EP1259819A2 - Diagnostic de maladies affectives bipolaires (bad) et de depressions unipolaires - Google Patents

Diagnostic de maladies affectives bipolaires (bad) et de depressions unipolaires

Info

Publication number
EP1259819A2
EP1259819A2 EP01905998A EP01905998A EP1259819A2 EP 1259819 A2 EP1259819 A2 EP 1259819A2 EP 01905998 A EP01905998 A EP 01905998A EP 01905998 A EP01905998 A EP 01905998A EP 1259819 A2 EP1259819 A2 EP 1259819A2
Authority
EP
European Patent Office
Prior art keywords
dpi
bad
related polypeptide
cells
antibody
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP01905998A
Other languages
German (de)
English (en)
Inventor
Herath Mudiyanselage Athula Chandrasiri Herath
Rajesh Bhikhu Parekh
Christian Rohlff
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Oxford Glycosciences UK Ltd
Original Assignee
Oxford Glycosciences UK Ltd
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
Priority claimed from GB0004412A external-priority patent/GB0004412D0/en
Priority claimed from GB0030050A external-priority patent/GB0030050D0/en
Application filed by Oxford Glycosciences UK Ltd filed Critical Oxford Glycosciences UK Ltd
Publication of EP1259819A2 publication Critical patent/EP1259819A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • 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
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/30Psychoses; Psychiatry
    • G01N2800/304Mood disorders, e.g. bipolar, depression

Definitions

  • The; present invention relates to the identification of proteins and protein isoforms that are associated with Bipolar Affective Disorder (BAD) and Unipolar depression and its onset and development, and of genes encoding the same, and to their use for e.g., clinical screening, diagnosis, prognosis, therapy and prophylaxis, as well as for drug screening and drug development in the treatment of a neuropsychiatric disorder, including, by way of example and not of limitation, attention deficit disorder, a schizoaffective disorder, a bipolar affective disorder or unipolar affective disorder. For convenience, such disorders are referred to herein as BAD unless the context suggests otherwise.
  • BAD Bipolar Affective Disorder
  • BAD manic-depressive illnesses
  • BP bipolar mood disorder
  • manic-depressive illness is one of the most common, severe and often life threatening neuropsychiatric disorders.
  • Suicide is the cause of death in 10% to 20% of individuals with either bipolar or recurrent disorders, and the risks of suicide in bipolar disorder may be higher than those in unipolar depression (reviewed by Simpson and Jamison, J Clin Psychiatry 1999, 60, 53-56).
  • BAD is characterized by episodes of elevated mood(mania) and depression (Goodwin et al. 1990, Manic Depressive Illness, Oxford University Press, New York).
  • BP-1 severe bipolar affective (mood) disorder
  • SAD-M schizoaffective manic type
  • BP-1 and SAD-M are always associated with at least one full episode of mania and may include episodes of depression (lowered mood and disturbances in rhythmic behaviors like sleeping, eating, and sexual activity). Since BP-1 and SAD-M follow similar clinical courses and segregate together in- family studies (Rosenthal et al. 1980, Arch. General Psychiat. 37, 804-810; Levinson and Levitt, 1987, Am J Psychiat. 144, 415- 426;Goddwin et al. 1990, Manic Depressive Illness, Oxford University Press, New York)they are frequently difficult to distinguish from one another. BP-1 often also co- segregates in families with unipolar major depressive disorder (MDD), which has a broadly defined phenotype (Freimer and Reus, 1992, in The Molecular and
  • Rates of depression co-occurring with other medical conditions are as follows: myocardial infarction:20-40%, Parkinson's disease: 40%, Alzheimer's disease: 30-35%, stroke: 25-50%,cancer: 3-50%, HIV/AIDS: 10-20 %, rheumatoid arthritis: 12 %, diabetes mellitus: 14-18%, chronic pain: 30%, disabling tinnitus: 60%, end-stage renal disease: 5-22%and spinal cord injury: 37% (Goldman et al.
  • Major depression is a frequent diagnosis in patients evaluated for both cognitive and affective disorders and many depressed patients, in fact, are clinically characterized by cognitive impairments (Emery and Oxman, 1992, Am J Psychiatry, 149, 305-317).
  • Current therapeutic can be categorized into the following major classes of agents: mood stabilizers: lithium, divalproex, carbamazepine, lamotrigine; antidepressants: tricyclic antidepressants (eg.
  • CSF cerebrospinal fluid
  • DAPs disease associated proteins
  • the present invention provides methods and compositions for clinical screening, diagnosis, prognosis, therapy and prophylaxis of BAD, for monitoring the effectiveness of BAD treatment, for selecting participants in clinical trials, for identifying patients most likely to respond to a particular therapeutic treatment and for screening and development of drugs for treatment of BAD.
  • a first aspect of the invention provides methods for diagnosis of BAD that comprise analyzing a sample of CSF by two-dimensional electrophoresis to detect the presence or level of at least one Depression-Associated Feature (DF), e.g., one or more of the DFs disclosed herein, or any combination thereof.
  • DF Depression-Associated Feature
  • a second aspect of the invention provides methods for diagnosis of BAD that comprise detecting in a sample of CSF the presence or level of at least one Depression- Associated Protein Isoform (DPI), e.g., one or more of the DPIs disclosed herein or any combination thereof.
  • DPI Depression- Associated Protein Isoform
  • a third aspect of the invention provides antibodies, e.g., monoclonal and polyclonal capable of immunospecific binding to a DPI, e.g., a DPI disclosed herein.
  • a fourth aspect of the invention provides a preparation comprising an isolated DPI, i.e., a DPI free from proteins or protein isoforms having a significantly different isoelectric point or a significantly different apparent molecular weight from the DPI.
  • a fifth aspect of the invention provides methods of treating BAD, comprising administering to a subject a therapeutically effective amount of an agent that modulates (e.g., upregulates or downregulates) the expression or activity (e.g., enzymatic or binding activity), or both, of a DPI in subjects having BAD, in order to prevent or delay the onset or development of BAD, to prevent or delay the progression of BAD, or to ameliorate the symptoms of BAD.
  • an agent that modulates e.g., upregulates or downregulates
  • the expression or activity e.g., enzymatic or binding activity
  • a sixth aspect of the invention provides methods of screening for agents that modulate (e.g., upregulate or downregulate) a characteristic of, e.g., the expression or the enzymatic or binding activity, of a DPI, a DPI analog, or a DPI-related polypeptide.
  • DPI analog refers to a polypeptide that possesses similar or identical function(s) as a DPI but need not necessarily comprise an amino acid sequence that is similar or identical to the amino acid sequence of the DPI, or possess a structure that is similar or identical to that of the DPI.
  • an amino acid sequence of a polypeptide is "similar" to that of a DPI if it satisfies at leastone of the following criteria: (a) the polypeptide has an amino acid sequence that is at least 30% (more preferably, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least85%, at least 90%, at least 95% or at least 99%) identical to the amino acid sequence of the DPI; (b) the polypeptide is encoded by a nucleotide sequence that hybridizes under stringent conditions to a nucleotide sequence encoding at least 5 amino acid residues (more preferably, at least 10 amino acid residues, at least 15 amino acid residues, at least 20 amino acid residues, at least 25 amino acid residues, at least 40amino acid residues, at least 50 amino acid residues, at least 60 amino residues, at least 70 amino acid residues,
  • a polypeptide with "similar structure" to that of a DPI refers to a polypeptide that has a similar secondary, tertiary or quartemary structure as that of the DPI.
  • the structure of a polypeptide can determined by methods known to those skilled in the art, including but not limited to, X-ray crystallography, nuclear magnetic resonance, and crystallographic electron microscopy.
  • DPI fusion protein refers to a polypeptide that comprises (i) an amino acid sequence of a DPI, a DPI fragment, a DPI-related polypeptide or a fragment of a DPI-related polypeptide and (ii) an amino acid sequence of a heterologous polypeptide (i.e., a non-DPI, non-DPI fragment or non- DPI-related polypeptide).
  • DPI homolog refers to a polypeptide that comprises an amino acid sequence similar to that of a DPI but does not necessarily possess a similar or identical function as the DPI.
  • DPI ortholog refers to a non-human polypeptide that(i) comprises an amino acid sequence similar to that of a DPI and (ii) possesses a similar or identical function to that of the DPI.
  • DPI-related polypeptide refers to a DPI homolog, a DPI analog, an isoform of DPI, a DPI ortholog, or any combination thereof.
  • derivative refers to a polypeptide that comprises an amino acid sequence of a second polypeptide which has been altered by the introduction of amino acid residue substitutions, deletions or additions. The derivative polypeptide possess a similar or identical function as the second polypeptide.
  • fragment refers to a peptide or polypeptide comprising an amino acid sequence of at least 5 amino acid residues (preferably, at least 10 amino acid residues, at least 15 amino acid residues, at least 20 amino acid residues, at least 25 amino acid residues, at least 40 amino acid residues, at least 50amino acid residues, at least 60 amino residues, at least 70 amino acid residues, at least 80 amino acid residues, at least 90 amino acid residues, at least 100 amino acid residues, at least 125 amino acid residues, at least 150 amino acid residues, at leastl75 amino acid residues, at least 200 amino acid residues, or at least 250 amino acid residues) of the amino acid sequence of a second polypeptide.
  • the fragment of a DPI may or may not possess a functional activity of the a second polypeptide.
  • fold change includes “fold increase” and “fold decrease” and refers to the relative increase or decrease in abundance of an DF or the relative increase or decrease in expression or activity of a polypeptide (e.g. a DPI) in a first sample or sample set compared to a second sample (or sample set).
  • a DF or polypeptide fold change may be measured by any technique known to those of skill in the art, albeit the observed increase or decrease will vary depending upon the technique used.
  • fold change is determined herein as described in the Examples infra.
  • isoform refers to variants of a polypeptide that are encoded by the same gene, but that differ in their pi or MW, or both. Such isoforms can differ in their amino acid composition (e.g. as a result of alternative mRNA or premRNA processing, e.g. alternative splicing or limited proteolysis) and in addition, or in the.alternative, may arise from differential post-translational modification (e.g., glycosylation, acylation, phosphorylation).
  • amino acid composition e.g. as a result of alternative mRNA or premRNA processing, e.g. alternative splicing or limited proteolysis
  • differential post-translational modification e.g., glycosylation, acylation, phosphorylation
  • modulate when used herein in reference to expression or activity of a DPI or a DPI-related polypeptide refers to any change, e.g., upregulation or downregulation, of the expression or activity of the DPI or a DPI-related polypeptide. Based on the present disclosure, such modulation can be determined by assays known to those of skill in the art or described herein.
  • the percent identity of two amino acid sequences or of two nucleic acid sequences is determined by aligning the sequences for optimal comparison purposes(e.g., gaps can be introduced in the first sequence for best alignment with the sequence) and comparing the amino acid residues or nucleotides at corresponding positions.
  • the "best alignment” is an alignment of two sequences which results in the highest percent identity.
  • the determination of percent identity between two sequences can be accomplished using a mathematical algorithm known to those of skill in the art.
  • An example of a mathematical algorithm for comparing two sequences is the algorithm of Kariin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Kariin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877.
  • the NBLAST and XBLAST programs of Altschul, et al. (1990) J. Mol. Biol. 215:403-410 have incorporated such an algorithm.
  • Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402.
  • PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Id.).
  • Figure 1 is an image obtained from 2-dimensional electrophoresis of normal CSF, which has been annotated to identify twelve landmark features, designated CSFlto CSF12.
  • Figure 2 shows the amino acid sequence of DPI-45 and DPI-213 ( Figure 2B).
  • FIG. 3 shows tissue distribution of DPI 45 and DPI-213 mRNA.
  • Levels of mRNA in normal tissues were quantified by real time RT-PCR. mRNA levels are expressed as the number of copies per nanogram cDNA. Note the 50 times difference in scale between the left-hand part of the graph, containing brain-related samples, and the right-hand part of the graph, containing body samples.
  • the invention described in detail below provides methods and compositions for clinical screening, diagnosis and prognosis of BAD in a mammalian subject, for monitoring the results of BAD therapy, for identifying patients most likely to respond to a particular therapeutic treatment and for drug screening and drug development.
  • the invention also encompasses the administration of therapeutic compositions to a mammalian subject to treat or prevent BAD.
  • the mammalian subject may be a non- human mammal, but is preferably human, more preferably a human adult, i.e. a human subject at least 21 (more preferably at least 35, at least 50, at least 60, at least 70, or at least 80) years old.
  • the invention will be described with respect to the analysis of CSF samples.
  • the assays and techniques described below can be applied to other types of samples, including a body fluid (e.g. blood, serum, plasma, saliva or urine), a tissue sample from a subject at risk of having or developing BAD (e.g. a biopsy such as a brain biopsy) or homogenate thereof.
  • a body fluid e.g. blood, serum, plasma, saliva or urine
  • a tissue sample from a subject at risk of having or developing BAD e.g. a biopsy such as a brain biopsy
  • homogenate thereof e.g. a body fluid
  • the methods and compositions of the present invention are useful for screening, diagnosis and prognosis of a living subject, but may also be used for postmortem diagnosis in a subject, for example, to identify family members of the subject who are at risk of developing the same disease.
  • cerebrospinal fluid refers to the fluid that surrounds the bulk of the central nervous system, as described in Physiological Basis of Medical Practice (J.B. West, ed., Williams and Wilkins, Baltimore, MD 1985). CSF includes ventricular CSF and lumbar CSF.
  • serum refers to the supernatant fluid produced by clotting and centrifugal sedimentation of a blood sample.
  • plasma refers to the supernatant fluid produced by inhibition of clotting (for example, by citrate or EDTA) and centrifugal sedimentation of a blood sample.
  • blood as used herein includes serum and plasma.
  • two-dimensional electrophoresis is used to analyze CSF from a subject, preferably a living subject, in order to detect or quantify the expression of one or more Depression- Associated Features (DFs) for screening, prevention or diagnosis of BAD, to determine the prognosis of a subject having BAD, to monitor progression of BAD, to monitor the effectiveness of BAD therapy, for identifying patients most likely to respond to a particular therapeutic treatment, or for drug development.
  • DFs Depression- Associated Features
  • two-dimensional electrophoresis means a technique comprising isoelectric focusing, followed by denaturing electrophoresis; this generates a two-dimensional gel (2D-gel) containing a plurality of separated proteins.
  • the step of denaturing electrophoresis uses polyacrylamide electrophoresis in the presence of sodium dodecyl sulfate (SDS- PAGE).
  • SDS- PAGE sodium dodecyl sulfate
  • the Preferred Technology described in International Application No.97GB3307 (published as WO 98/23950) and in U.S. Patent No. 6,064,754, both filed December 1, 1997, each of which is incorporated herein by reference in its entirety with particular reference to the protocol at pages 23-35.
  • the Preferred Technology provides efficient, computer-assisted methods and apparatus for identifying, selecting and characterizing biomolecules (e.g. proteins, including glycoproteins) in a biological sample.
  • a two-dimensional array is generated by separating biomolecules on a two-dimensional gel according to their electrophoretic mobility and isoelectric point.
  • a computer-generated digital profile of the array is generated, representing the identity, apparent molecular weight, isoelectric point, and relative abundance of a plurality of biomolecules detected in the two-dimensional array, thereby permitting computer-mediated comparison of profiles from multiple biological samples, as well as computer aided excision of separated proteins of interest.
  • the Basiji thesis provides a phase-sensitive detection system for discriminating modulated fluorescence from baseline noise due to laser scatter or homogeneous fluorescence, but the scanner can also be operated in a non-phase-sensitive mode.
  • This phase-sensitive detection capability would increase the sensitivity of the instrument by an order of magnitude or more compared to conventional fluorescence imaging systems. The increased sensitivity would reduce the sample-preparation load on the upstream instruments while the enhanced image quality simplifies image analysis downstream in the process.
  • a more highly preferred scanner is the Apollo 2 scanner (Oxford Glycosciences, Oxford, UK), which is a modified version of the above described scanner.
  • the gel is transported through the scanner on a precision lead-screw drive system. This is preferable to laying the glass plate on the belt-driven system that is described in the Basiji thesis, as it provides a reproducible means of accurately transporting the gel past the imaging optics.
  • the gel is secured against three alignment stops that rigidly hold the glass plate in a known position. By doing this in conjunction with the above precision transport system, the absolute position of the gel can be predicted and recorded. This ensures that co-ordinates of each feature on the gel can be determined more accurately and communicated, if desired, to a cutting robot for excision of the feature.
  • the carrier that holds the gel has four integral fluorescent markers for use to correct the image geometry. These markers are a quality control feature that confirms that the scanning has been performed correctly.
  • the optical components of the Apollo . 2 scanner have been inverted. In the Apollo 2 scanner, the laser, mirror, waveguide and other optical components are above the glass plate being scanned.
  • the scanner described in the Basiji thesis has these components underneath.
  • the glass plate is mounted onto the scanner gel side down, so that the optical path remains through the glass plate. By doing this, any particles of gel that may break away from the glass plate will fall onto the base of the instrument rather than into the optics. This does not affect the functionality of the system, but increases its reliability.
  • the Apollo 3 scanner in which the signal output is digitized to the full 16-bit data without any peak saturation or without square root encoding of the signal.
  • a compensation algorithm has also been applied to correct for any variation in detection sensitivity along the path of the scanning beam. This variation is due to anomalies in the optics and differences in collection efficiency across the waveguide.
  • a calibration is performed using a perspex plate with an even fluorescence throughout.
  • the data received from a scan of this plate are used to determine the multiplication factors needed to increase the signal from each pixel level to a target level. These factors are then used in subsequent scans of gels to remove any internal optical variations.
  • feature refers to a spot detected in a 2D gel
  • DF Depression-Associated Feature
  • a feature (or a protein isoform of DPI, as defined infra) is "differentially present" in a first sample with respect to a second sample when a method for detecting the feature, isoform or DPI (e.g., 2D electrophoresis or an immunoassay) gives a different signal when applied to the first and second samples.
  • isoform or DPI e.g., 2D electrophoresis or an immunoassay
  • a feature, isoform or DPI is "increased" in the first sample with respect to the second if the method of detection indicates that the feature, isoform or DPI is more abundant in the first sample than in the second sample, or if the feature, isoform or DPI is detectable in the first sample and undetectable in the second sample.
  • a feature, isoform or DPI is "decreased" in the first sample with respect to the second if the method of detection indicates that the feature, isoform or DPI is less abundant in the first sample than in the second sample or if the feature, isoform or DPI is undetectable in the first sample and detectable in the second sample.
  • the relative abundance of a feature in two samples is determined in two steps.
  • the signal obtained upon detecting the feature in a sample is normalized by reference to a suitable background parameter, e.g., (a) to the total protein in the sample being analyzed (e.g., total protein loaded onto a gel); (b) to an
  • Expression Reference Feature i.e., a feature whose abundance is invariant, within the limits of variability of the Preferred Technology, in the population of subjects being examined, e.g. the ERFs disclosed below, or (c) more preferably to the total signal detected from all proteins in the sample.
  • the normalized signal for the feature in one sample or sample set is compared with the normalized signal for the same feature in another sample or sample set in order to identify features that are "differentially present" in the first sample (or sample set) with respect to the second.
  • the DFs disclosed herein have been identified by comparing CSF samples from subjects having BAD against CSF samples from subjects free from BAD.
  • Subjects free from BAD include subjects with no known disease or condition (normal subjects) and subjects with BAD.
  • the first group consists of DFs that are decreased in the
  • DFs can be described by apparent molecular weight (MW) and isoelectric point(pI) as provided in Table I.
  • the second group consists of DFs that are increased in the CSF of subjects having BAD as compared with the CSF of subjects free from BAD.
  • These DFs can be described by apparent molecular weight (MW) and isoelectric point (pi) as provided in Table II.
  • the signal obtained upon analyzing CSF from subjects having BAD relative to the signal obtained upon analyzing CSF from subjects free from BAD will depend upon the particular analytical protocol and detection technique that is used. Accordingly, the present invention contemplates that each laboratory will, based on the present description, establish a reference range for each DF in subjects free from BAD according to the analytical protocol and detection technique in use, as is conventional in the diagnostic art.
  • at least one positive control CSF sample from a subject known to have BAD or at least one negative control CSF sample from a subject known to be free from BAD (and more preferably both positive and negative control samples) are included in each batch of test samples analyzed.
  • the level of expression of a feature is determined relative to a background value, which is defined as the level of signal obtained from a proximal region of the image that (a) is equivalent in area to the particular feature in question; and (b) contains no discernable protein feature.
  • the reference range depending upon the method of detection used and the conditions under which detection is carried out, can include no feature or isoform present, or non-detectable levels of feature or isoform present. Proteins described by pi and MW provided in Tables I and II can be identified by searching 2D-PAGE databases with those pi and MW values.
  • databases typically provide interactive 2D gels images for a given set of sample and preparation protocol, and the skilled artisan can obtain information relevant to a given feature by pointing and clicking the appropriate section of the image.
  • the signal associated with an DF in the CSF of a subject is normalized with reference to one or more ERFs detected in the same 2D gel.
  • ERFs may readily be determined by comparing different samples using the Preferred Technology. Suitable ERFs include (but are not limited to) that described in the following table.
  • the measured MW and pi of a given feature or protein isoform will vary to some extent depending on the precise protocol used for each step of the 2D electrophoresis and for landmark matching.
  • the terms "MW” and "pi" are defined, respectively, to mean the apparent molecular weight in Daltons and the apparent isoelectric point of a feature or protein isoform as measured in exact accordance with the Reference Protocol identified, in Section 6 below.
  • variation in the measured mean pi of a DF or DPI is typically less than 3% and variation in the measured mean MW of a DF or DPI is typically less than 5%.
  • DFs can be used for detection, prognosis, diagnosis, or monitoring of BAD or for drug development.
  • CSF from a subject e.g., a subject suspected of having BAD
  • 2D electrophoresis for quantitative detection of one or more of the following DFs: DF-3, DF-4, DF-5, DF-6, DF-7, DF-8, DF-9, DF-10, DF-11, DF-12, DF-13, DF-14, DF-15, DF-16, DF-17, DF-18, DF-19, DF-20, DF-21, DF-22, DF-23, DF-24, DF-25, DF-26, DF-27, DF-28, DF-29, DF-30, DF-31, DF-32, DF-33, DF-34, DF-35, DF-36, DF-37, DF-39, DF-40, DF-41, DF-42, DF-43,
  • CSF from a subject is analyzed by 2D electrophoresis for quantitative detection of one or more of the following DFs: DF-76, DF-77, DF-78, DF-82, DF-84, DF-86, DF-87, DF-94, DF-95, DF-96, DF-97, DF-98, DF-99, DF-100, DF-101, DF-102, DF-103, DF-104, DF-105, DF-106, DF-107, DF- 108, DF-109, DF-110, DF-111, DF-112, DF-113, DF-115, DF-117, DF-118, DF-120, DF-121, DF-123, DF-124, DF-125, DF-126, DF-127, DF-130, DF-131, DF-132, DF- 134, DF-135, DF-137, DF
  • CSF from a subject is analyzed by 2D electrophoresis for quantitative detection of (a) one or more DFs or any combination of them, whose decreased abundance indicates the presence of BAD, i.e., DF-3, DF-4, DF-5, DF-6, DF-7, DF-8, DF-9, DF-10, DF-11, DF-12, DF-13, DF-14, DF-15, DF-16, DF-17, DF-18, DF-19, DF-20, DF-21, DF-22, DF-23, DF-24, DF-25, DF-26, DF-27, DF-28, DF-29, DF-30, DF-31, DF-32, DF-33, DF-34, DF-
  • CSF from a subject is analyzed by 2D electrophoresis for quantitative detection of one or more of the following DFs: DF-3, DF-4, DF-5, DF-6, DF-7, DF-8, DF-9, DF-10, DF-11, DF-12, DF-13, DF-14, DF-15, DF-16, DF-17, DF-18, DF-19, DF-20, DF-21, DF-22, DF-23, DF-24, DF-25, DF-26, DF-27, DF-28, DF-29, DF-30, DF-31, DF-32, DF-33, DF-34, DF-35, DF-36, DF-37, DF-39, DF-40, DF-41, DF-42, DF-43, DF-44, DF-47, DF-48, DF-51, DF-52, DF-55, DF-56, DF-58,
  • a decrease in one or more DF/ERF ratios in a test sample relative to the DF/ERF ratios in a control sample or a reference range indicates the presence of BAD; DF-3, DF-4, DF-5, DF-6, DF-7, DF-8, DF-9, DF-10, DF-11, DF-12, DF-13, DF-14, DF-15, DF-16, DF-17, DF-18, DF-19, DF-20, DF-21, DF-22, DF-23, DF-24, DF-25, DF-26, DF-27, DF-28, DF-29, DF-30, DF-31, DF-32, DF-33, DF-34, DF-35, DF-36, DF-37, DF-39, DF-40, DF-41, DF-42, DF-43, DF-44, DF-47, DF-48, DF-51, DF-52, DF
  • an increase in one or more DF/ERF ratios in a test sample relative to the DF/ERF ratios' in a control sample or a reference range indicates the presence of BAD; DF-76, DF- 77, DF-78, DF-82, DF-84, DF-86, DF-87, DF-94, DF-95, DF-96, DF-97, DF-98, DF- 99, DF-100, DF-101, DF-102, DF-103, DF-104, DF-105, DF-106, DF-107, DF-108, DF-109, DF-110, DF-111, DF-112, DE-113, DF-115, DF-117, DF-118, DF-120, DF- 121, DF-123, DF-124, DF-125, DF-126, DF-127, DF-130, DF-131, DF-132, DF-134
  • 2D electrophoresis for quantitative detection of (a) one or more DFs, or any combination of them, whose decreased DF/ERF ratio(s) in a test sample relative to the DF/ERF ratio(s) in a control sample indicates the presence of BAD, i.e., DF-3, DF-4, DF-5, DF-6, DF-7, DF-8, DF-9, DF-10, DF-11, DF-12, DF-13, DF-14, DF-15, DF-16, DF-17, DF-18, DF-19, DF-20, DF-21, DF-22, DF-23, DF-24, DF-25, DF-26, DF-27, DF-28, DF-29, DF-30, DF-31, DF-32, DF-33, DF-34, DF-35, DF-36, DF-37, DF-39, DF-40, DF-41, DF-42, DF-43, DF-44
  • CSF from a subject is analyzed for quantitative detection of a plurality of DFs.
  • DPIs Depression-Associated Protein Isoforms
  • CSF from a subject is analyzed for quantitative detection of one or more Depression- Associated Protein Isoforms (DPIs) for screening or diagnosis of BAD, to determine the prognosis of a subject having BAD, to monitor the effectiveness of BAD therapy, for identifying patients most likely to respond to a particular therapeutic treatment or for drug development.
  • DPIs Depression-Associated Protein Isoforms
  • a given protein may be expressed as variants (isoforms) that differ in their amino acid composition (e.g. as a result of ⁇ alternative mRNA or premRNA processing, e.g.
  • differential post-translational modification e.g., glycosylation, phosphorylation, acylation
  • differential post-translational modification e.g., glycosylation, phosphorylation, acylation
  • differential presence of a protein isoform does not require differential expression of the gene encoding the protein in question.
  • the term "Depression- Associated Protein Isoform” refers to a protein isoform that is differentially present in CSF from a subject having BAD compared with CSF from a subject free from BAD.
  • DPIs Two groups of DPIs have been identified by amino acid sequencing of DFs. DPIs were isolated, subjected to proteolysis, and analyzed by mass spectrometry using the methods and apparatus of the Preferred Technology. One skilled in the art can identify sequence information from proteins analyzed by mass spectrometry and/or tandem mass spectrometry using various spectral interpretation methods and database searching tools. Examples of some of these methods and tools can be found at the Swiss Institute of Bioinformatics web site at http://www.expasy.ch/, and the European Molecular Biology Laboratory web site at www.mann.embl- heidelberg.de/Services/PeptideSearch . Identification of BPIs was performed primarily using the SEQUEST search program (Eng et al., 1994, J. Am.
  • the first group consists of DPIs that are decreased in the CSF of subjects having BAD as compared with the CSF of subjects free from BAD, where the differential presence is significant.
  • the amino acid sequences of tryptic digest peptides of these DPIs identified by tandem mass spectrometry and database searching as described in the Examples, infra are listed in Table IV, in addition to their corresponding pis and MWs.
  • DPI- 45 and DPI-213 the partial sequence information for these DPIs derived from tandem mass spectrometry was not found to be described in any known public database. These DPIs are listed as 'NOVEL' in Table IV, and further described below.
  • the second group consists of DPIs that are increased in the CSF of subjects having , BAD as compared with the CSF of subjects free from BAD, where the differential presence is significant.
  • the amino acid sequences of tryptic digest peptides of these DPIs identified by tandem mass spectrometry and database searching as described in the Examples, infra are listed in Table V, in addition to their corresponding pis and MWs.
  • the DPI is a protein comprising a peptide sequence described for that DPI (preferably comprising a plurality of, more preferably all of, the peptide sequences described for that DPI) and has a pi of about the value stated for that DPI (preferably within about 10%, more preferably within about 5% still more preferably within about 1% of the stated value) and has a MW of about the value stated for that DPI (preferably within about 10%, more preferably within about 5%, still more preferably within about 1% of the stated value).
  • Proteins comprising the peptide sequences provided in Table IV and V can be identified by searching sequence databases with those peptides using search tools known to those skilled in the art. Examples of search algorithm tools that can be used to identify proteins from peptide sequences include:
  • BLAST Basic Local Alignment Search Tool
  • BLASTP can be used to search a protein sequence against a protein database.
  • TBLASTN can be used to search a Protein Sequence against a
  • the nr protein database maintained at the National Center for Biotechnology Information (NCBI) (http://www.ncbi.nlm.nih.gov). The nr protein database is compiled of entries from various sources including SwissProt, SwissProt updates, PIR, and PDB. The BLAST resource is available for sequence searching.
  • the PIR-International Protein Sequence Database maintained by the Protein Information Resource (PIR), in collaboration with the Kunststoff Information Center for Protein Sequences (MIPS) and the Japanese International Protein Sequence Database (JIPID).
  • the Protein Identification Resource (PIR) is a division of the National Biomedical Research Foundation (NBRF) which is affiliated with Georgetown University Medical Center and can be found at http://www nbrf.georgetown.edu/pir/searchdb.html.
  • the database can be searched using BLAST and FASTA search algorithm tools. .
  • the Protein Data Bank maintained by Brookhaven National Laboratory (Long Island, New York, USA) which can be found at http://www.rcsb.org/pdb/.
  • the FASTA resource is available at this website for sequence searching.
  • CSF from a subject is analyzed for quantitative detection of one or more of the following DPIs: DPI-2, DPI-3, DPI-4, DPI-5, DPI-6, DPI-7, DPI-8, DPI-9, DPI-10, DPI-11, DPI-12, DPI-13, DPI-14, DPI-15, DPI-17, DPI-18, - DPI-19, DPI-20, DPI-21, DPI-22, DPI-23, DPI-24, DPI-25, DPI-29, DPI-30, DPI-34, DPI-35, DPI-37, DPI-38, DPI-39, DPI-44, DPI-45, DPI-47, DPI-49, DPI-50, DPI-51, DPI-52, DPI-103, DPI-104, DPI-105, DPI-106, DPI-107, DPI-108, DPI-109, DPI- 110, DPI-111, DPI-113, DPI-115, DPI-116, DPI-139,
  • CSF from a subject is analyzed for quantitative detection of one or more of the following DPIs: DPI-57, DPI-58, DPI-59, DPI-60, DPI-65, DPI-66, DPI-67, DPI-69, DPI-71, DPI-72, DPI-73, DPI-76, DPI-78, DPI-79, DPI-87, DPI-88, DPI-89, DPI-90, DPI-92, DPI-93, DPI-96, DPI-119, DPI- 120, DPI-121, DPI-123, DPI-124, DPI-127, DPI-128, DPI-129, DPI-135, DPI-147, DPI-151, DPI-152, DPI-154, DPI-155, DPI-159, DPI-160, DPI-161, DPI-162, DPI- 163, DPI-164, DPI-165, DPI-166, DPI-167, DPI-168, DPH69, DPI-57, DPI
  • CSF from a subject is analyzed for quantitative detection of (a) one or more DPIs, or any combination of them, whose decreased abundance indicates the presence of BAD, i.e., DPI-2, DPI-3, DPI-4, DPI-5, DPI-6, DPI-7, DPI-8, DPI-9, DPI-10, DPI-11, DPI-12, DPI-13, DPI-14, DPI-15, DPI-17, DPI-18, DPI-19, DPI-20, DPI-21, DPI-22, DPI-23, DPI-24, DPI-25, DPI-29, DPI-30, DPI-34, DPI-35, DPI-37, DPI-38, DPI-39, DPI-44, DPI-45, DPI-47, DPI-49, DPI-50, DPI-51, DPI-52, DPI-103, DPI-104, DPI-105, DPI-106, DPI-107, DPI-108, DPI-109, DPI-110, DPI
  • CSF from a subject is analyzed for quantitative detection of one or more DPIs and one or more previously known biomarkers of BAD (e.g., candidate markers such as hypersensitive platelet glutamate receptors (Berk et , al. Int Clin Psychopharmacol 1999 14, 119-22)).
  • candidate markers such as hypersensitive platelet glutamate receptors (Berk et , al. Int Clin Psychopharmacol 1999 14, 119-22)
  • the abundance of each DPI and known biomarker relative to a control or reference range indicates whether a subject has BAD.
  • ERPIs can be identified by partial amino acid sequencing of ERFs, which are described above, using the methods and apparatus of the Preferred Technology.
  • the partial amino acid sequences of an ERPI, and the known proteins to which it is homologous is presented in Table VI.
  • the DPIs described herein include previously unknown proteins, as well as isoforms of known proteins where the isoforms were not previously known to be associated with BAD.
  • the present invention additionally provides: (a) a preparation comprising the isolated DPI; (b) a preparation comprising one or more fragments of the DPI; and (c) antibodies that bind to said DPI, to said fragments, or both to said DPI and to said fragments.
  • a DPI is "isolated" when it is present in a preparation that is substantially free of contaminating proteins, i.e., a preparation in which less than 10% (preferably less than 5%, more preferably less than 1%) of the total protein present is contaminating protein(s).
  • a contaminating protein is a protein or protein isoform having a significantly different pi or MW from those of the isolated DPI, as determined by 2D electrophoresis.
  • a "significantly different" pl or MW is one that permits the contaminating protein to be resolved from the DPI on 2D electrophoresis, performed according to the Reference Protocol.
  • an isolated protein comprising a peptide with the amino acid sequence identified in Table IV or V for a DPI, said protein having a pi and MW within 10% (preferably within 5%, more preferably within 1%) of the values identified in Table IV or V for that DPI.
  • the DPIs of the invention can be qualitatively or quantitatively detected by any method known to those skilled in the art, including but not limited to the Preferred Technology described herein, kinase assays, enzyme assays, binding assays and other functional assays, immunoassays, and western blotting.
  • the DPIs are separated on a 2-D gel by virtue of their MWs and pis and visualized by staining the gel.
  • the DPIs are stained with a fluorescent dye and imaged with a fluorescence scanner. Sypro Red (Molecular Probes, Inc., Eugene, Oregon) is a suitable dye for this purpose.
  • a preferred fluorescent dye is Pyridinium, 4-[2-[4- (dipentylamino)-2-trifluoromethylphenyl] ethenyl]-l-(sulfobutyl)-, inner salt. See U.S. Application No. 09/412,168, filed on October 5, 1999, which is incorporated herein by reference in its entirety.
  • DPIs can be detected in an immunoassay.
  • an immunoassay is performed by contacting a sample from a subject to be tested with an anti-DPI antibody under conditions such that immunospecific binding can occur if the DPI is present, and detecting or measuring the amount of any immunospecific binding by the antibody.
  • Anti-DPI antibodies can be produced by the methods and techniques taught herein; examples of such antibodies known in the art are set forth in Table VII. These antibodies shown in Table VII are already known to bind to the protein of which the DPI is itself a family member.
  • the anti-DPI antibody preferentially binds to the DPI rather than to other isoforms of the same protein.
  • the anti-DPI antibody binds to the DPI with at least 2-fold greater affinity, more preferably at least 5 -fold greater affinity, still more preferably at least 10-fold greater affinity, than to said other isoforms of the same protein.
  • DPIs can be transferred from the gel to a suitable membrane (e.g. a PVDF membrane) and subsequently probed in suitable assays that include, without limitation, competitive and non-competitive assay systems using techniques such as western blots and "sandwich" immunoassays using anti-DPI antibodies as described herein, e.g., the antibodies identified in Table VII, or others raised against the DPIs of interest.
  • the immunoblots can be used to identify those anti-DPI antibodies displaying the selectivity required to immuno-specifically differentiate a DPI from other isoforms encoded by the same gene.
  • binding of antibody in tissue sections can be used to detect aberrant DPI localization or an aberrant level of one or more DPIs.
  • antibody to a DPI can be used to assay a tissue sample (e.g., a brain biopsy) from a subject for the level of the DPI where an aberrant level of DPI is indicative of BAD.
  • tissue sample e.g., a brain biopsy
  • an aberrant level means a level that is increased or decreased compared with the level in a subject free from BAD or a reference level. If desired, the comparison can be performed with a matched sample from the same subject, taken from a portion of the body not affected by BAD.
  • any suitable immunoassay can be used, including, without limitation, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement- fixation assays, immunoradiometric assays, fluorescent immunoassays and protein A immunoassays.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement- fixation assays, immunoradiometric assays, fluorescent immunoas
  • a DPI can be detected in a fluid sample (e.g., CSF, blood, urine, or tissue homogenate) by means of a two-step sandwich assay.
  • a capture reagent e.g., an anti-DPI antibody
  • the capture reagent can optionally be immobilized on a solid phase.
  • a directly or indirectly labeled detection reagent is used to detect the captured DPI.
  • the detection reagent is a lectin.
  • any lectin can be used for this purpose that preferentially binds to the DPI rather than to other isoforms that have the same core protein as the DPI or to other proteins that share the antigenic determinant recognized by the antibody.
  • the chosen lectin binds to the DPI with at least 2-fold greater affinity, more preferably at least 5-fold greater affinity, still more preferably at least 10- fold greater affinity, than to said other isoforms that have the same core protein as the DPI or to said other proteins that share the antigenic determinant recognized by the antibody.
  • a lectin that is suitable for detecting a given DPI can readily be identified by methods well known in the art, for instance upon testing one or more lectins enumerated in Table I on pages 158-159 of Sumar et al., Lectins as Indicators of Disease- Associated Glycoforms, In: Gabius H-J & Gabius S (eds.), 1993, Lectins and Glycobiology, at pp. 158-174 (which is incorporated herein by reference in its entirety).
  • Lectins with the desired oligosaccharide specificity can be identified, for example, by their ability to detect the DPI in a 2D gel, in a replica of a 2D gel following _dnsfer to a suitable solid substrate such as a nitrocellulose membrane, or in a two-step assay following capture by an antibody.
  • the detection reagent is an antibody, e.g., an antibody that immunospecifically detects other post- translational modifications, such as an antibody that immunospecifically binds to phosphorylated amino acids.
  • antibodies examples include those that bind to phosphotyrosine (BD Transduction Laboratories, catalog nos.: P11230-050/P11230- 150; P11120; P38820; P39020), those that bind to phosphoserine (Zymed Laboratories Inc., South San Francisco, CA, catalog no. 61-8100) and those that bind to phosphothreonine
  • a gene encoding a DPI, a related gene, or related nucleic acid sequences or subsequences, including complementary sequences can also be used in hybridization assays.
  • a nucleotide encoding a DPI, or subsequences thereof comprising at least 8 nucleotides, preferably at least 12 nucleotides, and most preferably at least 15 nucleotides can be used as a hybridization probe.
  • Hybridization assays can be used for detection, prognosis, diagnosis, or monitoring of conditions, disorders, or disease states, associated with aberrant expression of genes encoding DPIs, or for differential diagnosis of subjects with signs or symptoms suggestive of BAD.
  • such a hybridization assay can be carried out by a method comprising contacting a subject's sample containing nucleic acid with a nucleic acid probe capable of hybridizing to a DNA or RNA that encodes a DPI, under conditions such that hybridization can occur, and detecting or measuring any resulting hybridization.
  • Nucleotides can be used for therapy of subjects having BAD, as described below.
  • the methods and compositions for clinical screening, diagnosis and prognosis of BAD in a mammalian subject may be diagnostic of BAD or indicative of BAD. Diagnostic methods and compositions are based on Depression- Associated Features (DFs) and Depression- Associated Protein Isoforms (DPIs) which are specifically and particularly associated with BAD and are generally not associated with other diseases or conditions. Such diagnostic DFs or DPIs, which are specifically associated with BAD, are useful in screening, diagnosis and prognosis as indicators of BAD.
  • the administration of therapeutic compositions which are directed against or lead to modulation of diagnostic markers may have therapeutic value particularly in BAD.
  • Indicative methods and compositions are based on Depression- Associated Features (DFs) and Depression- Associated Protein Isoforms (DPIs) which are associated with BAD but may not be specific only for BAD, and may be associated with one or more other diseases or conditions.
  • Such indicative DFs or DPIs, which are associated with BAD, but not only with BAD, are useful in screening, diagnosis and prognosis as indicators of BAD.
  • Indicative methods and compositions are particularly useful in the initial or general screening, diagnosis and prognosis of an individual subject, whereby a first indication of a subset of conditions or diseases, including BAD, is thereby provided.
  • Additional assessment utilizing diagnostic or particular BAD DFs or DPIs may then be undertaken to provide specific, diagnostic screening, diagnosis and prognosis of the individual subject.
  • the administration of therapeutic compositions which are directed against or lead to modulation of indicative markers may have therapeutic value in BAD and other disorders as well, or may be useful therapeutically in more than one disease or condition.
  • a diagnostic marker changes (increases, decreases or otherwise alters form or character) significantly in only a single disease or condition or in only a small number of conditions, particularly in related conditions.
  • One such diagnostic marker, DF-60 is provided below in Table VIII.
  • An indicative marker changes (increases, decreases or otherwise alters form or character) significantly in more than one condition, particularly in BAD and one or more other distinct diseases or conditions.
  • One such indicative marker, DF-155 is found to increase in BAD and is provided in Table LX.
  • This same marker, identified or characterised by the same pi and MW, is noted as SF-255 as similarly found to be increased in Schizophrenia.
  • the DF-155/SF-255 marker is therefore indicative of Depression and/or Schizophrenia.
  • kits comprising an anti-DPI antibody.
  • a kit may optionally comprise one or more of the following: (1) instructions for using the anti-DPI antibody for diagnosis, prognosis, therapeutic monitoring or any combination of these applications; (2) a labeled binding partner to the antibody; (3) a solid phase (such as a reagent strip) upon which the anti-DPI antibody is immobilized; and (4) a label or insert indicating regulatory approval for diagnostic, prognostic or therapeutic use or any combination thereof.
  • the anti-DPI antibody itself can be labeled with a detectable marker, e.g., a chemiluminescent, enzymatic, fluorescent, or radioactive moiety.
  • kits comprising a nucleic acid probe capable of hybridizing to RNA encoding a DPI.
  • a kit comprises in one or more containers a pair of primers (e.g., each in the size range of 6-30 nucleotides, more preferably 10-30 nucleotides and still more preferably 10-20 nucleotides) that under appropriate reaction conditions can prime amplification of at least a portion of a nucleic acid encoding a DPI, such as by poiymerase chain reaction (see, e.g., Innis et al., 1990, PCR Protocols, Academic Press, Inc., San Diego, CA), ligase chain reaction (see EP 320,308) use of Q ⁇ replicase, cyclic probe reaction, or other methods known in the art.
  • poiymerase chain reaction see, e.g., Innis et al., 1990, PCR Protocols, Academic Press, Inc., San Diego, CA
  • ligase chain reaction see EP 320,
  • Kits are also provided which allow for the detection of a plurality of DPIs or a plurality of nucleic acids each encoding a DPI.
  • a kit can optionally further comprise a predetermined amount of an isolated DPI protein or a nucleic acid encoding a DPI, e.g., for use as a standard or control.
  • the uni-variate differential analysis tools are useful in identifying individual DFs or DPIs that are diagnostically associated with BAD or in identifying individual DPIs that regulate the disease process.
  • the disease process is associated with a combination of DFs or DPIs (and to be regulated by a combination of DPIs), rather than individual DFs and DPIs in isolation.
  • the strategies for discovering such combinations of DFs and DPIs differ from those for discovering individual DFs and DPIs. In such cases, each individual DF and DPI can be regarded as one variable and the disease can be regarded as a joint, multi-variate effect caused by interaction of these variables.
  • the first step is to identify a collection of DFs or DPIs that individually show significant association with BAD.
  • the association between the identified DFs or DPIs and BAD need not be as highly significant as is desirable when an individual DF or DPI is used as a diagnostic. Any of the tests discussed above (fold changes, wilcoxon rank sum test, etc.) can be used at this stage.
  • a sophisticated multi-variate analysis capable of identifying clusters can then be used to estimate the significant multivariate associations with BAD.
  • LDA Linear Discriminant Analysis
  • DFs or DPIs variables
  • BAD Linear Discriminant Analysis
  • a set of weights is associated with each variable (i.e., DF or DPI) so that the linear combination of weights and the measured values of the variables can identify the disease state by discriminating between subjects having BAD and subjects free from BAD.
  • Enhancements to the LDA allow stepwise inclusion (or removal) of variables to optimize the discriminant power of the model.
  • the result of the LDA is therefore a cluster of DFs or DPIs which can be used for diagnosis, prognosis, therapy or drug development.
  • LDA Flexible Discriminant Analysis
  • results of the discriminant analysis can be verified by post-hoc tests and also by repeating the analysis using alternative techniques such as classification trees.
  • a further category of DFs or DPIs can be identified by qualitative measures by comparing the percentage feature presence of an DF or DPI of one group of samples (e.g., samples from diseased subjects) with the percentage feature presence of an DF or DPI in another group of samples (e.g., samples from control subjects).
  • the "percentage feature presence" of an DF or DPI is the percentage of samples in a group of samples in which the DF or DPI is detectable by the detection method of choice.
  • the percentage feature presence of that DF in that sample group is 95 percent. If only 5 percent of samples from non-diseased subjects have detectable levels of the same DF, detection of that DF in the sample of a subject would suggest that it is likely that the subject suffers from BAD.
  • the diagnostic methods and compositions of the present invention can assist in monitoring a clinical study, e.g. to evaluate drugs for therapy of BAD.
  • candidate molecules are tested for their ability to restore DF or DPI levels in a subject having BAD to levels found in subjects free from BAD or, in a treated subject (e.g. after treatment with mood stabilizers: lithium, divalproex, carbamazepine, lamotrigine; antidepressants: tricyclic antidepressants (e.g.
  • the methods and compositions of the present invention are used to screen candidates for a clinical study to identify individuals having BAD; such individuals can then be excluded from the study or can be placed in a separate cohort for treatment or analysis. If desired, the candidates can concurrently be screened to identify individuals with attention deficit disorder, a schizoaffective disorder or a unipolar affective disorder.
  • the invention provides isolated mammalian DPIs, preferably human DPIs, and fragments thereof which comprise an antigenic determinant (i.e., can be recognized by an antibody) or which are otherwise functionally active, as well as nucleic acid sequences encoding the foregoing.
  • "Functionally active” as used herein refers to material displaying one or more functional activities associated with a full-length (wild- type) DPI, e.g., binding to a DPI substrate or DPI binding partner, antigenicity (binding to an anti-DPI antibody), immunogenicity, enzymatic activity and the like.
  • the invention provides fragments of a DPI comprising at least 5 amino acids, at least 10 amino acids, at least 50 amino acids, or at least 75 amino acids. Fragments lacking some or all of the regions of a DPI are also provided, as are proteins (e.g., fusion proteins) comprising such fragments. Nucleic acids encoding the foregoing are provided.
  • the gene product can be analyzed. This is achieved by assays based on the physical or functional properties of the product, including radioactive labeling of the product followed by analysis by gel electrophoresis, immunoassay, etc.
  • the DPIs identified herein can be isolated and purified by standard methods including chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • chromatography e.g., ion exchange, affinity, and sizing column chromatography
  • centrifugation e.g., centrifugation
  • differential solubility e.g., differential solubility
  • the entire amino acid sequence of the DPI can be deduced from the nucleotide sequence of the gene coding region contained in the recombinant nucleic acid.
  • the protein can be synthesized by standard chemical methods known in the art (e.g., see Hunkapiller et al., 1984, Nature 310:105-111).
  • native DPIs can be purified from natural sources, by standard methods such as those described above (e.g., immunoaffinity purification).
  • DPIs are isolated by the Preferred Technology described supra.
  • a narrow-range "zoom gel” having a pH range of 2 pH units or less is preferred for the isoelectric step, according to the method described in Westermeier, 1993, Electrophoresis in Practice (VCH, Weinheim, Germany), pp. 197-209 (which is incorporated herein by reference in its entirety); this modification permits a larger quantity of a target protein to be loaded onto the gel, and thereby increases the quantity of isolated DPI that can be recovered from the gel.
  • the Preferred Technology typically provides up to 100 ng, and can provide up to 1000 ng, of an isolated DPI in a single run.
  • a zoom gel can be used in any separation strategy which employs gel isoelectric focusing.
  • the invention thus provides an isolated DPI, an isolated DPI-related polypeptide, and an isolated derivative or fragment of a DPI or a DPI-related polypeptide; any of the foregoing can be produced by recombinant DNA techniques or by chemical synthetic methods.
  • nucleotide sequences of the present invention including DNA and RNA, and comprising a sequence encoding a DPI or a fragment thereof, or a DPI-related polypeptide, may be synthesized using methods known in the art, such as using conventional chemical approaches or poiymerase chain reaction (PCR) amplification.
  • PCR poiymerase chain reaction
  • the nucleotide sequences of the present invention also permit the identification and cloning of the gene encoding a DPI homolog or DPI ortholog including, for example, by screening cDNA libraries, genomic libraries or expression libraries.
  • oligonucleotides can be designed for all DPI peptide fragments identified as part of the same protein.
  • PCR reactions under a variety of conditions can be performed with relevant cDNA and genomic DNAs (e.g., from brain tissue or from cells of the immune system) from one or more species.
  • vectorette reactions can be performed on any available cDNA and genomic DNA using the oligonucleotides (which preferably are nested) as above.
  • Vectorette PCR is a method that enables the amplification of specific DNA fragments in situations where the sequence of only one primer is known.
  • Vectorette PCR may pe performed with probes that are, for example, anchored degenerate oligonucleotides (or most likely oligonucleotides) coding for DPI peptide fragments, using as a template a genomic library or cDNA library pools.
  • Anchored degenerate oligonucleotides can be designed for all DPI peptide fragments. These oligonucleotides may be labelled and hybridized to filters containing cDNA and genomic DNA libraries. Oligonucleotides to different peptides from the same protein will often identify the same members of the library.
  • the cDNA and genomic DNA libraries may be obtained from any suitable or desired mammalian species, for example from humans. Nucleotide sequences comprising a nucleotide sequence encoding a DPI or
  • DPI fragment of the present invention are useful for their ability to hybridize selectively with complementary stretches of genes encoding other proteins.
  • a variety of hybridization conditions may be employed to obtain nucleotide sequences at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99% identical, or 100% identical, to the sequence of a nucleotide encoding a DPI.
  • relatively stringent conditions are used to form the duplexes, such as low salt or high temperature conditions.
  • “highly stringent conditions” means hybridization to filter-bound DNA in 0.5 M NaHPO4, 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65 °C, and washing in O.lxSSC/0.1% SDS at 68 °C (Ausubel F.M. et al., eds., 1989, Current Protocols in Molecular Biology, Vol. I, Green Publishing Associates, Inc., and John Wiley & Sons, Inc., New York, at p.
  • hybridization conditions For some applications, less stringent conditions for duplex formation are required. As used herein "moderately stringent conditions” means washing in 0.2xSSC/0.1% SDS at 42 °C (Ausubel et al., 1989, supra). Hybridization conditions can also be rendered more stringent by the addition of increasing amounts of formamide, to destabilize the hybrid duplex. Thus, particular hybridization conditions can be readily manipulated, and will generally be chosen depending on the desired results.
  • DNA fragments are generated, some of which will encode parts or the whole of a DPI. Any suitable method for preparing DNA fragments may be used in the present invention.
  • the DNA may be cleaved at specific sites using various restriction enzymes.
  • one may use DNAse in the presence of manganese to fragment the DNA, or the DNA can be physically sheared, as for example, by sonication.
  • the DNA fragments can then be separated according to size by standard techniques, including but not limited to agarose and polyacrylamide gel electrophoresis, column chromatography and sucrose gradient centrifugation.
  • the DNA fragments can then be inserted into suitable vectors, including but not limited to plasmids, cosmids, bacteriophages lambda or T4, and yeast artificial chromosome (YAC).
  • suitable vectors including but not limited to plasmids, cosmids, bacteriophages lambda or T4, and yeast artificial chromosome (YAC).
  • the genomic library may be screened by nucleic acid hybridization to labeled probe (Benton and Davis, 1977, Science 196:180; Grunstein and Hogness, 1975, Proc. Natl. Acad. Sci. U.S.A. 72:3961).
  • the genomic libraries may be screened with labeled degenerate oligonucleotide probes corresponding to the amino acid sequence of any peptide of the DPI using optimal approaches well known in the art.
  • Any probe used is at least 10 nucleotides, at least 15 nucleotides, at least 20 nucleotides, at least 25 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, or at least 100 nucleotides.
  • a probe is 10 nucleotides or longer, and more preferably 15 nucleotides or longer.
  • DPIs disclosed herein correspond to isoforms of previously identified proteins encoded by genes whose sequences are publicly known.
  • any probe may be used that is complementary to the gene or its complement; preferably the probe is 10 nucleotides or longer, more preferably 15 nucleotides or longer.
  • SWISS-PROT and trEMBL databases (held by the Swiss Institute of Bioinformatics (SIB) and the European Bioinformatics Institute (EBI) which are available at http://www.expasy.ch/) and the GenBank database (held by the National Institute of Health (NTH) which is available at http://www.ncbi.nlm.nih.gov/) provide protein sequences for the DPIs listed in Tables IV and V under the following accession numbers and each sequence is incorporated herein by reference:
  • DPI-45 and DPI-213 the partial sequence information derived from tandem mass spectrometry was not found to be described as a transcribed protein in any known public database. DPI-45 and DPI-213 are therefore listed as 'NOVEL' in Table X. DPI-45 and DPI-213 have been cloned, and is further described below.
  • degenerate probes or probes taken from the sequences described above by accession number may be used for screening. In the case of degenerate probes, they can be constructed from the partial amino sequence information obtained from tandem mass spectra of tryptic digest peptides of the DPI. To screen such a gene, any probe may be used that is complementary to the gene or its complement; preferably the probe is 10 nucleotides or longer, more preferably 15 nucleotides or longer.
  • oligonucleotide probes When a library is screened, clones with insert DNA encoding the DPI or a fragment thereof will hybridize to one or more members of the corresponding set of degenerate oligonucleotide probes (or their complement). Hybridization of such oligonucleotide probes to genomic libraries is carried out using methods known in the art.
  • hybridization with one of the above-mentioned degenerate sets of oligonucleotide probes, or their complement can be performed under highly stringent or moderately stringent ' conditions as defined above, or can be carried out in 2X SSC, 1.0% SDS at 50 °C and washed using the washing conditions described supra for highly stringent or moderately stringent hybridization.
  • clones containing nucleotide sequences encoding the entire DPI, a fragment of a DPI, a DPI-related polypeptide, or a fragment of a DPI-related polypeptide or any of the foregoing may also be obtained by screening expression libraries.
  • DNA from the relevant source is isolated and random fragments are prepared and ligated into an expression vector (e.g., a bacteriophage, plasmid, phagemid or cosmid) such that the inserted sequence in the vector is capable of being expressed by the host cell into which the vector is then introduced.
  • an expression vector e.g., a bacteriophage, plasmid, phagemid or cosmid
  • Various screening assays can then be used to select for the expressed DPI or DPI-related polypeptides.
  • the various anti-DPI antibodies of the invention can be used to identify the desired clones using methods known in the art. See, for example, Harlow and Lane, 1988, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, Appendix TV. Colonies or plaques from the library are brought into contact with the antibodies to identify those clones that bind antibody. In an embodiment, colonies or plaques containing DNA that encodes a DPI, a fragment of a DPI, a DPI-related polypeptide, or a fragment of a DPI-related polypeptide can be detected using DYNA Beads according to Olsvick et al., 29th ICAAC, Houston, Tex. 1989, incorporated herein by reference.
  • Anti-DPI antibodies are crossiinked to tosylated DYNA Beads M280, and these antibody-containing beads are then contacted with colonies or plaques expressing recombinant polypeptides. Colonies or plaques expressing a DPI or DPI-related polypeptide are identified as any of those that bind the beads.
  • the anti-DPI antibodies can be nonspecifically immobilized to a suitable support, such as silica or Celite® resin. This material is then used to adsorb to bacterial colonies expressing the DPI protein or DPI-related polypeptide as described herein.
  • PCR amplification may be used to isolate from genomic DNA a substantially pure DNA (i.e., a DNA substantially free of contaminating nucleic acids) encoding the entire DPI or a part thereof.
  • a substantially pure DNA i.e., a DNA substantially free of contaminating nucleic acids
  • a DNA is at least 95% pure, more preferably at least 99% pure.
  • Oligonucleotide sequences, degenerate or otherwise, that correspond to peptide sequences of DPIs disclosed herein can be used as primers.
  • PCR can be carried out, e.g., by use of a Perkin-Elmer Cetus thermal cycler and Taq poiymerase (Gene Amp® or AmpliTaq DNA poiymerase).
  • the gene encoding a DPI can also be identified by mRNA selection by nucleic acid hybridization followed by in vitro translation. In this procedure, fragments are used to isolate complementary mRNAs by hybridization. Such DNA fragments may represent available, purified DNA encoding a DPI of another species (e.g., mouse, human). Immunoprecipitation analysis or functional assays (e.g., aggregation ability in vitro; binding to receptor) of the in vitro translation products of the isolated products of the isolated mRNAs identifies the mRNA and, therefore, the complementary DNA fragments that contain the desired sequences. In addition, specific mRNAs may be selected by adsorption of polysomes isolated from cells to immobilized antibodies that specifically recognize a DPI.
  • a radiolabelled cDNA encoding a DPI can be synthesized using the selected mRNA (from the adsorbed polysomes) as a template. The radiolabelled mRNA or cDNA may then be used as a probe to identify the DNA fragments encoding a DPI from among other genomic DNA fragments.
  • RNA for cDNA cloning of the gene encoding a DPI can be isolated from cells which express the DPI.
  • Any suitable eukaryotic cell can serve as the nucleic acid source for the molecular cloning of the gene encoding a DPI.
  • the nucleic acid sequences encoding the DPI can be isolated from vertebrate, mammalian, primate, human, porcine, bovine, feline, avian, equine, canine or murine sources.
  • the DNA may be obtained by standard procedures known in the art from cloned DNA (e.g., a DNA "library”), by chemical synthesis, by cDNA cloning, or by the cloning of genomic DNA, or fragments thereof, purified from the desired cell.
  • Clones derived from genomic DNA may contain regulatory and intron DNA regions in addition to coding regions; clones derived from cDNA will contain only exon sequences.
  • the identified and isolated gene or cDNA can then be inserted into any suitable cloning vector.
  • vector-host systems known in the art may be used.
  • the vector system chosen be compatible with the host cell used.
  • Such vectors include, but are not limited to, bacteriophages such as lambda derivatives, plasmids such as PBR322 or pUC plasmid derivatives or the Bluescript vector (Stratagene) or modified viruses such as adenoviruses, adeno-associated viruses or refroviruses.
  • the insertion into a cloning vector can be accomplished, for example, by ligating the DNA fragment into a cloning vector which has complementary cohesive termini.
  • the ends of the DNA molecules may be enzymatically modified.
  • any site desired may be produced by ligating nucleotide sequences (linkers) onto the DNA termini; these ligated linkers may comprise specific chemically synthesized oligonucleotides encoding restriction endonuclease recognition sequences.
  • the cleaved vector and the gene encoding a DPI may be modified by homopolymeric tailing. Recombinant molecules can be introduced into host cells via transformation, transfection, infection, electroporation, etc., so that many copies of the gene sequence are generated.
  • transformation of host cells with recombinant DNA molecules that incorporate the isolated gene encoding the DPI, cDNA, or synthesized DNA sequence enables generation of multiple copies of the gene.
  • the gene may be obtained in large quantities by growing transformants, isolating the recombinant DNA molecules from the transformants and, when necessary, retrieving the inserted gene from the isolated recombinant DNA.
  • nucleotide sequences of the present invention include nucleotide sequences encoding amino acid sequences with substantially the same amino acid sequences as native DPIs, nucleotide sequences encoding amino acid sequences with functionally equivalent amino acids, nucleotide sequences encoding DPIs, a fragments of DPIs, DPI-related polypeptides, or fragments of DPI-related polypeptides.
  • an isolated nucleic acid molecule encoding a DPI- related polypeptide can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of a DPI such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.
  • Standard techniques known to those of skill in the art can be used to introduce mutations, including, for example, site-directed mutagenesis and PCR- mediated mutagenesis.
  • conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues.
  • a "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a side chain with a similar charge.
  • Families of amino acid residues having side chains with similar charges have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.
  • mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity. Following mutagenesis, the encoded protein can be expressed and the activity of the protein can be determined.
  • tandem mass spectra of the peptides were interpreted manually, using methods known in the art as described in the Examples, infra.
  • the following pairs of amino acids could not be distinguished from each other: leucine and isoleucine; and, under certain circumstances, glutamine and lysine, and phenylalanine and oxidized methionine.
  • an amino acid sequence "as determined by mass spectrometry" refers to the set of amino acid sequences containing at the indicated positions, one or other member of the designated pairs of amino acids.
  • amino acid sequence P[L/I]A indicates the amino acid sequences PLA and PIA.
  • a sequence containing n designated pairs indicates 2 n amino acid sequences.
  • Table XI each possible amino acid sequence is listed for each sequence determined by mass spectrometry.
  • the masses determined by mass spectrometry have an error of mass measurement of 100 parts-per- million (ppm) or less.
  • M having an error of mass measurement of z ppm
  • the error of mass measurement can be calculated as ⁇ M x z ⁇ 1000000).
  • the "mass of the peptide analyzed by mass spectrometry” is the mass of the singly protonated peptide measured by mass spectrometry, and corresponds to the total mass of the constituent amino acid residues of the peptide with the addition of a water molecule (H2O) and a single proton (H+).
  • the "mass to N- terminus” corresponds to the total mass of the constituent amino acid residues extending from the start of the partial sequence to the N-terminus of the peptide.
  • the "mass to C-terminus” corresponds to the total mass of the constituent amino acid residues extending from the end of the partial sequence to the C-terminus of the peptide with the addition of a water molecular (H2O), and a single proton (H+).
  • EST AA326679 shows 44% amino acid identity with a putative human protein derived from a conceptual translation of the cDNA CAB07646.1 (available at http://www.ncbi.nlm.nih.gov/entrez/).
  • the C terminus of this protein sequence shows a similar level of homology with a further brain tissue derived EST (AI589129) (TBlastN, BLAST, Altschul, Stephen F., Gish, Warren, Miller, Webb, Myers, Eugene W., and Lipman, David J. (1990). Basic local alignment search tool. J. Mol.
  • PCR primers (1 & 2 from Table XII) from EST AA326679 and EST AI589129 were used in a PCR reaction (1 ml of Advantage 2 cDNA poiymerase mix (Clontech) in a buffer containing 50mM KCl, 10 mM Tris-HCl, 1.5 mM MgC12, ⁇ H8.3; 0.2mM each of dATP, dCTP, dGTP, dTTP and 10 pmoles of oligonucleotide primers.
  • Reactions were routinely made to a final volume of 50ml and amplification carried out in a PE GeneAmpSystems 9700 PCR machine with the following cycling conditions: initial denaturation of 94 °C for 1 minute followed by 30 cycles of 94 °C for 30 seconds, 55 °C for 30 seconds and 72 °C for 2 minutes. Reaction products were resolved by standard agarose gel electrophoresis and stained with Ethidium Bromide) on lOng of whole brain cDNA (Clontech, USA). The resulting 1.6kb fragment was purified from primers and buffers (Qiagen, UK) and sequenced using the primers given in Table XII (1,2, 3 & 4). This generated overlapping sequence for the entire product.
  • SEQ ID no.l and Figure 2A show the DNA sequence.
  • SEQ ID no. 2 and Figure 2B show the protein sequence of the open reading frame (ORF) seen in SEQ ID no.l, demonstrating the presence of the two peptides identified by mass spectrometry.
  • gag tgg gtg gcc ate gag age gac tct gtc cag cct gtg cct Glu Trp Val Ala He Glu Ser Asp Ser Val Gin Pro Val Pro
  • DPI 45 and DPI-213 were also found to be differentially present in a sample of CSF from a subject having Schizophrenia compared with a sample of CSF from a subject free from Schizophrenia, being decreased 1.50 fold.
  • WO99/58660 disclosed 97 human secreted proteins. These included a sequence, identified as Gene No: 21, which corresponds to DPI 45 discussed herein.
  • SEQ ID no.l shares 44% identity with a putative human protein derived from a conceptual translation of the cDNA CAB07646.1 (available at http://www.ncbi.nlm.nih.gov/entrez/), no function has been assigned to CAB07646.1.
  • PSORT Nai, K. and Kanehisa, M., A knowledge base for predicting protein localization sites in eukaryotic cells, Genomics 14, 897-911 (1992)
  • analysis of SEQ ID no.l identifies only a signal sequence at amino acids 1-20, with proteolytic cleavage predicted between amino acids 20 and 21. Thus the mRNA expression and protein structure analyses are consistent with this protein being secreted from brain tissues and being assayable in CSF.
  • the nucleotide sequence coding for a DPI, a DPI analog, a DPI-related peptide, or a fragment or other derivative of any of the foregoing, can be inserted into an appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted protein-coding sequence.
  • the necessary transcriptional and translational signals can also be supplied by the native gene encoding the DPI or its flanking regions, or the native gene encoding the DPI- related polypeptide or its flanking regions.
  • a variety of host- vector systems may be utilized in the present invention to express the protein-coding sequence.
  • mammalian cell systems infected with virus e.g., vaccinia virus, adenovirus, etc.
  • insect cell systems infected with virus e.g., baculovirus
  • microorganisms such as yeast containing yeast vectors; or bacteria transformed with bacteriophage, DNA, plasmid DNA, or cosmid DNA.
  • the expression elements of vectors vary in their strengths and specificities. Depending on the host- vector system utilized, any one of a number of suitable transcription and translation elements may be used.
  • a nucleotide sequence encoding a human gene or a nucleotide sequence encoding a functionally active portion of a human DPI
  • a fragment of a DPI comprising a domain of the DPI is expressed.
  • any of the methods previously described for the insertion of DNA fragments into a vector may be used to construct expression vectors containing a chimeric gene consisting of appropriate transcriptional and translational control signals and the protein coding sequences. These methods may include in vitro recombinant DNA and synthetic techniques and in vivo recombinants (genetic recombination). Expression of nucleic acid sequence encoding a DPI or fragment thereof may be regulated by a second nucleic acid sequence so that the DPI or fragment is expressed in a host transformed with the recombinant DNA molecule. For example, expression of a DPI may be controlled by any promoter or enhancer element known in the art.
  • Promoters which may be used to control the expression of the gene encoding a DPI or a DPI- related polypeptide include, but are not limited to, the SV40 early promoter region (Bernoist and Chambon, 1981, Nature 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al., 1980, Cell 22:787- 797), the herpes thymidine kinase promoter (Wagner et al., 1981, Proc. Natl. Acad. Sci. U.S.A.
  • promoter elements from yeast or other fungi such as the Gal 4 promoter, the ADC (alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter, alkaline phosphatase promoter, and the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: elastase I gene control region which is active in pancreatic acinar cells (Swift et al., 1984, Cell 38:639- 646; Ornitz et al., 1986, Cold Spring Harbor Symp.
  • mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al., 1986, Cell 45:485-495), albumin gene control region which is active in liver (Pinkert et al., 1987, Genes and Devel. 1:268- 276), alpha-fetoprotein gene control region which is active in liver (Krumlauf et al., 1985, Mol. Cell. Biol. 5:1639-1648; Hammer et al., 1987, Science 235:53-58; alpha 1- antitrypsin gene control region which is active in the liver (Kelsey et al., 1987, Genes and Devel.
  • beta-globin gene control region which is active in myeloid cells (Mogram et al., 1985, Nature 315:338-340; Kollias et al., 1986, Cell 46:89-94; myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al., 1987, Cell 48:703-712); myosin light chain-2 gene control region which is active in skeletal muscle (Sani, 1985, Nature 314:283-286); neuronal-specific enolase (NSE) which is active in neuronal cells (Morelli et al., 1999, Gen. Virol.
  • NSE neuronal-specific enolase
  • BDNF brain-derived neurotrophic factor
  • GFAP glial fibrillary acidic protein promoter which is active in astrocytes
  • Gomes et al., 1999, Braz J Med Biol Res 32(5):619-631; Morelli et al., 1999, Gen. Virol. 80:571-83) and gonadotropic releasing hormone gene control region which is active in the hypothalamus (Mason et al., 1986, Science 234:1372-1378).
  • a vector is used that comprises a promoter operably linked to a DPI-encoding nucleic acid, one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic resistance gene).
  • an expression construct is made by subcloning a DPI or a DPI-related polypeptide coding sequence into the EcoRl restriction site of each of the three pGEX vectors (Glutathione S-Transferase expression vectors; Smith and Johnson, 1988, Gene 7:31-40). This allows for the expression of the DPI product or DPI-related polypeptide from the subclone in the correct reading frame.
  • a number of viral-based expression systems may be utilized.
  • the DPI coding sequence or DPI-related polypeptide coding sequence may be ligated to an adenovirus transcription translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination.
  • Insertion in a non-essential region of the viral genome will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts, (e.g., see Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:355-359).
  • Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert.
  • These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
  • Expression vectors containing inserts of a gene encoding a DPI or a DPI- related polypeptide can be identified by three general approaches: (a) nucleic acid hybridization, (b) presence or absence of "marker" gene functions, and (c) expression of inserted sequences.
  • the presence of a gene encoding a DPI inserted in an expression vector can be detected by nucleic acid hybridization using probes comprising sequences that are homologous to an inserted gene encoding a DPI.
  • the recombinant vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, etc.) caused by the insertion of a gene encoding a DPI in the vector.
  • certain "marker" gene functions e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, etc.
  • recombinant expression vectors can be identified by assaying the gene product (i.e., DPI) expressed by the recombinant.
  • DPI gene product expressed by the recombinant.
  • assays can be based, for example, on the physical or functional properties of the DPI in in vitro assay systems, e.g., binding with anti-DPI antibody.
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus, expression of the genetically engineered DPI or DPI-related polypeptide may be controlled.
  • different host cells have characteristic and specific mechanisms for the translational and post-translational processing and modification (e.g., glycosylation, phosphorylation of proteins). Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the foreign protein expressed. For example, expression in a bacterial system will produce an unglycosylated product and expression in yeast will produce a glycosylated product.
  • Eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • mammalian host cells include but are not limited to CHO, VERO, BHK, Hela, COS, MDCK, 293, 3T3, WI38, and in particular, neuronal cell lines such as, for example, SK-N-AS, SK-N-FI, SK-N-DZ human neuroblastomas (Sugimoto et al., 1984, J. Natl. Cancer Inst. 73: 51-57), SK-N-SH human neuroblastoma (Biochim. Biophys.
  • cell lines which stably express the differentially expressed or pathway gene protein may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched medium, and then are switched to a selective medium.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • This method may advantageously be used to engineer cell lines which express the differentially expressed or pathway gene protein.
  • Such engineered cell lines may be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the differentially expressed or pathway gene protein.
  • a number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler, et al., 1977, Cell 11 :223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1962, Proc. Natl. Acad. Sci. USA 48:2026), and adenine phosphoribosyltransferase (Lowy, et al., 1980, Cell 22:817) genes can be employed in tk-, hgprt- or aprt- cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methofrexate (Wigler, et al., 1980, Natl. Acad. Sci. USA 77:3567; O ⁇ are, et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenohc acid (Mulligan & Berg, 1981, Proc. Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin, et al, 1981, J. Mol. Biol.
  • the DPI, fragment, analog, or derivative may be expressed as a fusion, or chimeric protein product (comprising the protein, fragment, analog, or derivative joined via a peptide bond to a heterologous protein sequence).
  • the polypeptides of the present invention may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHI , CH2, CH3, or any combination thereof and portions thereof) resulting in chimeric polypeptides.
  • Such fusion proteins may facilitate purification, increase half- life in vivo, and enhance the delivery of an antigen across an epithelial barrier to the immune system.
  • An increase in the half-life in vivo and facilitated purification has been shown for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins. See, e.g., EP 394,827; Traunecker et al., Nature, 331:84-86 (1988).
  • antigens e.g., insulin
  • FcRn binding partner such as IgG or Fc fragments
  • Nucleic acids encoding a DPI, a fragment of a DPI, a DPI-related polypeptide, or a fragment of a DPI-related polypeptide can be fused to an epitope tag (e.g., the hemagglutinin ("HA") tag or flag tag) to aid in detection and purification of the expressed polypeptide.
  • an epitope tag e.g., the hemagglutinin ("HA") tag or flag tag
  • HA hemagglutinin
  • a system described by Janknecht et al. allows for the. ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al., 1991, Proc. Natl. Acad. Sci. USA 88:8972-897).
  • Fusion proteins can be made by ligating the appropriate nucleic acid sequences encoding the desired amino acid sequences to each other by methods known in the art, in the proper coding frame, and expressing the chimeric product by methods commonly known in the art.
  • a fusion protein may be made by protein synthetic techniques, e.g., by use of a peptide synthesizer.
  • cDNA and genomic sequences can be cloned and expressed.
  • Domains of some DPIs are known in the art and have been described in the scientific literature. Moreover, domains of a DPI can be identified using techniques known to those of skill in the art. For example, one or more domains of a DPI can be identified by using one or more of the following programs: ProDom, TMpred, and SAPS.
  • ProDom compares the amino acid sequence of a polypeptide to a database of compiled domains (see, e.g., http://www.toulouse.inra.fr/prodom.html; Corpet F., Gouzy J. & Kahn D., 1999, Nucleic Acids Res., 27:263-267).
  • TMpred predicts membrane-spanning regions of a polypeptide and their orientation.
  • TMbase a database of naturally occuring transmembrane proteins
  • the SAPS program analyzes polypeptides for statistically . significant features like charge-clusters, repeats, hydrophobic regions, compositional domains (see, e.g., Brendel et al., 1992, Proc. Natl. Acad. Sci. USA 89: 2002-2006).
  • the skilled artisan can identify domains of a DPI having enzymatic or binding activity, and further can identify nucleotide sequences encoding such domains. These nucleotide sequences can then be used for recombinant expression of a DPI fragment that retains the enzymatic or binding activity of the DPI.
  • the skilled artisan can identify domains of a DPI having enzymatic or binding activity, and further can identify nucleotide sequences encoding such domains. These nucleotide sequences can then be used for recombinant expression of DPI fragments that retain the enzymatic or binding activity of the DPI.
  • a DPI has an amino acid sequence sufficiently similar to an identified domain of a known polypeptide.
  • the term "sufficiently similar” refers to a first amino acid or nucleotide sequence which contains a sufficient number of identical or equivalent (e.g., with a similar side chain) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences have or encode a common structural domain or common functional activity or both.
  • a DPI domain can be assessed for its function using techniques well known to those of skill in the art.
  • a domain can be assessed for its kinase activity or for its ability to bind to DNA using techniques known to the skilled artisan.
  • Kinase activity can be assessed, for example, by measuring the ability of a polypeptide to phosphorylate a substrate.
  • DNA binding activity can be assessed, for example, by measuring the ability of a polypeptide to bind to a DNA binding element in a electromobility shift assay.
  • the function of a domain of a DPI is determined using an assay described in one or more of the references identified in Table XIII, infra.
  • a DPI, DPI analog, DPI-related protein or a fragment or derivative of any of the foregoing may be used as an immunogen to generate antibodies which immunospecifically bind such an immunogen.
  • immunogens can be isolated by any convenient means, including the methods described above.
  • Antibodies of the invention include, but are not limited to polyclonal, monoclonal, bispecific, humanized or chimeric antibodies, single chain antibodies, Fab fragments and F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • antibody refers to immunoglobulin, molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen.
  • the immunoglobulin molecules of the invention can be of any class (e.g., IgG, IgE, IgM, IgD and IgA ) or subclass of immunoglobulin molecule.
  • antibodies that recognize gene products of genes encoding DPIs are publicly available.
  • antibodies that recognize these DPIs and/or their isoforms include antibodies recognizing, DPI-3, DPI-5, DPI-8, DPI-9, DPI-10, DPI-11, DPI-12, DPI-13, DPI-14, DPI-15, DPI-17, DPI-19, DPI-20, DPI-23, DPI-24, DPI-25, DPI-29, DPI-30, DPI-35, DPI-37, DPI-44, DPI-51, DPI-57, DPI-58, DPI-59, DPI-60, DPI-65, DPI-66, DPI-67, DPI-71, DPI-72, DPI-73, DPI-75, DPI-76, DPI-79, DPI-87, DPI-88, DPI-89, DPI-90, DPI-92, DPI-108, DPI-109, DPI-110, DPI-113, DPI-115, DPI-
  • antibodies to a specific domain of a DPI are produced.
  • hydrophilic fragments of a DPI are used as immunogens for antibody production.
  • screening for the desired antibody can be accomplished by techniques known in the art, e.g. ELISA (enzyme-linked immunosorbent assay).
  • ELISA enzyme-linked immunosorbent assay
  • an antibody that specifically binds a first DPI homolog but which does not specifically bind to (or binds less avidly to) a second DPI homolog one can select on the basis of positive binding to the first DPI homolog and a lack of binding to (or reduced binding to) the second DPI homolog.
  • the present invention provides ari antibody (preferably a monoclonal antibody) that binds with greater affinity (preferably at least 2-fold, more preferably at least 5-fold still more preferably at least 10-fold greater affinity) to a DPI than to a different isoform or isoforms (e.g., glycoforms) of the DPI.
  • Polyclonal antibodies which may be used in the methods of the invention are heterogeneous populations of antibody molecules derived from the sera of immunized animals. Unfractionated immune serum can also be used. Various procedures known in the art may be used for the production of polyclonal antibodies to a DPI, a fragment of a DPI, a DPI-related polypeptide,. or a fragment of a DPI-related polypeptide. In a particular embodiment, rabbit polyclonal antibodies to an epitope of a DPI or a DPI- related polypeptide can be obtained.
  • various host animals can be immunized by injection with the native or a synthetic (e.g., recombinant) version of a DPI, a fragment of a DPI, a DPI- related polypeptide, or a fragment of a DPI-related polypeptide, including but not limited to rabbits, mice, rats, etc.
  • the Preferred Technology described herein provides isolated DPIs suitable for such immunization. If the DPI is purified by gel electrophoresis, the DPI can be used for immunization with or without prior extraction from the polyacrylamide gel.
  • adjuvants may be used to enhance the immunological response, depending on the host species, including, but not limited to, complete or incomplete Freund's adj ⁇ vant, a mineral gel such as aluminum hydroxide, surface active substance such as lysolecithin, pluronic polyol, a polyanion, a peptide, an oil emulsion, keyhole limpet hemocyanin, dinitrophenol, and an adjuvant such as BCG (bacille Calmette-Guerin) or corynebacterium parvum. Additional adjuvants are also well known in the art.
  • any technique which provides for the production of antibody molecules by continuous cell lines in culture may be used.
  • the hybridoma technique originally developed by Kohler and Milstein (1975, Nature 256:495-497), as well as the frioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies Colde et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R.
  • Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof.
  • the hybridoma producing the mAbs of the invention may be cultivated in vitro or in vivo.
  • monoclonal antibodies can be produced in germ-free animals utilizing known technology (PCT/US90/02545, incorporated herein by reference).
  • the monoclonal antibodies include but are not limited to human monoclonal antibodies and chimeric monoclonal antibodies (e.g., human-mouse chimeras).
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a human immunoglobulin constant region and a variable region derived from a murine mAb.
  • Humanized antibodies are antibody molecules from non- human species having one or more complementarily determining regions (CDRs) from the non-human species and a framework region from a human immunoglobulin molecule.
  • CDRs complementarily determining regions
  • Chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in PCT Publication No. WO 87/02671; European Patent Application 184,187; European Patent Application 171 ,496; European Patent Application 173 ,494; PCT Publication No. WO 86/01533; U.S. Patent No. 4,816,567; European Patent Application 125,023; Better et al., 1988, Science 240:1041-1043; Liu et al., 1987, Proc. Natl. Acad. Sci. USA 84:3439-3443; Liu et al., 1987, J. Immunol.
  • Fully human antibodies are particularly desirable for therapeutic treatment of human subjects.
  • Such antibodies can be produced using transgenic mice which are incapable of expressing endogenous immunoglobulin heavy and light chains genes, but which can express human heavy and light chain genes.
  • the transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a DPI of the invention.
  • Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation.
  • phage display methods functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
  • Phage used in these methods are typically filamentous phage including fd and Ml 3 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein.
  • Phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177- 186 (1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994); Persic et al., Gene 187 9-18 (1997); Burton et al., Advances in Immunology 57: 191-280 (1994); PCT Application No.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below.
  • techniques to recombinantly produce Fab, Fab 1 and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in PCT publication WO 92/22324; Mullinax et al., BioTechniques 12(6):864-869 (1992); and Sawai et al., AJRI 34:26-. 34 (1995); and Better et al., Science 240:1041-1043 (1988) (said references incorporated by reference in their entireties).
  • the invention further provides for the use of bispecific antibodies, which can be made by methods known in the art.
  • Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Milstein et al., 1983, Nature 305:537-539). Because of the randorh assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al., 1991, EMBO J. 10:3655-3659 .
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHI) containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co-transfected into a suitable host organism.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation.
  • This approach is disclosed in WO 94/04690 published March 3,1994.
  • For further details for generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 1986, 121:210.
  • the invention provides functionally active fragments, derivatives or analogs of the anti-DPI immunoglobulin molecules.
  • Functionally active means that the fragment, derivative of analog is able to elicit anti-anti-idiotype antibodies (i.e., tertiary antibodies) that recognize the same antigen that is recognized by the antibody from which the fragment, derivative or analog is derived.
  • antigenicity of the idiotype of the immunoglobulin molecule may be enhanced by deletion of framework and CDR sequences that are C-terminal to the CDR sequence that specifically recognizes the antigen.
  • the present invention provides antibody fragments such as, but not limited to, F(ab')2 fragments and Fab fragments.
  • Antibody fragments which recognize specific epitopes may be generated by known techniques.
  • F(ab')2 fragments consist of the variable region, the light chain constant region and the CHI domain of the heavy chain and are generated by pepsin digestion of the antibody molecule.
  • Fab fragments are generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • the invention also provides heavy chain and light chain dimers of the antibodies of the invention, or any minimal fragment thereof such as Fvs or single chain antibodies (SCAs) (e.g., as described in U.S. Patent 4,946,778; Bird, 1988, Science 242:423-42; Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; and Ward et al., 1989, Nature 334:544-54), or any other molecule with the same specificity as the antibody of the invention.
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in E.
  • the invention provides fusion proteins of the immunoglobulins of the invention (or functionally active fragments thereof), for example in which the immunoglobulin is fused via a covalent bond (e.g., a peptide bond), at either the N-terminus or the C-terminus to an amino acid sequence of another protein (or portion thereof, preferably at least 10, 20 or 50 amino acid portion of the protein) that is not the immunoglobulin.
  • a covalent bond e.g., a peptide bond
  • the immunoglobulin, or fragment thereof is covalently linked to the other protein at the N-terminus of the constant domain.
  • immunoglobulins of the invention include analogs and derivatives that are either modified, i.e, by the covalent attachment of any type of molecule as long as such covalent attachment that does not impair immunospecific binding.
  • the derivatives and analogs of the immunoglobulins include those that have been further modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, etc. Additionally, the analog or derivative may contain one or more non-classical amino acids.
  • the foregoing antibodies can be used in methods known in the art relating to the localization and activity of the DPIs of the invention, e.g., for imaging these proteins, measuring levels thereof in appropriate physiological samples, in diagnostic methods, etc. 5.10 Expression Of Antibodies
  • the antibodies of the invention can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or by recombinant expression, and are preferably produced by recombinant expression techniques.
  • a nucleic acid encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., 1994, BioTechniques 17:242), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding antibody, annealing and ligation of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.
  • the nucleic acid encoding the antibody may be obtained by cloning the antibody. If a clone containing the nucleic acid encoding the particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the antibody may be obtained from a suitable source (e.g., an antibody cDNA library, or cDNA library generated from any tissue or cells expressing the antibody) by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence.
  • a suitable source e.g., an antibody cDNA library, or cDNA library generated from any tissue or cells expressing the antibody
  • antibodies specific for a particular antigen may be generated by any method known in the art, for example, by immunizing an animal, such as a rabbit, to generate polyclonal antibodies or, more preferably, by generating monoclonal antibodies.
  • a clone encoding at least the Fab portion of the antibody may be obtained by screening Fab expression libraries (e.g., as described in Huse et al., 1989, Science 246:1275-1281) for clones of Fab fragments that bind the specific antigen or by screening antibody libraries (See, e.g., Clackson et al., 1991, Nature 352:624; Hane et al., 1997 Proc. Natl. Acad. Sci. USA 94:4937).
  • nucleic acid encoding at least the variable domain of the antibody molecule may be introduced into a vector containing the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Patent No. 5,122,464).
  • Vectors containing the complete light or heavy chajn for co-expression with the nucleic acid to allow the expression of a complete antibody molecule are also available.
  • the nucleic acid encoding the antibody can be used to introduce the nucleotide substitution(s) or deletion(s) necessary to substitute (or delete) the one or more variable region cysteine residues participating in an intrachain disulfide bond with an amino acid residue that does not contain a sulfhydyl group.
  • Such modifications can be carried out by any method known in the art for the introduction of specific mutations or deletions in a nucleotide sequence, for example, but not limited to, chemical mutagenesis, in vitro site directed mutagenesis (Hutchinson et al., 1978, J. Biol. Chem. 253:6551), PCT based methods, etc.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human antibody constant region, e.g., humanized antibodies.
  • the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art.
  • methods for preparing the protein of the invention by expressing nucleic acid containing the antibody molecule sequences are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing an antibody molecule coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. See, for example, the techniques described in Sambrook et al.
  • the expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody of the invention.
  • the host cells used to express a recombinant antibody of the invention may be either bacterial cells such as Escherichia coli, or, preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule.
  • mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., 1986, Gene 45:101; Cockett et al., 1990, Bio/Technology 8:2).
  • host-expression vector systems riiay be utilized to express an antibody molecule of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express the antibody molecule of the invention in situ.
  • These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionem , promoter) or
  • a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed.
  • vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al., 1983, EMBO J. 2:1791), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, 1985, Nucleic Acids Res.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-fransferase (GST).
  • GST glutathione S-fransferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to a matrix glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • the antibody coding sequence may be cloned individually into non- essential regions (for example the polyhedrin gene) of the virus and placed under confrol of an AcNPV promoter (for example the polyhedrin promoter).
  • a number of viral-based expression systems e.g., an adenovirus expression system
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired.
  • Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • stable expression is preferred.
  • cells lines that stably express an antibody of interest can be produced by fransfecting the cells with an expression vector comprising the nucleotide sequence of the antibody and the nucleotide sequence of a selectable (e.g., neomycin or hygromycin), and selecting for expression of the selectable marker.
  • a selectable e.g., neomycin or hygromycin
  • the expression levels of the antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • vector amplification for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • a marker in the vector system expressing antibody is amplifiable
  • increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., 1983, Mol. Cell. Biol. 3:257).
  • the host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide.
  • the two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used which encodes both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, 1986, Nature 322:52; Kohler, 1980, Proc. Natl. Acad. Sci. USA 77:2197).
  • the coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
  • the antibody molecule of the invention may be purified by any method known in the art for purification of an antibody molecule, for example, by chromatography (e.g., ion exchange chromatography, affinity chromatography such as with protein A or specific antigen, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • chromatography e.g., ion exchange chromatography, affinity chromatography such as with protein A or specific antigen, and sizing column chromatography
  • centrifugation e.g., centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • any fusion protein may be readily purified by utilizing an antibody specific for the fusion protein being expressed.
  • a system described by Janknecht et al. allows for the ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al., 1991, Proc. Natl. Acad. Sci. USA 88:8972-897).
  • the gene of interest is subcloned into a vaccinia recombination plasmid such that the open reading frame of the gene is translationally fused to an amino-terminal tag consisting of six histidine residues.
  • the tag serves as a matrix binding domain for the fusion protein. Extracts from cells infected with recombinant vaccinia virus are loaded onto Ni2+ nitriloacetic acid-agarose columns and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
  • anti-DPI antibodies or fragments thereof are conjugated to a diagnostic or therapeutic moiety.
  • the antibodies can be used for diagnosis or to determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive nuclides, positron emitting metals (for use in positron emission tomography), and nonradioactive paramagnetic metal ions. See generally U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention.
  • Suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylchohnesterase; suitable prosthetic groups include streptavidin, avidin and biotin; suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride and phycoerythrin; suitable luminescent materials include luminol; suitable bioluminescent materials include luciferase, luciferin, and aequorin; and suitable radioactive nuclides include 1251, 1311, l l lln and 99Tc.
  • Anti-DPI antibodies or fragments thereof can be conjugated to a therapeutic agent or drug moiety to modify a given biological response.
  • the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, a thrombotic agent or an anti-angiogenic agent, e.g., angiostatin or endostatin; or, a biological response modifier such as a lymphokine, interleukin-1 (IL-1), interleukin-2 (IL-2), interleukin-6 (IL-6), granulocyte macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), nerve growth factor (NGF) or other growth factor.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin
  • a protein such as tumor necrosis factor,
  • an antibody can be conjugated to a second antibody to form an antibody heteroco ⁇ jugate as described by Segal in U.S. Patent No. 4,676,980.
  • An antibody with or without a therapeutic moiety conjugated to it can be used as a therapeutic that is administered alone or in combination with cytotoxic factor(s) and or cytokine(s).
  • test samples of cerebrospinal fluid (CSF), serum, plasma or urine obtained from a subject suspected of having or known to have BAD can be used for diagnosis or monitoring.
  • CSF cerebrospinal fluid
  • a decreased, abundance of one or more DFs or DPIs (or any combination of them) in a test sample relative to a control sample (from a subject or subjects free from BAD) or a previously determined reference range indicates the presence of BAD; DFs and DPIs suitable for this purpose are identified in Tables I and IV, respectively, as described in detail above.
  • an increased abundance of one or more DFs or DPIs (or any combination of them) in a test sample compared to a confrol sample or a previously determined reference range indicates the presence of BAD; DFs and DPIs suitable for this purpose are identified in Tables II and VI, respectively, as described in detail above.
  • the relative abundance of one or more DFs or DPIs (or any combination of them) in a test sample compared to a control sample or a previously determined reference range indicates a subtype of BAD (e.g., familial or sporadic BAD).
  • the relative abundance of one or more DFs or DPIs (or any combination of them) in a test sample relative to a control sample or a previously determined reference range indicates the degree or severity of BAD.
  • detection of one or more DPIs described herein may optionally be combined with detection of one or more additional biomarkers for BAD.
  • any suitable method in the art can be employed to measure the level of DFs and DPIs, including but not limited to the Preferred Technology described herein, kinase assays, immunoassays to detect and/or visualize the DPI (e.g., Western blot, immunoprecipitation followed by sodium dodecyl sulfate polyacrylamide gel electrophoresis, immunocytochemistry, etc.).
  • kinase assays e.g., Western blot, immunoprecipitation followed by sodium dodecyl sulfate polyacrylamide gel electrophoresis, immunocytochemistry, etc.
  • an assay for that function may be used to measure DPI expression.
  • a decreased abundance of mRNA encoding one or more DPIs identified in Table TV (or any combination of them) in a test sample relative to a control sample or a previously determined reference range indicates the presence of BAD.
  • an increased abundance of mRNA encoding one or more DPIs identified in Table V (or any combination of them) in a test sample relative to a control sample or previously determined reference range indicates the presence of BAD.
  • Any suitable hybridization assay can be used to detect DPI expression by detecting and/or visualizing mRNA encoding the DPI (e.g., Northern assays, dot blots, in situ hybridization, etc.).
  • labeled antibodies, derivatives and analogs thereof, which specifically bind to a DPI can be used for diagnostic purposes to detect, diagnose, or monitor BAD.
  • BAD is detected in an animal, more preferably in a mammal and most preferably in a human.
  • the invention provides methods for identifying agents (e.g., candidate compounds or test compounds) that bind to a DPI or have a stimulatory or inhibitory effect on the expression or activity of a DPI.
  • the invention also provides methods of identifying agents, candidate compounds or test compounds that bind to a DPI-related polypeptide or a DPI fusion protein or have a stimulatory or inhibitory effect on the expression or activity of a DPI-related polypeptide or a DPI fusion protein.
  • agents, candidate compounds or test compounds include, but are not limited to, nucleic acids (e.g., DNA and RNA), carbohydrates, lipids, proteins, peptides, peptidomimetics, small molecules and other drugs.
  • Agents can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one- compound” library method; and synthetic library methods using affinity chromatography selection.
  • the biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, 1997, Anticancer Drug Des. 12:145; U.S. Patent No. 5,738,996; and U.S. Patent No.5,807,683, each of which is incorporated herein in its entirety by reference).
  • Libraries of compounds may be presented, e.g., presented in solution (e.g., Houghten, 1992, Bio/Techniques 13:412-421), or on beads (Lam, 1991, Nature 354:82- 84), chips (Fodor, 1993, Nature 364:555-556), bacteria (U.S. Patent No. 5,223,409), spores (Patent Nos. 5,571,698; 5,403,484; and 5,223,409), plasmids (Cull et al., 1992, Proc. Natl. Acad. Sci.
  • agents that interact with (i.e., bind to) a DPI, a DPI fragment (e.g. a functionally active fragment), a DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein are identified in a cell-based assay system.
  • a DPI fragment e.g. a functionally active fragment
  • a DPI-related polypeptide e.g. a functionally active fragment
  • a DPI-related polypeptide e.g. a fragment of a DPI-related polypeptide, or a DPI fusion protein
  • cells expressing a DPI, a fragment of a DPI, a DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein are contacted with a candidate compound or a control compound and the ability of the candidate compound to interact with the DPI is determined.
  • this assay may be used to screen a plurality (e
  • the cell for example, can be of prokaryotic origin (e.g., E. coli) or eukaryotic origin (e.g., yeast or mammalian). Further, the cells can express the DPI, fragment of the DPI, DPI- related polypeptide, a fragment of the DPI-related polypeptide, or a DPI fusion protein endogenously or be genetically engineered to express the DPI, fragment of the DPI, DPI-related polypeptide, a fragment of the DPI-related polypeptide, or a DPI fusion protein.
  • prokaryotic origin e.g., E. coli
  • eukaryotic origin e.g., yeast or mammalian.
  • the cells can express the DPI, fragment of the DPI, DPI- related polypeptide, a fragment of the DPI-related polypeptide, or a DPI fusion protein endogenously or be genetically engineered to express the DPI, fragment of the DPI, DPI-related polypeptide, a
  • the DPI, fragment of the DPI, DPI-related polypeptide, a fragment of the DPI-related polypeptide, or a DPI fusion protein or the candidate compound is labeled, for example with a radioactive label (such as 32 P, 35 S or 125 I) or a fluorescent label (such as fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde or fluorescamine) to enable detection of an interaction between a DPI and a candidate compound.
  • a radioactive label such as 32 P, 35 S or 125 I
  • a fluorescent label such as fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde or fluorescamine
  • the ability of the candidate compound to interact directly or indirectly with a DPI, a fragment of a DPI, a DPI- related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein can be determined by methods known to those of skill in the art.
  • the interaction between a candidate compound and a DPI, a fragment of a DPI, a DPI- related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein can be determined by flow cytometry, a scintillation assay, immunoprecipitation or western blot analysis.
  • agents that interact with (i.e., bind to) a DPI, a DPI fragment (e.g., a functionally active fragment) a DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein are identified in a cell-free assay system.
  • a native or recombinant DPI or fragment thereof, or a native or recombinant DPI-related polypeptide or fragment thereof, or a DPI-fusion protein or fragment thereof is contacted with a candidate compound or a control compound and the ability of the candidate compound to interact with the DPI or DPI-related polypeptide, or DPI fusion protein is determined.
  • this assay may be used to screen a plurality (e.g. a library) of candidate compounds.
  • the DPI, DPI fragment, DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI-fusion protein is first immobilized, by, for example, contacting the DPI, DPI fragment, DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein with an immobilized antibody which specifically recognizes and binds it, or by contacting a purified preparation of the DPI, DPI fragment, DPI-related polypeptide, fragment of a DPI-related polypeptide, or a DPI fusion protein with a surface designed to bind proteins.
  • the DPI, DPI fragment, DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein may be partially or completely purified (e.g., partially or completely free of other polypeptides) or part of a cell lysate. Further, the DPI, DPI fragment, DPI- related polypeptide, a fragment of a DPI-related polypeptide may be a fusion protein comprising the DPI or a biologically active portion thereof, or DPI-related polypeptide and a domain such as glutathionine-S- transferase.
  • the DPI, DPI fragment, DPI-related polypeptide, fragment of a DPI-related polypeptide or DPI fusion protein can be biotinylated using techniques well known to those of skill in the art (e.g., biotinylation kit, Pierce Chemicals; Rockford, IL).
  • biotinylation kit Pierce Chemicals; Rockford, IL.
  • the ability of the candidate compound to interact with a DPI, DPI fragment, DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein can be determined by methods known to those of skill in the art.
  • a cell-based assay system is used to identify agents that bind to or modulate the activity of a protein, such as an enzyme, or a biologically active portion thereof, which is responsible for the production or degradation of a DPI or is responsible for the post- translational modification of a DPI.
  • a protein such as an enzyme, or a biologically active portion thereof, which is responsible for the production or degradation of a DPI or is responsible for the post- translational modification of a DPI.
  • a plurality e.g., a library
  • cells that naturally or recombinantly express: (i) a DPI, an isoform of a DPI, a DPI homolog a DPI-related polypeptide, a DPI fusion protein, or a biologically active fragment of any of the foregoing; and (ii) a protein that is responsible for processing of the DPI, DPI isoform, DPI homolog, DPI-related polypeptide, DPI fusion protein, or fragment in order to identify compounds that modulate the production, degradation, or post- translational modification of the DPI, DPI isoform, DPI homolog, DPI-related polypeptide, DPI fusion protein or fragment.
  • compounds identified in the primary screen can then be assayed in a secondary screen against cells naturally or recombinantly expressing the specific DPI of interest.
  • the ability of the candidate compound to modulate the production, degradation or post-translational modification of a DPI, isoform, homolog, DPI-related polypeptide, or DPI fusion protein can be determined by methods known to those of skill in the art, including without limitation, flow cytometry, a scintillation assay, immunoprecipitation and western blot analysis.
  • agents that competitively interact with (i.e., bind to) a DPI, DPI fragment, DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein are identified in a competitive binding assay.
  • cells expressing a DPI, DPI fragment, DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein are contacted with a candidate compound and a compound known to interact with the DPI, DPI fragment, DPI-related polypeptide, a fragment of a DPI-related polypeptide or a DPI fusion protein; the ability of the candidate compound to competitively interact with the DPI, DPI fragment, DPI-related polypeptide, fragment of a DPI-related polypeptide, or a DPI fusion protein is then determined.
  • agents that competitively interact with (i.e., bind to) a DPI, DPI fragment, DPI-related polypeptide or fragment of a DPI-related polypeptide are identified in a cell-free assay system by contacting a DPI, DPI fragment, DPI-related polypeptide, fragment of a DPI-related polypeptide, or a DPI fusion protein with a candidate compound and a compound known to interact with the DPI, DPI- related polypeptide or DPI fusion protein.
  • the ability of the candidate compound to interact with a DPI, DPI fragment, DPI-related polypeptide, a fragment of a DPI-related polypeptide, or a DPI fusion protein can be determined by methods known to those of skill in the art. These assays, whether cell- based or cell-free, can be used to screen a plurality (e.g., a library) of candidate compounds.
  • agents that modulate i.e., upregulate or downregulate the expression of a DPI, or a DPI-related polypeptide are identified by contacting cells (e.g., cells of prokaryotic origin or eukaryotic origin) expressing the DPI, or DPI-related polypeptide with a candidate compound or a control compound (e.g., phosphate buffered saline (PBS)) and determining the expression of the DPI, DPI-related polypeptide, or DPI fusion protein, mRNA encoding the DPI, or mRNA encoding the DPI-related polypeptide.
  • a candidate compound or a control compound e.g., phosphate buffered saline (PBS)
  • the level of expression of a selected DPI, DPI-related polypeptide, mRNA encoding the DPI, or mRNA encoding the DPI- related polypeptide in the presence of the candidate compound is compared to the level of expression of the DPI, DPI- elated polypeptide, mRNA encoding the DPI, or mRNA encoding the DPI- related polypeptide in the absence of the candidate compound (e.g., in the presence of a control compound).
  • the candidate compound can then be identified as a modulator of the expression of the DPI, or a DPI-related polypeptide based on this comparison.
  • the candidate compound when expression of the DPI or mRNA is significantly greater in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of expression of the DPI or mRNA.
  • the candidate compound when expression of the DPI or mRNA is significantly less in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of the expression of the DPI or mRNA.
  • the level of expression of a DPI or the mRNA that encodes it can be determined by methods known to those of skill in the art. For example, mRNA expression can be assessed by Northern blot analysis or RT-PCR, and protein levels can be assessed by western blot analysis.
  • agents that modulate the activity of a DPI, or a DPI- related polypeptide are identified by contacting a preparation containing the DPI or DPI- related polypeptide, or cells (e.g., prokaryotic or eukaryotic cells) expressing the DPI or DPI-related polypeptide with a test compound or a control compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the DPI or DPI-related polypeptide.
  • the activity of a DPI or a DPI- related polypeptide can be assessed by detecting induction of a cellular signal transduction pathway of the DPI or DPI-related polypeptide (e.g.
  • telomeres intracellular Ca2+, diacylglycerol, IP3, etc.
  • detecting catalytic or enzymatic activity of the target on a suitable substrate detecting the induction of a reporter gene (e.g., a regulatory element that is responsive to a DPI or a DPI-related polypeptide and is operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase)j or detecting a cellular response, for example, cellular differentiation, or cell proliferation.
  • a reporter gene e.g., a regulatory element that is responsive to a DPI or a DPI-related polypeptide and is operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase
  • a cellular response for example, cellular differentiation, or cell proliferation.
  • the candidate compound can then be identified as a modulator of the activity of a DPI or DPI-related polypeptide by comparing the effects of the candidate compound to the control compound.
  • Suitable control compounds include phosphate buffered saline (PBS) and normal saline (NS).
  • agents that modulate i.e., upregulate or downregulate) the expression, activity or both the expression and activity of a DPI or DPI-related polypeptide are identified in an animal model.
  • suitable animals include, but are not limited to, mice, rats, rabbits, monkeys, guinea pigs, dogs and cats.
  • the animal used represent a model of BAD (a number of animal models for psychiatric disorders have had significant value in the search for new treatments and in the study of mechanisms. Most notably, the Porsolt forced swim test model of depression is frequently used in both these contexts (Kirby and Lucki, 1997; Rossetti et al., 1993).
  • test compound or a control compound is administered (e.g., orally, rectally or parenterally such as intraperitoneally or intravenously) to a suitable animal and the effect on the expression, activity or both expression and activity of the DPI is determined.
  • test compound or a control compound is administered (e.g., orally, rectally or parenterally such as intraperitoneally or intravenously) to a suitable animal and the effect on the expression, activity or both expression and activity or both expression and activity of the DPI or DPI-related polypeptide is determined. Changes in the expression of a DPI or DPI-related polypeptide can be assessed by the methods outlined above.
  • a DPI or DPI-related polypeptide is used as a "bait protein" in a two-hybrid assay or three hybrid assay to identify other proteins that bind to or interact with a DPI or DPI-related polypeptide
  • a DPI or DPI-related polypeptide see, e.g., U.S. Patent No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J. Biol. Chem. 268:12046-12054; Bartel et al. (1993) Bio/Techniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and PCT Publication No.
  • binding proteins are also likely to be involved in the propagation of signals by the DPIs of the invention as, for example, upstream or downstream elements of a signaling pathway involving the DPIs of the invention.
  • Table XIII enumerates scientific publications describing suitable assays for detecting or quantifying enzymatic or binding activity of a DPI, a DPI analog, a DPI- related polypeptide, or a fragment of any of the foregoing. Each such reference is hereby incorporated in its entirety.
  • an assay referenced in Table XIII is used in the screens and assays described herein, for example to screen for or identify a compound that modulates the activity of (or that modulates both the expression and activity of) a DPI, DPI analog, or DPI-related polypeptide, a fragment of any of the foregoing or a DPI fusion protein.
  • This invention further provides novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.
  • the invention provides for treatment or prevention of various diseases and disorders by administration of a therapeutic compound.
  • Such compounds include but are not limited to: DPIs, DPI analogs, DPI-related polypeptides and derivatives (including fragments) thereof; antibodies to the foregoing; nucleic acids encoding DPIs, DPI analogs, DPI-related polypeptides and fragments thereof; antisense nucleic acids to a gene encoding a DPI or DPI-related polypeptide; and modulator (e.g., agonists and antagonists) of a gene encoding a DPI or DPI-related polypeptide.
  • An important feature of the present invention is the identification of genes encoding DPIs involved in BAD. BAD can be treated (e.g.
  • one or more antibodies each specifically binding to a DPI are administered alone or in combination with one or more additional therapeutic compounds or treatments. Examples of such treatments include mood stabilizers: lithium, divalproex, carbamazepine, lamotrigine; antidepressants: tricyclic antidepressants (eg.
  • a biological product such as an antibody is allogeneic to the subject to which it is administered.
  • a human DPI or a human DPI- related polypeptide is administered to a human subject for therapy (e.g. to ameliorate symptoms or to retard onset or progression) or prophylaxis.
  • Unipolar depression or BAD is treated or prevented by administration to a subject suspected of having or known to have BAD or to be at risk of developing BAD of a compound that modulates (i.e., increases or decreases) the level or activity (i.e., function) of one or more DPIs ⁇ or the level of one or more DFs ⁇ that are differentially present in the CSF of subjects having BAD compared with CSF of subjects free from BAD.
  • BAD is treated or prevented by administering to a subject suspected of having or known to have BAD or to be at risk of developing BAD a compound that upregulates (i.e., increases) the level or activity (i.e., function) of one or more DPIs « or the level of one or more DFs ⁇ that are decreased in the CSF of subjects having BAD.
  • a compound is administered that downregulates the level or activity (i.e., function) of one or more DPIs -- or the level of one or more DFs ⁇ that are increased in the CSF of subjects having BAD.
  • Examples of such a compound include but are not limited to: DPIs, DPI fragments and DPI-related polypeptides; nucleic acids encoding a DPI, a DPI fragment and a DPI-related polypeptide (e.g., for use in gene therapy); and, for those DPIs or DPI-related polypeptides with enzymatic activity, compounds or molecules known to modulate that enzymatic activity.
  • BAD is also freated or prevented by administration to a subject suspected of having or known to have BAD or to be at risk of developing BAD of a compound that downregulates the level or activity of one or more DPIs ⁇ or the level of one or more DFs ⁇ that are increased in the CSF of subjects having BAD.
  • a compound is administered that upregulates the level or activity of one or more DPIs - - or the level of one or more DFs -- that are decreased in the CSF of subjects having BAD.
  • Examples of such a compound include, but are not limited to, DPI antisense oligonucleotides, ribozymes, antibodies directed against DPIs, and compounds that inhibit the enzymatic activity of a DPI.
  • Other useful compounds e.g., DPI antagonists and small molecule DPI antagonists, can be identified using in vitro assays.
  • therapy or prophylaxis is tailored to the needs of an individual subject.
  • compounds that promote the level or function of one or more DPIs, or the level of one or more DFs are therapeutically or prophylactically administered to a subject suspected of having or known to have BAD, in whom the levels or functions of said one or more DPIs, or levels of said one or more DFs, are absent or are decreased relative to a control or normal reference range.
  • compounds that promote the level or function of one or more DPIs, or the level of one or more DFs are therapeutically or prophylactically administered to a subject suspected of having or known to have BAD in whom the levels or functions of said one or more DPIs, or levels of said one or more DFs, are increased relative to a control or to a reference range.
  • compounds that decrease the level or function of one or more DPIs, or the level of one or more DFs are therapeutically or prophylactically administered to a subject suspected of having or known to have BAD in whom the levels or functions of said one or more DPIs, or levels of said one or more DFs, are increased relative to a control or to a reference range.
  • compounds that decrease the level or function of one or more DPIs, or the level of one or more DFs are therapeutically or prophylactically administered to a subject suspected of having or known to have BAD in whom the levels or functions of said one or more DPIs, or levels of said one or more DFs, are decreased relative to a control or to a reference range.
  • the change in DPI function or level, or DF level, due to the administration of such compounds can be readily detected, e.g., by obtaining a sample (e.g., a sample of CSF, blood or urine or a tissue sample such as biopsy tissue) and assaying in vitro the levels of said DFs or the levels or activities of said DPIs, or the levels of mRNAs encoding said DPIs. or any combination of the foregoing. Such assays can be performed before and after the administration of the compound as described herein.
  • the compounds of the invention include but are not limited to any compound, e.g., a small organic molecule, protein, peptide, antibody, nucleic acid, etc.
  • antidepressants tricyclic antidepressants (eg. Desipramine, chlorimipramine, nortriptyline), selective serotonin reuptake inhibitors (SSRIs including fluoxetine (Prozac), sertraltrine
  • nucleic acids comprising a sequence encoding a DPI, a DPI fragment, DPI-related polypeptide or fragment of a DPI-related polypeptide, are administered to promote DPI function by way of gene therapy.
  • Gene therapy refers to administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acid produces its encoded polypeptide that mediates a therapeutic effect by promoting DPI f nction.
  • the compound comprises a nucleic acid encoding a DPI or fragment or chimeric protein thereof, said nucleic acid being part of an expression vector that expresses a DPI or fragment or chimeric protein thereof in a suitable host.
  • a nucleic acid has a promoter operably linked to the DPI coding region, said promoter being inducible or constitutive (and, optionally, tissue-specific).
  • a nucleic acid molecule is used in which the DPI coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the DPI nucleic acid (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al., 1989, Nature 342:435-438).
  • Delivery of the nucleic acid into a subject may be direct, in which case the subject is directly exposed to the nucleic acid or nucleic acid-carrying vector; this approach is known as in vivo gene therapy.
  • delivery of the nucleic acid into the subject may be indirect, in which case cells are first transformed with the nucleic acid in vitro and then transplanted into the subject; this approach is known as ex vivo gene therapy.
  • the nucleic acid is directly administered in vivo, where it is expressed to produce the encoded product.
  • This can be accomplished by any of numerous methods known in the art, e.g., by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by infection using a defective or attenuated retroviral or other viral vector (see U.S. Patent No.
  • a nucleic acid-ligand complex can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180 dated April 16, 1992 (Wu et al.); WO 92/22635 dated December 23, 1992 (Wilson et al.); WO92/20316 dated November 26, 1992 (Findeis et al.); WO93/14188 dated July 22, 1993 (Clarke et al.), WO 93/20221 dated October 14, 1993 (Young)).
  • the nucleic acid can be introduced infracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al., 1989, Nature 342:435-438).
  • a viral vector that contains a nucleic acid encoding a DPI is used.
  • a retroviral vector can be used (see Miller et al., 1993, Meth. Enzymol. 217:581-599). These retroviral vectors have been modified to delete retroviral sequences that are not necessary for packaging of the viral genome and integration into host cell DNA.
  • the nucleic acid encoding the DPI to be used in gene therapy is cloned into the vector, which facilitates delivery of the gene into a subject.
  • retroviral vectors More detail about retroviral vectors can be found in Boesen et al., 1994, Biotherapy 6:291-302, which describes the use of a retroviral vector to deliver the mdrl gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy.
  • Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al., 1994, J. Clin. Invest. 93:644-651; Kiem et al., 1994, Blood 83:1467- 1473; Salmons and Gunzberg, 1993, Human Gene Therapy 4:129-141; and Grossman and Wilson, 1993 , Curr. Opin. in Genetics and Devel. 3:110-114.
  • Adenoviruses are other viral vectors that can be used in gene therapy. Adenoviruses are especially attractive vehicles for delivering genes to respiratory epithelia. Adenoviruses naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson, 1993, Current Opinion in Genetics and Development 3:499-503 present a review of adenovirus- based gene therapy.
  • Adeno-associated virus has also been proposed for use in gene therapy (Walsh et al., 1993, Proc. Soc. Exp. Biol. Med. 204:289-300; U.S. Patent No. 5,436,146).
  • Another approach to gene therapy involves transferring a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated, transfection, or viral infection.
  • the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a subject.
  • the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell.
  • introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell- mediated gene transfer, spheroplast fusion, etc.
  • Numerous techniques are known in the art for the introduction of foreign genes into cells (see, e.g., Loeffler and Behr, 1993, Meth. Enzymol. 217:599-618; Cohen et al., 1993, Meth. Enzymol.
  • the technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
  • the resulting recombinant cells can be delivered to a subject by various methods known in the art.
  • epithelial cells are injected, e.g., subcutaneously.
  • recombinant skin cells may be applied as a skin graft onto the subject.
  • Recombinant blood cells e.g., hematopoietic stem or progenitor cells
  • the amount of cells envisioned for use depends on the desired effect, the condition of the subject, etc., and can be determined by one skilled in the art.
  • Cells into which a nucleic acid can be introduced for purposes of gene therapy encompass any desired, available cell type, and include but are not limited to neuronal cells, glial cells (e.g., oligodendrocytes or astrocytes), epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granul ⁇ cytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord blood, peripheral blood or fetal liver.
  • glial cells e.g., oligodendrocytes or astrocytes
  • epithelial cells e.g., endothelial cells
  • keratinocytes kerat
  • the cell used for gene therapy is autologous to the subject that is treated.
  • a nucleic acid encoding a DPI is introduced into the cells such that it is expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect.
  • stem or progenitor cells are used. Any stem or progenitor cells which can be isolated and maintained in vitro can be used in accordance with this embodiment of the present invention (see e.g. PCT Publication WO 94/08598, dated April 28, 1994; Stemple and Anderson, 1992, Cell 71:973-985; Rheinwald, 1980, Meth. Cell Bio.
  • the nucleic acid to be introduced for purposes of gene therapy comprises an inducible promoter operably linked to the coding region, such that expression of ithe nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
  • Direct injection of a DNA coding for a DPI may also be performed according to, for example, the techniques described in United States Patent No. 5,589,466.
  • naked DNA i.e., isolated DNA molecules in the absence of Hposomes, cells, or any other material besides a suitable carrier.
  • the injection of DNA encoding a protein and operably linked to a suitable promoter results in the production of the protein in cells near the site of injection and the elicitation of an immune response in the subject to, the protein encoded by the injected DNA.
  • naked DNA comprising (a) DNA encoding a DPI and (b) a promoter are injected into a subject to elicit an immune response to the DPI.
  • BAD is treated or prevented by administration of a compound that antagonizes (inhibits) the level(s) and/or function(s) of one or more DPIs which are elevated in the CSF of subjects having BAD as compared with CSF of subjects free from Unipolar depression or BAD.
  • Compounds useful for this purpose include but are not limited to anti-DPI antibodies (and fragments and derivatives containing the binding region thereof), DPI antisense or ribozyme nucleic acids, and nucleic acids encoding dysfunctional DPIs that are used to "knockout" endogenous DPI function by homologous recombination (see, e.g., Capecchi, 1989, Science 244:1288-1292).
  • Other compounds that inhibit DPI function can be identified by use of known in vitro assays, e.g., assays for the ability of a test compound to inhibit binding of a DPI to another protein or a binding partner, or to inhibit a known DPI function.
  • Such inhibition is assayed in vitro or in cell culture, but genetic assays may also be employed.
  • the Preferred Technology can also be used to detect levels of the DPI before and after the adminisfration of the compound.
  • suitable in vitro or in vivo assays are utilized to determine the effect of a specific compound and whether its administration is indicated for treatment of the affected tissue, as described in more detail below.
  • a compound that inhibits a DPI function is administered therapeutically or prophylactically to a subject in whom an increased CSF level or functional activity of the DPI (e.g., greater than the normal level or desired level) is detected as compared with CSF of subjects free from Unipolar depression or BAD or a predetermined reference range.
  • an increased CSF level or functional activity of the DPI e.g., greater than the normal level or desired level
  • Methods standard in the art can be employed to measure the increase in a DPI level or function, as outlined above.
  • Preferred DPI inhibitor compositions include small molecules, i.e., molecules of 1000 daltons or less. Such small molecules can be identified by the screening methods described herein.
  • DPI expression is inhibited by use of DPI antisense nucleic acids.
  • the present invention provides the therapeutic or prophylactic use of nucleic acids comprising at least six nucleotides that are antisense to a gene or cDNA encoding a DPI or a portion thereof.
  • a DPI "antisense" nucleic acid refers to a nucleic acid capable of hybridizing by virtue of some sequence complementarity to a portion of an RNA (preferably mRNA) encoding a DPI.
  • the antisense nucleic acid may be complementary to a coding and/or noncoding region of an mRNA encoding a DPI.
  • Such antisense nucleic acids have utility as compounds that inhibit DPI expression, and can be used in the treatment or prevention of BAD.
  • the antisense nucleic acids of the invention are double-stranded or single- stranded oligonucleotides, RNA or DNA or a modification or derivative thereof, and can be directly administered to a cell or produced intracellularly by transcription of exogenous, introduced sequences.
  • the invention further provides pharmaceutical compositions comprising an effective amount of the DPI antisense nucleic acids of the invention in a pharmaceutically acceptable carrier, as described infra.
  • the invention provides methods for inhibiting the expression of a DPI nucleic acid sequence in a prokaryotic or eukaryotic cell comprising providing the cell with an effective amount of a composition comprising a DPI antisense nucleic acid of the invention. DPI antisense nucleic acids and their uses are described in detail below.
  • the DPI antisense nucleic acids are of at least six nucleotides and are preferably oligonucleotides ranging from 6 to about 50 oligonucleotides.
  • the oligonucleotide is at least 10 nucleotides, at least 15 nucleotides, at least 100 nucleotides, or at least 200 nucleotides.
  • the oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof and can be single-stranded or double-stranded.
  • the oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone.
  • the oligonucleotide may include other appended groups such as peptides; agents that facilitate transport across the cell membrane (see, e.g., Letsinger et al., 1989, Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al., 1987, Proc. Natl. Acad. Sci. 84:648-652; PCT Publication No. WO 88/09810, published December 15, 1988) or blood-brain barrier (see, e.g., PCT Publication No.
  • a DPI antisense oligonucleotide is provided, preferably of single-stranded DNA.
  • the oligonucleotide may be modified at any position on its structure with substituents generally known in the art.
  • the DPI antisense oligonucleotide may comprise at least one of the following modified base moieties: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5- carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylammomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1- methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2- methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5- methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta- D- mannosylqueos ⁇ n
  • the oligonucleotide comprises at least one modified sugar moiety, e.g., one of the following sugar moieties: arabinose, 2-fluoroarabinose, xylulose, and hexose.
  • the oligonucleotide comprises at least one of the following modified phosphate backbones: a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, a formacetal, or an analog of formacetal.
  • the oligonucleotide is an ⁇ -anomeric oligonucleotide.
  • An ⁇ -anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other (Gautier et al., 1987, Nucl. Acids Res. 15:6625-6641).
  • the oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent.
  • Oligonucleotides of the invention may be synthesized by standard methods known in the art, e.g., by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, etc.).
  • an automated DNA synthesizer such as are commercially available from Biosearch, Applied Biosystems, etc.
  • phosphorothioate oligonucleotides may be synthesized by the method of Stein et al. (1988, Nucl. Acids Res. 16:3209), and methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al., 1988, Proc. Natl. Acad. Sci. USA 85:7448-7451).
  • the DPI antisense nucleic acid of the invention is produced intracellularly by transcription from an exogenous sequence.
  • a vector can be introduced in vivo such that it is taken up by a cell, within which cell the vector or a portion thereof is transcribed, producing an antisense nucleic acid (RNA) of the invention.
  • RNA antisense nucleic acid
  • Such a vector would contain a sequence encoding the DPI antisense nucleic acid.
  • Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA.
  • Such vectors can be constructed by recombinant DNA technology standard in the art.
  • Vectors can be plasmid, viral, or others known in the art, used for replication and expression in mammalian cells.
  • Expression of the sequence encoding the DPI antisense RNA can be by any promoter known in the art to act in mammalian, preferably human, cells. Such promoters can be inducible or constitutive. Examples of such promoters are outlined above.
  • the antisense nucleic acids of the invention comprise a sequence complementary to at least a portion of an RNA transcript of a gene encoding a DPI, preferably a human gene encoding a DPI.
  • absolute complementarity although preferred, is not required.
  • a sequence "complementary to at least a portion of an RNA,” as referred to herein, means a sequence having sufficient complementarity to be able to hybridize under stringent conditions (e.g., highly stringent conditions comprising hybridization in 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65 C and washing in O.lxSSC/0.1% SDS at 68 °C, or moderately stringent conditions comprising washing in 0.2xSSC/0.1 % SDS at 42 °C ) with the RNA, forming a stable duplex; in the case of double-stranded DPI antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed.
  • stringent conditions e.g., highly stringent conditions comprising hybridization in 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65 C and washing in O.lxSSC/0.1% SDS
  • the ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid. Generally, the longer the hybridizing nucleic acid, the more base mismatches with an RNA encoding a DPI it may contain and still form a stable duplex (or triplex, as the case may be).
  • One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex.
  • the DPI antisense nucleic acids can be used to treat or prevent BAD when the target DPI is overexpressed in the CSF of subjects suspected of having or suffering from BAD.
  • a single-stranded DNA antisense DPI oligonucleotide is used.
  • Cell types which express or overexpress RNA encoding a DPI can be identified by various methods known in the art. Such cell types include but are not limited to leukocytes (e.g., neutrophils, macrophages, monocytes) and resident cells (e.g., astrocytes, glial cells, neuronal cells, and ependymal cells).
  • Such methods include, but are not limited to, hybridization with a DPI-specific nucleic acid (e.g., by Northern hybridization, dot blot hybridization, in situ hybridization), observing the ability of RNA from the cell type to be translated in vitro into a DPI, immunoassay, etc.
  • primary tissue from a subject can be assayed for DPI expression prior to treatment, e.g., by immunocytochemistry or in situ hybridization.
  • compositions of the invention comprising an effective amount of a DPI antisense nucleic acid in a pharmaceutically acceptable carrier, can be administered to a subject having BAD.
  • the amount of DPI antisense nucleic acid which will be effective in the treatment of BAD can be determined by standard clinical techniques.
  • compositions comprising one or more DPI antisense nucleic acids are administered via Hposomes, microparticles, or microcapsules.
  • such compositions may be used to achieve sustained release of the DPI antisense nucleic acids.
  • symptoms of BAD may be ameliorated by decreasing the level of a DPI or DPI activity by using gene sequences encoding the DPI in conjunction with well-known gene "knock-out,” ribozyme or triple helix methods to decrease gene expression of a DPI.
  • ribozyme or triple helix molecules are used to modulate the activity, expression or synthesis of the gene encoding the DPI, and thus to ameliorate the symptoms of BAD.
  • Such molecules may be designed to reduce or inhibit expression of a mutant or non-mutant target gene. Techniques for the production and use of such molecules are well known to those of skill in the art.
  • Ribozyme molecules designed to catalytically cleave gene mRNA transcripts encoding a DPI can be used to prevent translation of target gene mRNA and, therefore, expression of the gene product.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage event.
  • the composition of ribozyme molecules must include one or more sequences complementary to the target gene mRNA, and must include the well known catalytic sequence responsible for mRNA cleavage. For this sequence, see, e.g., U.S. Patent No. 5,093,246, which is incorporated herein by reference in its entirety.
  • ribozymes that cleave mRNA at site specific recognition sequences can be used to destroy mRNAs encoding a DPI
  • the use of hammerhead ribozymes is preferred.
  • Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA. The sole requirement is that the target mRNA have the following sequence of two bases: 5'- UG-3'.
  • ribozymes are engineered so that the cleavage recognition site is located near the 5' end of the mRNA encoding the DPI, i.e., to increase efficiency and minimize the intracellular accumulation of non-functional mRNA transcripts.
  • the ribozymes of the present invention also include RNA endoribonucleases (hereinafter "Cech-type ribozymes”) such as the one that occurs naturally in Tetrahymena thermophila (known as the IVS, or L-19 IVS RNA) and that has been extensively described by Thomas Cech and collaborators (Zaug, et al., 1984, Science, 224, 574-578; Zaug and Cech, 1986, Science, 231, 470-475; Zaug, et al., 1986, Nature, 324, 429-433; published International patent application No. WO 88/04300 by University Patents Inc.; Been and Cech, 1986, Cell, 47, 20,7-216).
  • Cech-type ribozymes such as the one that occurs naturally in Tetrahymena thermophila (known as the IVS, or L-19 IVS RNA) and that has been extensively described by Thomas Cech and collaborators (Zaug, et al., 1984, Science, 224,
  • the Cech-type ribozymes have an eight base pair active site which hybridizes to a target RNA sequence whereafter cleavage of the target RNA takes place.
  • the invention encompasses those Cech-type ribozymes which target eight base-pair active site sequences that are present in the gene encoding the DPI.
  • the ribozymes can be composed of modified oligonucleotides (e.g., for improved stability, targeting, etc.) and should be delivered to cells that express the DPI in vivo.
  • a preferred method of delivery involves using a DNA construct "encoding" the ribozyme under the confrol of a strong constitutive pol III or pol II promoter, so that transfected cells will produce sufficient quantities of the ribozyme to destroy endogenous mRNA encoding the DPI and inhibit translation. Because ribozymes, unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficacy.
  • Endogenous DPI expression can also be reduced by inactivating or "knocking out” the gene encoding the DPI, or the promoter of such a gene, using targeted homologous recombination (e.g., see Smithies, et al., 1985, Nature 317:230-234; Thomas and Capecchi, 1987, Cell 51:503-512; Thompson et al., 1989, Cell 5:313- 321; and Zijlstra et al., 1989, Nature 342:435-438, each of which is incorporated by reference herein in its entirety).
  • targeted homologous recombination e.g., see Smithies, et al., 1985, Nature 317:230-234; Thomas and Capecchi, 1987, Cell 51:503-512; Thompson et al., 1989, Cell 5:313- 321; and Zijlstra et al., 1989, Nature 342:435-438, each of which is incorporated by reference herein in its entirety).
  • a mutant gene encoding a nonfunctional DPI (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous gene (either the coding regions or regulatory regions of the gene encoding the DPI) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express the target gene in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the target gene.
  • Such approaches are particularly suited in the agricultural field where modifications to ES (embryonic stem) cells can be used to generate animal offspring with an inactive target gene (e.g., see Thomas and Capecchi, 1987 and Thompson, 1989, supra).
  • the endogenous expression of a gene encoding a DPI can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of the gene (i.e., the gene promoter and/or enhancers) to form triple helical structures that prevent transcription of the gene encoding the DPI in target cells in the body.
  • deoxyribonucleotide sequences complementary to the regulatory region of the gene i.e., the gene promoter and/or enhancers
  • triple helical structures that prevent transcription of the gene encoding the DPI in target cells in the body.
  • Nucleic acid molecules to be used in triplex helix formation for the inhibition of transcription should be single stranded and composed of deoxynucleotides.
  • the base composition of these oligonucleotides must be designed to promote triple helix formation via Hoogsteen base pairing rules, which generally require sizeable sfretches of either purines or pyrimidines to be present on one strand of a duplex.
  • Nucleotide sequences may be pyrimidine-based, which will result in TAT and CGC+ triplets across the three associated strands of the resulting triple helix.
  • the pyrimidine-rich molecules provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand.
  • nucleic acid molecules may be chosen that are purine-rich, for example, contain a stretch of G residues. These molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting in GGC triplets across the three strands in the triplex.
  • the potential sequences that can be targeted for triple helix formation may be increased by creating a so called "switchback" nucleic acid molecule.
  • Switchback molecules are synthesized in an alternating 5'-3', 3'-5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • the technique may so efficiently reduce or inhibit the transcription (triple helix) or translation (antisense, ribozyme) of mRNA produced by normal gene alleles of a DPI that the situation may arise wherein the concentration of DPI present may be lower than is necessary for a normal phenotype.
  • gene therapy may be used to introduce into cells nucleic acid molecules that encode and express the DPI that exhibit normal gene activity and that do not contain sequences susceptible to whatever antisense, ribozyme, or triple helix treatments are being utilized.
  • Antisense RNA and DNA, ribozyme, and triple helix molecules of the invention may be prepared by any method known in the art for the synthesis of DNA and RNA molecules, as discussed above. These include techniques for chemically synthesizing ohgodeoxyribonucleotides and ohgoribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule.
  • DNA sequences may be incorporated into a wide variety of vectors that incorporate suitable RNA poiymerase promoters such as the T7 or SP6 poiymerase promoters.
  • RNA poiymerase promoters such as the T7 or SP6 poiymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines. 5.15 Assays For Therapeutic Or Prophylactic Compounds
  • the present invention also provides assays for use in drug discovery in order to identify or verify the efficacy of compounds for treatment or prevention of BAD.
  • Test compounds can be assayed for their ability to restore DF or DPI levels in a subject having BAD towards levels found in subjects free from BAD or to produce similar changes in experimental animal models of BAD.
  • Compounds able to restore DF or DPI levels in a subject having BAD towards levels found in subjects free from BAD or to produce similar changes in experimental animal models of BAD can be used as lead compounds for further drug discovery, or used therapeutically.
  • DF and DPI expression can be assayed by the Preferred Technology, immunoassays, gel electrophoresis followed by visualization, detection of DPI activity, or any other method taught herein or known to those skilled in the art.
  • Such assays can be used to screen candidate drugs, in clinical monitoring or in drug development, where abundance of a DF or DPI can serve as a surrogate marker for clinical disease.
  • in vitro assays can be carried out with cells representative of cell types involved in a subject's disorder, to determine if a compound has a desired effect upon such cell types.
  • Compounds for use in therapy can be tested in suitable animal model systems prior to testing in humans, including but not limited to rats, mice, chicken, cows, monkeys, rabbits, etc.
  • suitable animal model systems prior to adminisfration to humans, any animal model system known in the art may be used.
  • animal models of BAD include, but are not limited to, animals that express human familial BAD genes and the Porsolt forced swim test model of depression is frequently used in both these contexts (Kirby and Lucki, 1997; Rossetti et al., 1993).
  • the two major clinical states observed in bipolar disorder (depression and mania) have also been successfully modeled (Cappeliez and Moore Prog Neuropsychopharmacol Biol Psychiatry 1990 14, 347-58).
  • Psychostimulant treatment can produce a range of behaviors similar to that of mania including hyperactivity, heightened sensory awareness, and alertness, and for this reason has become a very useful model for mania which exhibits (to some extent) face, construct and predictive validity.
  • Another model that has been utilized for the development of bipolar illness is behavioral sensitization, In this model, the repeated administration of many psychostimulant drugs leads to a gradual increase or sensitization of the drug-induced behavioral; this model also has considerable construct and face validity for mania (Koob et al. Pharmacol Biochem Behav 1997 57, 513-21)), which can be utilised to test compounds that modulate DF or DPI levels.
  • transgenic animals can be produced with "knock-out” mutations of the gene or genes encoding one or more DPIs.
  • a "knock-out" mutation of a gene is a mutation that causes the mutated gene to not be expressed, or expressed in an aberrant form or at a low level, such that the activity associated with the gene product is nearly or entirely absent.
  • the transgenic animal is a mammal, more preferably, the transgenic animal is a mouse.
  • test compounds that modulate the expression of a DPI are identified in non-human animals (e.g., mice, rats, monkeys, rabbits, and guinea pigs), preferably non-human animal models for BAD, expressing the DPI.
  • non-human animals e.g., mice, rats, monkeys, rabbits, and guinea pigs
  • a test compound or a control compound is administered to the animals, and the effect of the test compound on expression of one or more DPIs is determined.
  • a test compound that alters the expression of a DPI can be identified by comparing the level of the selected DPI or DPIs (or mRNA(s) encoding the same) in an animal or group of animals treated with a test compound with the level of the DPI(s) or rnRNA(s) in an animal or group of animals treated with a control compound.
  • Techniques known to those of skill in the art can be used to determine the mRNA and protein levels, for example, in situ hybridization. The animals may or may not be sacrificed to assay the effects of a test compound.
  • test compounds that modulate the activity of a DPI or a biologically active portion thereof are identified in non-human animals (e.g., mice, rats, monkeys, rabbits, and guinea pigs), preferably non-human animal models for BAD, expressing the DPI.
  • non-human animals e.g., mice, rats, monkeys, rabbits, and guinea pigs
  • a test compound or a confrol compound is administered to the animals, and the effect of a test compound on the activity of a DPI is determined.
  • a test compound that alters the activity of a DPI can be identified by assaying animals treated with a control compound and animals treated with the test compound.
  • the activity of the DPI can be assessed by detecting induction of a cellular second messenger of the DPI (e.g., intracellular Ca2+, diacylglycerol, IP3, etc.), detecting catalytic or enzymatic activity of the DPI or binding partner thereof, detecting the induction of a reporter gene (e.g., a regulatory element that is responsive to a DPI of the invention operably linked to a nucleic acid encoding a detectable marker, such as luciferase or green fluorescent protein), or detecting a cellular response (e.g., cellular differentiation or cell proliferation).
  • a reporter gene e.g., a regulatory element that is responsive to a DPI of the invention operably linked to a nucleic acid encoding a detectable marker, such as luciferase or green fluorescent protein
  • a cellular response e.g., cellular differentiation or cell proliferation.
  • test compounds that modulate the level or expression of a DPI are identified in human subjects having BAD, preferably those having mild to severe BAD and most preferably those having mild BAD.
  • a test compound or a control compound is administered to the human subject, and the effect of a test compound on DPI expression is determined by analyzing the expression of the DPI or the mRNA encoding the same in a biological sample (e.g., CSFj serum, plasma, or urine).
  • a biological sample e.g., CSFj serum, plasma, or urine.
  • a test compound that alters the expression of a DPI can be identified by comparing the level of the DPI or mRNA encoding the same in a subject or group of subjects treated with a control compound to that in a subject or group of subjects freated with a test compound.
  • alterations in the expression of a DPI can be identified by comparing the level of the DPI or mRNA encoding the same in a subject or group of subjects before and after the administration of a test compound.
  • Techniques known to those of skill in the art can be used to obtain the biological sample and analyze the mRNA or protein expression. For example, the Preferred Technology described herein can be used to assess changes in the level of a DPI.
  • test compounds that modulate the activity of a DPI are identified in human subjects having BAD, preferably those having mild to severe BAD and most preferably those with mild BAD.
  • a test compound or a control compound is administered to the human subject, and the effect of a test compound on the activity of a DPI is determined.
  • a test compound that alters the activity of a DPI can be identified by comparing biological samples from subjects treated with a control compound to samples from subjects treated with the test compound.
  • alterations in the activity of a DPI can be identified by comparing the activity of a DPI in a subject or group of subjects before and after the administration of a test compound.
  • the activity of the DPI can be assessed by detecting in a biological sample (e.g., CSF, serum, plasma, or urine) induction of a cellular signal transduction pathway of the DPI (e.g., intracellular Ca2+, diacylglycerol, IP3, etc.), catalytic or enzymatic activity of the DPI or a binding partner thereof, or a cellular response, for example, cellular differentiation, or cell proliferation.
  • a biological sample e.g., CSF, serum, plasma, or urine
  • a cellular signal transduction pathway of the DPI e.g., intracellular Ca2+, diacylglycerol, IP3, etc.
  • catalytic or enzymatic activity of the DPI or a binding partner thereof e.g., intracellular Ca2+, diacylglycerol, IP3, etc.
  • a cellular response for example, cellular differentiation, or cell proliferation.
  • Techniques known to those of skill in the art can be used to detect changes in the in
  • a test compound that changes the level or expression of a DPI towards levels detected in confrol subjects is selected for further testing or therapeutic use.
  • a test compound that changes the activity of a DPI towards the activity found in control subjects is selected for further testing or therapeutic use.
  • test compounds that reduce the severity of one or more symptoms associated with BAD are identified in human subjects having BAD, preferably subjects having mild to severe BAD and most preferably subjects with mild BAD.
  • a test compound or a control compound is administered to the subjects, and the effect of a test compound on one or more symptoms of BAD is determined.
  • a test compound that reduces one or more symptoms can be identified by comparing the subjects freated with a control compound to the subjects treated with the test compound. Techniques known to physicians familiar with BAD can be used to determine whether a test compound reduces one or more symptoms associated with BAD. For example, a test compound that enhances memory or reduces confusion in a subject having BAD will be beneficial for treating subjects having BAD.
  • a test compound that reduces the severity of one or more symptoms associated with BAD in a human having BAD is selected for further testing or therapeutic use.
  • the invention provides methods of treatment (and prophylaxis) comprising administering to a subject an effective amount of a compound of the invention.
  • the compound is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects).
  • the subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human. In a specific embodiment, a non-human mammal is the subject.
  • Formulations and methods of administration that can be employed when the compound comprises a nucleic acid are described above; additional appropriate formulations and routes of adminisfration are described below.
  • a compound of the invention e.g., encapsulation in Hposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor- mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction can be'enteral or parenteral and include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, infranasal, epidural, and oral routes.
  • the compounds may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • Pulmonary adminisfration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • compositions of the invention may be desirable to administer locally to the area in need of treatment; this may be achieved, for example, and not by way of limitation, by local infusion during surgery, topical application, e.g., by injection, by means of a catheter, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • administration can be by direct injection into CSF or at the site (or former site) of neurodegeneration or to CNS tissue.
  • the compound in another embodiment, can be delivered in a vesicle, in particular a Hposome (see Langer, 1990, Science 249:1527-1533; Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317- 327; see generally ibid.)
  • the compound can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng.
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailabihty, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., 1983, Macromol. Sci. Rev. Macromol. Chem. 23:61; see also Levy et al., 1985, Science 228:190; During et al., 1989, Ann.
  • a controlled release system can be placed in proximity of the therapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Patent No.
  • a nucleic acid can be introduced infracellularly and incorporated within host cell DNA for expression, by homologous recombination.
  • compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin.
  • Such compositions will contain a therapeutically effective amount of - the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject.
  • the formulation should suit the mode of administration.
  • composition is formulated in accordance with
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as Hdocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • compositions are administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to adminisfration.
  • the compounds of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine, etc.
  • the amount of the compound of the invention which will be effective in the treatment of BAD can be determined by standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances.
  • suitable dosage ranges for intravenous administration are generally about 20- 500 micrograms of active compound per kilogram body weight.
  • Suitable dosage ranges for infranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • Suppositories generally contain active ingredient in the range of 0.5% to 10% by weight; oral formulations preferably contain 10% to 95% active ingredient.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects (a) approval by the agency of manufacture, use or sale for human administration, (b) directions for use, or both.
  • CSF depletion Removal of albumin, haptoglobin, transferrin and immunoglobin G (IgG) from CSF ("CSF depletion") was achieved by an affinity chromatography purification step in which the sample was passed through a series of 'Hi-Trap' columns containing immobilized antibodies for selective removal of albumin, haptoglobin and transferrin, and protein G for selective removal of immunoglobin G.
  • Two affinity columns in a tandem assembly were prepared by coupling antibodies to protein G-sepharose contained in Hi-Trap columns (Protein G-Sepharose Hi-Trap columns (1 ml) Pharmacia Cat. No. 17-0404-01).
  • the chromatographic procedure was automated using an Akta Fast Protein Liquid Chromatography (FPLC) System such that a series of up to seven runs could be performed sequentially.
  • the samples were passed through the series of 3 Hi-Trap columns in which the affinity chromatography media selectively bind the above proteins thereby removing them from the sample.
  • Fractions typically 3 ml per tube
  • Flowthrough fractions unbound material
  • Bound/Eluted fractions bound proteins
  • the eluate containing unbound material was collected in fractions which were pooled, desalted/concentrated by centrifugal ulfrafiltration and stored to await further analysis by 2D PAGE.
  • a volume of depleted CSF containing approximately 100-150 ⁇ g of total protein was aliquoted and an equal volume of 10% (w/v) SDS (Fluka 71729), 2.3%) (w/v) dithiothreitol (BDH 443852A) was added.
  • the sample was heated at 95 °C for 5 mins, and then allowed to cool to 20 °C .
  • 125 ⁇ l of the following buffer was then added to the sample:
  • Isoelectric focusing was performed using the Immobiline® DryStrip Kit (Pharmacia BioTech), following the procedure described in the manufacturer's instructions, see Instructions for Immobiline® DryStrip Kit, Pharmacia, # 18-1038- 63, Edition AB (incorporated herein by reference in its entirety).
  • Immobilized pH Gradient (IPG) sfrips (18cm, pH 3-10 non-linear strips; Pharmacia Cat. # 17-1235-01) were rehydrated overnight at 20 °C in a solution of 8M urea, 2% (w/v) CHAPS, lOmM DTT, 2% (v/v) Resolytes 3.5-10, as described in the Immobiline DryStrip Users Manual.
  • the strips were immediately removed and immersed for 10 mins at 20 °C in a first solution of the following composition: 6M urea; 2% (w/v) DTT; 2% (w/v) SDS; 30% (v/v) glycerol (Fluka 49767); 0.05M Tris/HCl, pH 6.8 (Sigma Cat T-l 503).
  • the strips were removed from the first solution and immersed for 10 mins at 20 °C in a second solution of the following composition: 6M urea; 2% (w/v) iodoacetamide (Sigma 1-6125); 2% (w/v) SDS; 30% (v/v) glycerol; 0.05M Tris/HCl, pH 6.8.
  • the strips were loaded onto supported gels for SDS-PAGE according to Hochstrasser et al. ⁇ 1988, Analytical Biochemistry 173: 412-423 (incorporated herein by reference in its entirety), with modifications as specified below.
  • the gels were cast between two glass plates of the following dimensions: 23cm wide x 24cm long (back plate); 23cm wide x 24cm long with a 2cm deep notch in the central 19cm (front plate).
  • the back plate was treated with a 0.4% solution of ⁇ -methacryl- oxypropyltrimethoxysilane in ethanol (BindSilaneTM; Pharmacia Cat. # 17-1330-01).
  • the front plate was treated with (RepelSilaneTM Pharmacia Cat. # 17-1332-01) to reduce adhesion of the gel. Excess reagent was removed by washing with water, and the plates were allowed to dry..
  • an adhesive bar-code was attached to the back plate in a position such that it would not come into contact with the gel matrix.
  • the dried plates were assembled into a casting box with a capacity of 13 gel sandwiches.
  • the front and back plates of each sandwich were spaced by means of lmm thick spacers, 2.5 cm wide.
  • the sandwiches were interleaved with acetate sheets to facilitate separation of the sandwiches after gel polymerization. Casting was then carried out according to Hochstrasser et al., op. cit.
  • a 9- 16% linear polyacrylamide gradient was cast, extending up to a point 2cm below the level of the notch in the front plate, using the Angelique gradient casting system (Large Scale Biology).
  • Stock solutions were as follows. Acrylamide (40% in water) was from Serva (Cat. # 10677).
  • the cross-linking agent was PDA (BioRad 161- 0202), at a concentration of 2.6% (w/w) of the total starting monomer content.
  • the gel buffer was 0.375M Tris/HCl, pH 8.8.
  • the polymerization catalyst was 0.05% (v/v) TEMED (BioRad 161-0801), and the initiator was 0.1% (w/v) APS (BioRad 161-0700). No SDS was included in the gel and no stacking gel was used.
  • the cast gels were allowed to polymerize at 20 °C overnight, and then stored individually at 4 °C in sealed polyethylene bags with 6ml of gel buffer, and were used within 4
  • a solution of 0.5% (w/v) agarose (Fluka Cat 05075) was prepared in running buffer (0.025M Tris, 0.198M glycine (Fluka 50050), 1% (w/v) SDS, supplemented by a trace of bromophenol blue).
  • the agarose suspension was heated to 70 °C with stirring, until the agarose had dissolved.
  • the top of the supported 2nd D gel was filled with the agarose solution, and the equilibrated strip was placed into the agarose, and tapped gently with a palette knife until the gel was intimately in contact with the 2nd D gel.
  • the gels were placed in the 2nd D running tank, as described by Amess et al., 1995, Electrophoresis 16: 1255-1267 (incorporated herein by reference in its entirety).
  • the tank was filled with running buffer (as above) until the level of the buffer was just higher than the top of the region of the 2nd D gels which contained polyacrylamide, so as to achieve efficient cooling of the active gel area.
  • Running buffer was added to the top buffer compartments formed by the gels, and then voltage was applied immediately to the gels using a Consort E-833 power supply. For 1 hour, the gels were run at 20mA/gel.
  • the wattage limit was set to 150W for a tank containing 6 gels, and the voltage limit was set to 600V.
  • the gels were then run at 40mA/gel, with the same voltage and wattage limits as before, until the bromophenol blue line was 0.5cm from the bottom of the gel.
  • the temperature of the buffer was held at 16 °C throughout the run. Gels were not run in duplicate.
  • the gels were immediately removed from the tank for fixation.
  • the top plate of the gel cassette was carefully removed, leaving the gel bonded to the bottom plate.
  • the bottom plate with its attached gel was then placed into a staining apparatus, which can accommodate 12 gels.
  • the gels were completely immersed in fixative solution of 40% (v/v) ethanol (BDH 28719), 10% (v/v) acetic acid (BDH 100016X), 50% (v/v) water (MilliQ- Millipore), which was continuously circulated over the gels.
  • the fixative was drained from the tank, and the gels were primed by immersion in 7.5% (v/v) acetic acid, 0.05% (w/v) SDS, 92.5% (v/v) water for 30 mins.
  • the priming solution was then drained, and the gels were stained by complete immersion for 4 hours in a staining solution of Pyridinium, 4-[2-[4-(dipentylamino)- 2-trifluoromethylphenyl] ethenyl]-l- (sulfobutyl)-, inner salt, prepared by diluting a stock solution of this dye (2mg/ml in DMSO) in 7.5% (v/v) aqueous acetic acid to give a final concenfration of 1.2 mg/l; the staining solution was vacuum filtered through a 0.4 ⁇ m filter (Duropore) before use.
  • a staining solution of Pyridinium, 4-[2-[4-(dipentylamino)- 2-trifluoromethylphenyl] ethenyl]-l- (sulfobutyl)-, inner salt prepared by diluting a stock solution of this dye (2mg/ml in DMSO) in 7.5%
  • a computer-readable output was produced by imaging the fluorescently stained gels with the Apollo 2 scanner (Oxford Glycosciences, Oxford, UK) described in section 5.1, supra.
  • This scanner has a gel carrier with four integral fluorescent markers (Designated Ml, M2, M3) that are used to correct the image geometry and are a quality control feature to confirm that the scanning has been performed correctly.
  • the gels were removed from the stain, rinsed with water and allowed to air dry briefly, and imaged on the Apollo 2. After imaging, the gels were sealed in polyethylene bags containing a small volume of staining solution, and then stored at 4 °C.
  • the output from the scanner was first processed using the MELANIE® II 2D
  • M4 to autocrop the images (i.e., to eliminate signals originating from areas of the scanned image lying outside the boundaries of the gel, e.g. the reference frame); to filter out artifacts due to dust; to detect and quantify features; and to create image files in GIF format.
  • Features were detected using the following parameters:
  • Landmark identification was used to determine the pi and MW of features detected in the images. Twelve landmark features, designated CSFl to CSF12, were identified in a standard CSF image obtained from a pooled sample. These landmark features are identified in Figure 1 and were assigned the pi and/or MW values identified in Table XIV.
  • each feature in the study gels was then assigned a pi value by linear interpolation or extrapolation (using the MELANIE®-II software) to the two nearest landmarks, and was assigned a MW value by linear interpolation or extrapolation (using the MELANIE®-II software) to the two. nearest landmarks.
  • 6.1.10 Matching With Primary Master Image Images were edited to remove gross artifacts such as dust, to reject images which had gross abnormalities such as smearing of protein features, or were of too low a loading or overall image intensity to allow identification of more than the most intense features, or were of too poor a resolution to allow accurate detection of features. Images were then compared by pairing with one common image from the whole sample set. This common image, the "primary master image", was selected on the basis of protein load (maximum load consistent with maximum feature detection), a well resolved myoglobin region, (myoglobin was used as an internal standard), and general image quality. Additionally, the primary master image was chosen to be an image which appeared to be generally representative of all those to be included in the analysis.
  • each study gel was adjusted for maximum alignment between its pattern of protein features, and that of the primary master, as follows.
  • Each of the study gel images was individually transformed into the geometry of the primary master image using a multi-resolution warping procedure. This procedure corrects the image geometry for the distortions brought about by small changes in the physical parameters of the electrophoresis separation process from one sample to another. The observed changes are such that the distortions found are not simple geometric distortions, but rather a smooth flow, with variations at both local and global scale.
  • the fundamental principle in multi-resolution modeling is that smooth signals may be modeled as an evolution through 'scale space', in which details at successively finer scales are added to a low resolution approximation to obtain the high resolution signal.
  • This type of model is applied to the flow field of vectors (defined at each pixel position on the reference image) and allows flows of arbitrary smoothness to be modeled with relatively few degrees of freedom.
  • Each image is first reduced to a stack, or pyramid, of images derived from the initial image, but smoothed and reduced in resolution by a factor of 2 in each direction at every level (Gaussian pyramid) and a corresponding difference image is also computed at each level, representing the difference between the smoothed image and its progenitor (Laplacian pyramid).
  • the Laplacian images represent the details in the image at different scales.
  • the warping process brought about good alignment between the common features in the primary master image, and the images for the other samples.
  • the MELANIE® II 2D PAGE analysis program was used to calculate and record approximately 500-700 matched feature pairs between the primary master and each of the other images.
  • the accuracy of this program was significantly enhanced by the alignment of the images in the manner described above.
  • all pairings were finally examined by eye in the MelView interactive editing program and residual recognizably incorrect pairings were removed. Where the number of such recognizably incorrect pairings exceeded the overall reproducibility of the Preferred Technology (as measured by repeat analysis of the same biological sample) the gel selected to be the primary master gel was judged to be insufficiently representative of the study gels to serve as a primary master gel.
  • the gel chosen as the primary master gel was rejected, and different gel was selected as the primary master gel, and the process was repeated. All the images were then added together to create a composite master image, and the positions and shapes of all the gel features of all the component images were super- imposed onto this composite master as described below.
  • a composite master image was thus generated by initialising the primary master image with its feature descriptors. As each image was transformed into the primary master geometry, it was digitally summed pixel by pixel into the composite master image, and the features that had not been paired by the procedure outlined above were likewise added to the composite master image description, with their cenfroids adjusted to the master geometry using the flow field correction.
  • MCI molecular cluster index
  • LIMS Laboratory Information Management System
  • a second non-overlapping selection strategy is based on the fold change.
  • a fold change representing the ratio of the average normalized protein abundances of the DFs within an MCI was calculated for each MCI between each set of confrols and BAD samples.
  • An 80% confidence limit for the mean of the fold changes was calculated.
  • the MCIs with fold changes which fall outside the confidence limit were selected as DFs which met the criteria of the significant fold change threshold with . 80% selectivity. Because the MCI fold changes are based on an 80% confidence limit, it follows that the significant fold change threshold is itself 80%.
  • a third non-overlapping selection strategy is based on qualitative presence or absence alone.
  • Proteins in DFs were robotically excised and processed to generate tryptic digest peptides. Tryptic peptides were analyzed by mass spectrometry using a . PerSeptive Biosystems Voyager- DETM STR Matrix- Assisted Laser Desorption Ionization Time-of- Flight (MALDI-TOF) mass spectrometer, and selected tryptic peptides were analyzed by tandem mass spectrometry (MS/MS) using a Micromass Quadrupole Time-of-Flight (Q-TOF) mass spectrometer (Micromass, Altrincham, U.K.) equipped with a nanoflowTM elecfrospray Z-spray source.
  • MALDI-TOF Micromass Quadrupole Time-of-Flight
  • the database searched was database constructed of protein entries in the non-redundant database held by the National Centre for Biotechnology Information (NCBI) which is accessible at http://www.ncbi.nlm.nih.gov/.
  • NCBI National Centre for Biotechnology Information
  • masses detected in MALDI-TOF mass spectra were assigned to tryptic digest peptides within the proteins identified.
  • tandem mass spectra of the peptides were interpreted manually, using methods known in the art. (In the case of interpretation of low-energy fragmentation mass spectra of peptide ions see Gaskell et al., 1992, Rapid Commun. Mass Spectrom. 6:658-662)
  • the following example illustrates the use of a DPI of the invention for screening or diagnosis of BAD, determining the prognosis of a BAD patient, or monitoring the effectiveness of BAD therapy.
  • the following example also illustrates the use of modulators (e.g., agonist or antagonists) of a DPI of the invention to treat or prevent BAD.
  • Dickkopf-3 belongs to a family a secreted glycoproteins that antagonise the Wnt signalling pathway.
  • Wnts are a large family of cysteine-rich, secreted glycoproteins, which bind to frizzled seven-fransmembrane-span receptors, and regulate cell fate and embryonic patterning (Eastmann and Grosschedl, Regulation of LEF-1/TCF transcription factors by Wnt and other signals, Curr Opin Cell Biol 1999 Apr 11:2 233-40).
  • beta- catenin is phosphorylated by glycogen synthase kinase3beta (GSK3beta), which triggers ubiquitination of beta-catenin by betaTrCP and degradation in proteasomes. Phosphorylation of beta-catenin occurs in a multiprotein complex assembled by the scaffolding protein axin or conductin. Wnt binding to its receptor Frizzled leads to activation of the Dishevelled protein (Klingensmith J, Nusse R, Perrimon N: The Drosophila segment polarity gene dishevelled encodes a novel protein required for response to the wingless signal.
  • GSK3beta glycogen synthase kinase3beta
  • GSK phosphorylation blocks its ability to phosphorylate beta-catenin, leading to increased stability and accumulation (Munemitsu S, Albert I, Rubinfeld B, Polakis P, Deletion of an amino-terminal sequence beta-catenin in vivo and promotes hyperphosporylation of the adenomatous polyposis coli tumor suppressor protein, Mol Cell Biol 1996 Aug 16:8 4088-94.; Pai LM, Orsulic S, Bejsovec A, Peifer M, Negative regulation of Armadillo, a Wingless effector in Drosophila, Development 1997 Jun 124:11 2255-66; Yost C, Torres M, Miller JR, Huang E, Kimelman D, Moon RT, The axis-inducing activity, stability, and subcellular distribution of beta-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3, Genes Dev 1996 Jun 15 10:12 1443-54).
  • beta-catenin can interact with members of T cell factor (TCF)/lymphoid enhancer factor (LCF) family in the nucleus, which regulate Wnt target genes (Wodarz A, Nusse R, Mechanisms of Wnt signaling in development, Annu Rev Cell Dev Biol 1998 14: 59-88).
  • TCF T cell factor
  • LCF lymphoid enhancer factor
  • the human Dkk gene family is composed of Dkk-1, Dkk-2, Dkk-3, Dkk-4 and a unique Dkk-3 related protein termed Soggy (Sgy) (Krupnik VE, Sharp JD, Jiang C, Robison K, Chickering TW, Amaravadi L, Brown DE, Guyot D, Mays G, Leiby K, Chang B, Duong T, Goodearl AD, Gearing DP, Sokol SY, McCarthy SA, Functional and structural diversity of the human Dickkopf gene family, Gene 1999 Oct 1 238:2 301-13).
  • Soggy Soggy
  • Dkk-3 mRNA is highly expressed in brain and heart and low levels can be detected in spleen, kidney, liver, small intestine, placenta and lung (Tsuji T, Miyazaki M, Sakaguchi M, Inoue Y, Namba M, A REIC gene shows down-regulation in human immortalized cells and human tumor-derived cell lines, Biochem Biophys Res Commun 2000 Feb 5 268:1 20-4).
  • Murine Dkk-3 mRNA is expressed in neurones of the cortex and hippocampus (Krupnik et al., supra).
  • beta-catenin degradation is modulated by the casein kinase CK1 and by the protein phosphatases PP2A and PP2C.
  • Stabilised beta- catenin enters the cell nucleus and associates with TCF transcription factors, which leads to the transcription of Wnt-target genes.
  • Smad4, Tsh, XSoxl7 and the histone acetyl transferase CBP modulate target gene expression.
  • beta-catenin is absent, certain TCFs repress transcription by interacting with the co-repressors groucho and CtBP.
  • beta-catenin also binds to cadherin cell adhesion molecules and provides a link to the actin cytoskeleton.
  • Data for the Wnt pathway have been obtained from a variety of systems and organisms.
  • the following example illustrate the use of DPI-6, DPI- 186 or DPI- 192 of the invention for screening or diagnosis of a neuropsychiatric or neurological diseases, determining the prognosis of a subject having a neuropsychiatric or neurological disease, or monitoring the effectiveness of a neuropsychiatric or neurological disease therapy.
  • modulators e.g., agonist or antagonists
  • a colipase fold in the carboxy-terminal domain of Dkks may enable Dkks proteins to interact with lipids and subsequently Wnt proteins, in order to regulate Wnt function (Aravind L, Koonin EV, A colipase fold in the carboxy-terminal domain of the Wnt antagonists ⁇ the Dickkopfs, Curr Biol 1998 Jul 2 8:14 R477-8), since Wnt proteins are known to be tightly associated with the cell surface (Smolich BD, McMahon JA, McMahon AP, Papkoff J, Wnt family proteins are secreted and associated with the cell surface, Mol Biol Cell 1993 Dec 4: 12 1267-75).
  • DPI-6, DPI-186 and DPI-192 of Dickkopf with molecular weights and pi values of 62182 Da and pi of 4.29, 53154 Da and pi of 4.29, 63376 Da and pi of 4.31 respectively have been shown herein to be significantly differentially expressed in the cerebrospinal fluid (CSF) of subjects having Depression as compared with the CSF of subjects free from a neuropsychiatric or neurological disease.
  • CSF cerebrospinal fluid
  • quantitative detection of DPI-6, DPI-186 or DPI-192 in CSF can be used to diagnose neuropsychiatric or neurological diseases, determine the progression of a neuropsychiatric or neurological disease or monitor the effectiveness of a therapy for a neuropsychiatric or neurological disease.
  • compounds that modulate i.e., upregulate or downregulate) the expression, activity or both the expression and activity of DPI-6 are administered to a subject in need of freatment or for prophylaxis of a neuropsychiatric or neurological disease.
  • Antibodies that modulate the expression, activity or both the expression and activity of DPI-6, DPI-186 or DPI-192 are suitable for this purpose.
  • nucleic acids coding for all or a portion of DPI-6, DPI-186 and DPI-192, or nucleic acids complementary to all or a portion of DPI-6, DPI-186 or DPI-192 may be administered.
  • DPI-6, DPI-186 or DPI-192, or fragments of the DPI-6, DPI-186 or DPI-192 polypeptides may also be administered.
  • the invention also provides screening assays to identify additional compounds that modulate the expression of DPI-6, DPI-186 or DPI-192, or activity of DPI-6, DPI-186 or DPI-192.
  • Compounds that modulate the expression of DPI-6, DPI-186 and DPI-192 in vitro can be identified by comparing the expression of DPI-6, DPI- 186 or DPI-192 in cells treated with a test compound to the expression of DPI-6, DPI- 186 or DPI-192 in cells treated with a control compound (e.g., saline).
  • a control compound e.g., saline
  • Methods for detecting expression of DPI-6, DPI-186 or DPI-192 include measuring the level of DPI-6, DPI-186 or DPI-192 RNA (e.g., by northern blot analysis or RT-PCR) and measuring DPI-6, DPI-186 or DPI-192 protein (e.g., by immunoassay or western blot analysis).
  • Compounds that modulate the activity of DPI-6, DPI- 186 or DPI- 192 can be identified by comparing the ability of a test compound to agonize or antagonize a function of DPI-6, DPI-186 or DPI-192 , such as its ability to block Frizzled activation, activity or the binding of Wnts to the Frizzled receptor, activation of Disheveled, GSK-3 phosphorylation, changes in expression of Wnt regulated genes, to the ability of a confrol compound (e.g., saline) to inhibit the same function of DPI-6, DPI-186 or DPI-192 .
  • a confrol compound e.g., saline
  • Frizzled can be determined by, for example, contacting DPI-6, DPI-186 or DPI-192 with cells known to express the Wnt and or the Frizzled receptor and assaying the extent of binding between DPI-6, DPI-186 or DPI-192 and Wnt of the cell surface receptor, or by contacting DPI-6, DPI-186 or DPI-192 with its receptor in a cell-free assay, i.e., an assay where the DPI-6, DPI-186 or DPI-192 and Wnt and or the
  • Frizzled receptor are isolated, and, preferably, recombinantly produced, and assaying the extent of binding between DPI-6, DPI-186 or DPI-192 and Wnt and or the Frizzled receptor. Through the use of such assays, candidate compounds may be tested for their ability to agonize or antagonize the binding of DPI-6, DPI-186 or DPI- 192 to its Wnt and or the Frizzled receptor.

Abstract

La présente invention concerne des procédés et des compositions permettant le criblage, le diagnostic et le pronostic de BAD, le contrôle de l'efficacité de traitements de BAD et la mise au point de médicaments. La présente invention concerne également des caractéristiques associées aux BAD (DFs) qui peuvent être détectées par une électrophorèse bidimensionnelle de liquide céphalo-rachidien, de sérum ou de plasma. La présente invention concerne également des isoformes de protéines associées aux BAD (DPIs) qui peuvent être détectées dans du liquide céphalo-rachidien, du sérum ou du plasma, des préparations comprenant des DPIs isolées, des anticorps immunospécifiques aux DPIs, ainsi que des trousses comprenant ces dernières.
EP01905998A 2000-02-24 2001-02-23 Diagnostic de maladies affectives bipolaires (bad) et de depressions unipolaires Withdrawn EP1259819A2 (fr)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GB0004412 2000-02-24
GB0004412A GB0004412D0 (en) 2000-02-24 2000-02-24 Proteins and their use for diagnosis and treatment of bipolar affective disorder and unipolar depression
GB0030050 2000-12-08
GB0030050A GB0030050D0 (en) 2000-12-08 2000-12-08 Proteins, genes and their use for diagnosis and treatment of bipolar affective disorder (BAD) and unipolar depression
US25483000P 2000-12-12 2000-12-12
US254830P 2000-12-12
PCT/GB2001/000791 WO2001063294A2 (fr) 2000-02-24 2001-02-23 Proteines, genes et leur utilisation dans le diagnostic et le traitement de maladies affectives bipolaires (bad) et de depressions unipolaires

Publications (1)

Publication Number Publication Date
EP1259819A2 true EP1259819A2 (fr) 2002-11-27

Family

ID=27255558

Family Applications (2)

Application Number Title Priority Date Filing Date
EP01905998A Withdrawn EP1259819A2 (fr) 2000-02-24 2001-02-23 Diagnostic de maladies affectives bipolaires (bad) et de depressions unipolaires
EP01905994A Withdrawn EP1259539A1 (fr) 2000-02-24 2001-02-23 Diagnostic et traitement de la maladie affective bipolaire

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP01905994A Withdrawn EP1259539A1 (fr) 2000-02-24 2001-02-23 Diagnostic et traitement de la maladie affective bipolaire

Country Status (3)

Country Link
EP (2) EP1259819A2 (fr)
AU (2) AU2001233957A1 (fr)
WO (2) WO2001063294A2 (fr)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7314762B2 (en) 2001-11-21 2008-01-01 Nanogen, Inc. Apolipoprotein biopolymer markers indicative of insulin resistance
US7052849B2 (en) * 2001-11-23 2006-05-30 Syn X Pharma, Inc. Protein biopolymer markers predictive of insulin resistance
US20030113808A1 (en) * 2001-12-13 2003-06-19 George Jackowski Apolipoprotein biopolymer markers predictive of alzheimers disease
DE10225144A1 (de) * 2002-05-29 2003-12-18 Immatics Biotechnologies Gmbh An MHC-Moleküle bindende Tumor-assoziierte Peptide
EP1394549A1 (fr) 2002-08-23 2004-03-03 Bayer HealthCare AG Biomarqueurs pour le diagnostic de la maladie d'Alzheimer
CA2745569C (fr) * 2002-08-23 2015-07-14 Bayer Schering Pharma Aktiengesellschaft Biomarqueurs permettant le diagnostic de la maladie d'alzheimer
CA2522985A1 (fr) * 2003-04-17 2004-10-28 Affectis Pharmaceuticals Ag Moyens et methodes pour le diagnostic et le traitement de troubles affectifs
KR101219519B1 (ko) * 2011-05-06 2013-01-09 한국기초과학지원연구원 렉틴을 이용한 암 진단 방법
RU2488118C1 (ru) * 2011-11-21 2013-07-20 Учреждение Российской Академии Медицинских Наук Научно-Исследовательский Институт Психического Здоровья Сибирского Отделения Рамн Способ прогнозирования формирования органического расстройства личности
CN110361242B (zh) * 2019-08-14 2022-01-18 武汉赛维尔生物科技有限公司 一种用于眼球组织的固定液以及眼球组织制片的预处理方法

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990014363A1 (fr) * 1989-05-19 1990-11-29 Amgen Inc. Inhibiteur de metalloproteinase
EP0910641A1 (fr) * 1997-02-13 1999-04-28 Smithkline Beecham Plc Variantes d'epissage de molecule d'adherence cellulaire neuronale
US5866412A (en) * 1997-03-27 1999-02-02 Millennium Pharmaceuticals, Inc. Chromosome 18 marker
WO1999004825A1 (fr) * 1997-07-22 1999-02-04 Millennium Pharmaceuticals, Inc. Procedes et compositions permettant le diagnostic et le traitement de troubles neuropsychiatriques
WO1999021886A1 (fr) * 1997-10-24 1999-05-06 Aventis Pharma S.A. PEPTIDES CAPABLES D'INHIBER L'INTERACTION ENTRE LES PRESENILINES ET LE PEPTIDE β-AMYLOIDE OU SON PRECURSEUR
WO1999035290A1 (fr) * 1998-01-08 1999-07-15 Millennium Pharmaceuticals, Inc. Procedes et compositions permettant de diagnostiquer et de traiter des troubles neuropsychiatriques
WO1999055380A1 (fr) * 1998-04-27 1999-11-04 Pacific Northwest Cancer Foundation GENE Nr-CAM, ACIDES NUCLEIQUES ET PRODUITS D'ACIDE NUCLEIQUE UTILISES DANS UNE THERAPIE OU UN DIAGNOSTIC DE TUMEURS
CA2332109A1 (fr) * 1998-05-12 1999-11-18 Human Genome Sciences, Inc. 97 proteines humaines secretees
NZ527292A (en) * 1999-03-23 2005-03-24 Genentech Inc Secreted and transmembrane polypeptides PRO1484 and nucleic acids encoding the same, with homology to an adipocyte complement-related protein

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0163294A3 *

Also Published As

Publication number Publication date
WO2001063294A3 (fr) 2002-04-11
WO2001062787A1 (fr) 2001-08-30
AU2001233957A1 (en) 2001-09-03
AU2001233955A1 (en) 2001-09-03
WO2001063294A2 (fr) 2001-08-30
EP1259539A1 (fr) 2002-11-27

Similar Documents

Publication Publication Date Title
US20070015217A1 (en) Nucleic acid molecules, polypeptides and uses therefor, including diagnosis and treatment of Alzheimer's disease
US20040110938A1 (en) Proteins, genes and their use for diagnosis and treatment of schizophrenia
US20040053309A1 (en) Proteins, genes and their use for diagnosis and treatment of kidney response
EP1354199A2 (fr) Diagnostic et traitement de la sclerose en plaques
EP1999150A1 (fr) Nouvelles proteines isoformes et leur utilisation
EP1408333A2 (fr) Diagnostic et traitement de la maladie d'Alzheimer
US20030064419A1 (en) Proteins, genes and their use for diagnosis and treatment of breast cancer
US20030064411A1 (en) Nucleic acid molecules, polypeptides and uses therefor, including diagnosis and treatment of Alzheimer's disease
US20030152935A1 (en) Proteins, genes and their use for diagnosis and treatment of breast cancer
WO2002046767A2 (fr) Molecules d'acide nucleique, polypeptides et utilisations associees, parmi lesquelles diagnostic et traitement de la maladie d'alzheimer
EP1259819A2 (fr) Diagnostic de maladies affectives bipolaires (bad) et de depressions unipolaires
US20020142303A1 (en) Proteins, genes and their use for diagnosis and treatment of Schizophrenia
US20030032200A1 (en) Proteins, genes and their use for diagnosis and treatment of bipolar affective disorder (BAD) and unipolar depression
EP1259818A2 (fr) Diagnostic et traitement de la schizophrenie
WO2001069261A2 (fr) Proteines, genes et leur utilisation dans le diagnostic et le traitement de la demence vasculaire
US20040203022A1 (en) Proteins and genes for diagnosis and treatment of ErbB2-related cancer
US20050163789A9 (en) Nucleic acid molecules, polypeptides and uses therefor, including diagnosis and treatment of Alzheimer's disease
US20040043425A1 (en) Proteins, genes and their use for diagnosis and treatment of chronic asthma
EP1368647A2 (fr) Proteines, genes et leur utilisation pour le diagnostic et le traitement de la reponse vasculaire
US20030092197A1 (en) Proteins, genes and their use for diagnosis and treatment of cardiac response
US20030077603A1 (en) Methods and compositions for diagnosis of hepatoma
EP1356283A2 (fr) Proteines, genes et leur utilisation pour le diagnostic et le traitement de la reponse cardiaque

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20020920

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

17Q First examination report despatched

Effective date: 20021213

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20040319