransferase inhibitor, or an anticholinergic.
PCT/US2021/024539 2020-03-27 2021-03-27 Alzheimer's diagnostic method WO2021195602A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063001195P 2020-03-27 2020-03-27
US63/001,195 2020-03-27

Publications (1)

Publication Number Publication Date
WO2021195602A1 true WO2021195602A1 (en) 2021-09-30

Family

ID=77892698

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/024539 WO2021195602A1 (en) 2020-03-27 2021-03-27 Alzheimer's diagnostic method

Country Status (1)

Country Link
WO (1) WO2021195602A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130071860A1 (en) * 2010-02-24 2013-03-21 Matthew B. Hale Methods for autoimmune disease diagnosis, prognosis, and treatment`
WO2013050413A1 (en) * 2011-10-03 2013-04-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for obtaining a population of regulatory t cells
US20170248604A1 (en) * 2008-07-10 2017-08-31 Nodality, Inc. Methods for diagnosis, prognosis and methods of treatment

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170248604A1 (en) * 2008-07-10 2017-08-31 Nodality, Inc. Methods for diagnosis, prognosis and methods of treatment
US20130071860A1 (en) * 2010-02-24 2013-03-21 Matthew B. Hale Methods for autoimmune disease diagnosis, prognosis, and treatment`
WO2013050413A1 (en) * 2011-10-03 2013-04-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for obtaining a population of regulatory t cells

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PHONGPREECHA THANAPHONG, CHOLERTON BRENNA, MATA IGNACIO F., ZABETIAN CYRUS P., POSTON KATHLEEN L., AGHAEEPOUR NIMA, TIAN LU, QUINN: "Multivariate prediction of dementia in Parkinson’s disease", NPJ PARKINSON'S DISEASE, vol. 6, no. 1, 1 December 2020 (2020-12-01), XP055861298, DOI: 10.1038/s41531-020-00121-2 *
PHONGPREECHA THANAPHONG, FERNANDEZ ROSEMARY, MRDJEN DUNJA, CULOS ANTHONY, GAJERA CHANDRESH R, WAWRO ADAM M, STANLEY NATALIE, GAUD: "Single-cell peripheral immunoprofiling of Alzheimer's and Parkinson's diseases", SCI. ADV, 1 January 2020 (2020-01-01), XP055861301, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7688332/pdf/abd5575.pdf> [retrieved on 20211115] *

Similar Documents

Publication Publication Date Title
Butovsky et al. Targeting mi R‐155 restores abnormal microglia and attenuates disease in SOD 1 mice
Larochelle et al. Melanoma cell adhesion molecule–positive CD 8 T lymphocytes mediate central nervous system inflammation
US11034751B1 (en) Methods and compositions for treating cancer using serotonin receptor inhibitors
US20140193827A1 (en) Characterizing a glatiramer acetate related drug product
Singer The neurochemistry of Tourette syndrome
Mangalam et al. Absence of IFN-γ Increases Brain Pathology in Experimental Autoimmune Encephalomyelitis–Susceptible DRB1* 0301. DQ8 HLA Transgenic Mice through Secretion of Proinflammatory Cytokine IL-17 and Induction of Pathogenic Monocytes/Microglia into the Central Nervous System
US11147829B2 (en) Inhibition of expansion and function of pathogenic age-associated B cells and use for the prevention and treatment of autoimmune disease
Peligero-Cruz et al. IL18 signaling promotes homing of mature Tregs into the thymus
Boddupalli et al. Neuroinflammation in neuronopathic Gaucher disease: Role of microglia and NK cells, biomarkers, and response to substrate reduction therapy
Nuvolone et al. Cystatin F is a biomarker of prion pathogenesis in mice
JP7244423B2 (en) PCNT as a Target Protein for Treatment or Diagnosis of Cranial Nervous System Diseases
US20220091111A1 (en) Methods and compositions for diagnosing neurodegenerative disease
US20170003277A1 (en) Biological characterization of a glatiramer acetate related drug product using mammalian and human cells
Gillett et al. Interleukin 18 receptor 1 expression distinguishes patients with multiple sclerosis
EP2855516B1 (en) Inflammation-enabling polypeptides and uses thereof
WO2021195602A1 (en) Alzheimer&#39;s diagnostic method
US11208475B1 (en) Methods and compositions for treating inflammatory or autoimmune diseases or conditions using serotonin receptor activators
US10385395B2 (en) Diagnostic tools for response to 6-thiopurine therapy
TW201610169A (en) Biological characterization of a GLATIRAMER acetate related drug product using mammalian and human cells
AU4848899A (en) Methods and agents for the detection and modulation of cellular immunity to immune privileged antigens
Borna et al. FOXP3 deficiency, from the mechanisms of the disease to curative strategies
US20240131050A1 (en) Inhibition of expansion and function of pathogenic age-associated b cells and use for the prevention and treatment of autoimmune disease
US11059886B1 (en) Methods and compositions for treating inflammatory or autoimmune diseases or conditions using GRM8 activators
Williams et al. PINK1 is a target of T cell responses in Parkinson's disease
Dominguez-Pinilla et al. Extreme Phenotypes With Identical Mutations: Two Patients With Same Non-sense NHEJ1 Homozygous Mutation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21775734

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21775734

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 21775734

Country of ref document: EP

Kind code of ref document: A1