WO2001063293A2 - Proteines et genes et leur utilisation dans le diagnostic et le traitement de la schizophrenie - Google Patents

Proteines et genes et leur utilisation dans le diagnostic et le traitement de la schizophrenie Download PDF

Info

Publication number
WO2001063293A2
WO2001063293A2 PCT/GB2001/000783 GB0100783W WO0163293A2 WO 2001063293 A2 WO2001063293 A2 WO 2001063293A2 GB 0100783 W GB0100783 W GB 0100783W WO 0163293 A2 WO0163293 A2 WO 0163293A2
Authority
WO
WIPO (PCT)
Prior art keywords
spi
schizophrenia
related polypeptide
cells
antibody
Prior art date
Application number
PCT/GB2001/000783
Other languages
English (en)
Other versions
WO2001063293A3 (fr
WO2001063293A8 (fr
Inventor
Herath Mudiyanselage Athula Chandrasiri Herath
Rajesh Bhikhu Parekh
Christian Rohlff
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 GB0004415A external-priority patent/GB0004415D0/en
Application filed by Oxford Glycosciences (Uk) Ltd. filed Critical Oxford Glycosciences (Uk) Ltd.
Priority to EP01905991A priority Critical patent/EP1259818A2/fr
Priority to AU2001233954A priority patent/AU2001233954A1/en
Publication of WO2001063293A2 publication Critical patent/WO2001063293A2/fr
Publication of WO2001063293A8 publication Critical patent/WO2001063293A8/fr
Publication of WO2001063293A3 publication Critical patent/WO2001063293A3/fr

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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • G01N2800/302Schizophrenia

Definitions

  • PROTEINS GENES AND THEIR USE FOR DIAGNOSIS AND TREATMENT OF SCHIZOPHRENIA
  • the present invention relates to the identification of proteins and protein isoforms that are associated with Schizophrenia 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.
  • neuropsychiatric disorders In the majority of psychiatric disorders, little is known about a link between changes at a cellular and/or molecular level and nervous system structure and function.
  • the paucity of detectable neuralgic defects distinguishes neuropsychiatric disorders such as Schizophrenia from neurological disorders, where manifestations of anatomical and biochemical changes have been identified in many cases. Consequently the identification and characterization of cellular and/or molecular causative defects and neuropathologies are necessary for improved treatment of neuropsychiatric disorders.
  • Schizophrenia is characterized by episodes of positive symptoms such as delusions, hallucinations, paranoia and psychosis and/or negative symptoms such as flattened affect, impaired attention, social withdrawal, and cognitive impairments (Ban et al, Psychiatr. Dev. (1984) 2:179-199). It afflicts 1.1% of the U.S. population and imposes a disproportionately large economic burden due to long-term expenditures for hospitalisation, treatment and rehabilitation, and lost productivity. Cost-of-illness studies have estimated that in 1990 the total economic burden of Schizophrenia in the US was $32.5 billion. (Rice J Clin Psychiatry (1999) 60 Suppl 1 4-6).
  • DAPs disease associated proteins
  • Schizophrenia Currently diagnosis of Schizophrenia remains clinically based on the presence of certain types of hallucinations, delusions and thought disorders (Andreasen Lancet (1995) 346:477-481). It is made on the basis of a careful clinical interview and mental status examination according to international established manuals, in particular the DSM-IV or ICD 10.
  • the core clinical symptoms comprise formal thought disorders, delusions, hallucinations (also summarized as positive symptoms), and negative symptoms such as lack of drive and affect flattening.
  • Neuroimaging techniques such as magnetic resonance imaging or positron emission tomography show subtle changes of the frontal and temporal lobes and the basal ganglia (Buchsbaum, Schiz. Bull. (1990) 16:379-389) in the majority of patients.
  • Neuroleptic agents are essential for the treatment of Schizophrenia. While typical neuroleptics effect primarily the dopaminergic system, newer atypical neuroleptics also afflict serotonergic synapses. In general, the latter have greater effects on negative symptoms and cause less extrapyramidal side effects than typical neuroleptic compounds. It is generally accepted that early and continuous neuroleptic treatment may improve the outcome of the disorder. Nevertheless, regardless of the particular drug used, neuroleptic treatment is still considered to be solely symptomatic and does not inhibit the causes of the disorder.
  • Schizophrenia has no objective biochemical markers useful for diagnosis and prognosis in living patients.
  • Many CNS pathologies involve increased neuronal loss and such neuronal loss or impaired synaptogenesis may result in disease associated alterations of neuronal and CSF proteins.
  • Synaptic pathologies have been implicated in Schizophrenia (Heinonen et al, Neuroscience (1995) 64:375-384; Benes Schiz. Bull. (1993) 19:537-549.).
  • the present invention provides methods and compositions for clinical screening, diagnosis, prognosis, therapy and prophylaxis of Schizophrenia, for monitoring the effectiveness of Schizophrenia 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 Schizophrenia.
  • a first aspect of the invention provides methods for diagnosis of Schizophrenia that comprise analyzing a sample of cerebrospinal fluid (CSF) by two-dimensional electrophoresis to detect the presence or level of at least one Schizophrenia- Associated Feature (SF), e.g., one or more of the SFs disclosed herein or any combination thereof.
  • CSF cerebrospinal fluid
  • SF Schizophrenia- Associated Feature
  • a second aspect of the invention provides methods for diagnosis of Schizophrenia that comprise detecting in a sample of CSF the presence or level of at least one Schizophrenia- Associated Protein Isoform (SPI), e.g., one or more of the SPIs disclosed herein or any combination thereof.
  • SPI Schizophrenia- Associated Protein Isoform
  • a third aspect of the invention provides antibodies, e.g. monoclonal and polyclonal antibodies capable of immunospecific binding to an SPI, e.g., an SPI disclosed herein.
  • a fourth aspect of the invention provides a preparation comprising an isolated SPI, i.e., an SPI free from proteins or protein isoforms having a significantly different isoelectric point or a significantly different apparent molecular weight from the SPI.
  • a fifth aspect of the invention provides methods of treating Schizophrenia, 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 an SPI in subjects having Schizophrenia, in order to prevent or delay the onset or development of Schizophrenia, to prevent or delay the progression of Schizophrenia, or to ameliorate the symptoms of Schizophrenia.
  • 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 an SPI, an SPI analog, or an SPI-related polypeptide.
  • SPI analog refers to a polypeptide that possesses a similar or identical function as an SPI but need not necessarily comprise an amino acid sequence that is similar or identical to the amino acid sequence of the SPI, or possess a structure that is similar or identical to that of the SPI.
  • an amino acid sequence of a polypeptide is "similar" to that of an SPI if it satisfies at least one 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 least 85%, at least 90%, at least 95% or at least 99%) identical to the amino acid sequence of the SPI; (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 a'mino 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 50 amino acid residues, at least 60 amino residues, at least 70 amino acid residue
  • a polypeptide with "similar structure" to that of an SPI refers to a polypeptide that has a similar secondary, tertiary or quarternary structure as that of 'the SPI.
  • 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.
  • SPI fusion protein refers to a polypeptide that comprises (i) an amino acid sequence of an SPI, an SPI fragment, an SPI-related polypeptide or a fragment of an SPI-related polypeptide and (ii) an amino acid sequence of a heterologous polypeptide (i. e. , a non-SPI, non-SPI fragment or non-SPI- related polypeptide).
  • SPI homolog refers to a polypeptide that comprises an amino acid sequence similar to that of an SPI but does not necessarily possess a similar or identical function as the SPI.
  • SPI ortholog refers to a non-human polypeptide that (i) comprises an amino acid sequence similar to that of an SPI and (ii) possesses a similar or identical function to that of the SPI.
  • SPI-related polypeptide refers to an SPI homolog, an SPI analog, an isoform of SPI, an SPI 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 50 amino 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 least 175 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 an SPI may or may not possess a functional activity of the second polypeptide.
  • fold change includes “fold increase” and “fold decrease” and refers to the relative increase or decrease in abundance of an SF or the relative increase or decrease in expression or activity of a polypeptide (e.g. an SPI) in a first sample or sample set compared to a second sample (or sample set).
  • a polypeptide e.g. an SPI
  • An SF or polypeptide fold change may be measured by any technique known to those of skill in the art, however 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.
  • isoforms can differ in their amino- acid composition (e.g. as a result of alternative splicing or limited proteolysis) and in addition, or in the alternative, may arise from differential post-translational modification (e.g., glycosylation, acylation, phosphorylation).
  • isoform also refers to a protein that exists in only a single form, i.e., it is not expressed as several variants.
  • modulate when used herein in reference to expression or activity of an SPI or an SPI-related polypeptide refers to the upregulation or downregulation of the expression or activity of the SPI or an SPI-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 Karlin and Altschul Proc. Natl. Acad. Sci. USA (1990) 87:2264-2268, modified as in Karlin and Altschul (1993) Proc. Nail. Acad. Sci. USA 90:5873-5877.
  • the NBLAST and XBLAST programs of Altschul et al, J. Mol. Biol. (1990) 215:403-410 have incorporated such an algorithm.
  • Gapped BLAST can be utilized as described in Altschul et al, Nucleic Acids Res. (1997) 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 human CSF, which has been annotated to identify twelve landmark features, designated CSF1 to CSF 12.
  • Figure 2 comprises amino acid sequences of SPI-206 (Figure 2A) and the nucleic acid sequence encoding the amino acid sequence of Figure 2A ( Figure 2B). Peptides of SPI-206 identified by mass spectrometry are underlined in the sequence of Figure 2A, and the amino acid sequences determined by mass spectrometry are highlighted.
  • FIG. 3 shows tissue distribution of SPI-206 mRNA.
  • Levels of mRNA in samples of normal tissue were quantified by real time RT-PCR.
  • the mRNA levels are expressed as the number of copies per nanogram cDNA. Note the 25 times difference in scale between the graph containing brain-related samples, and the graph containing body samples.
  • Figure 4 comprises amino acid sequences of SPI-238 and SPI-240 ( Figure 2A) and the nucleic acid sequence encoding the amino acid sequence of Figure 2A ( Figure 2B).
  • FIG. 5 shows tissue distribution of SPI-238 and SPI-240 mRNA. Levels of mRNA in samples of normal tissue were quantified by real time RT-PCR. The mRNA levels are expressed as the number of copies per nanogram cDNA. Note the 25 times difference in scale between the graph containing brain-related samples, and the graph containing body samples.
  • the invention described in detail below provides methods and compositions for clinical screening, diagnosis and prognosis of Schizophrenia in a mammalian subject for identifying patients most likely to respond to a particular therapeutic treatment, for monitoring the results of Schizophrenia therapy, for drug screening and drug development.
  • the invention also encompasses the administration of therapeutic compositions to a mammalian subject to treat or prevent Schizophrenia.
  • 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.
  • a body fluid e.g. blood, serum, plasma, saliva or urine
  • a tissue sample from a subject at risk of having or developing Schizophrenia e.g. a biopsy such as a brain biopsy
  • 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 Schizophrenia-Associated Features (SFs) for screening, prevention or diagnosis of Schizophrenia, to determine the prognosis of a subject having Schizophrenia, to monitor progression of Schizophrenia, to monitor the effectiveness of Schizophrenia therapy, for identifying patients most likely to respond to a particular therapeutic treatment, or for drug development.
  • SFs Schizophrenia-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 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. In the Apollo 2 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 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.
  • the term “feature” refers to a spot detected in a 2D gel
  • the term “Schizophrenia-Associated Feature” (SF) refers to a feature that is differentially present in a sample (e.g. a sample of CSF) from a subject having Schizophrenia compared with a sample (e.g. a sample of CSF) from a subject free from Schizophrenia.
  • a feature (or a protein isoform of SPI, 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 SPI (e.g., 2D electrophoresis or an immunoassay) gives a different signal when applied to the first and second samples.
  • a feature, isoform or SPI is "increased" in the first sample with respect to the second if the method of detection indicates that the feature, isoform or SPI is more abundant in the first sample than in the second sample, or if the feature, isoform or SPI is detectable in the first sample and undetectable in the second sample.
  • a feature, isoform or SPI is "decreased" in the first sample with respect to the second if the method of detection indicates that the feature, isoform or SPI is less abundant in the first sample than in the second sample or if the feature, isoform or SPI 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 (ERF) 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.
  • 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 (ERF) 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 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 SFs disclosed herein have been identified by comparing CSF samples from subjects having Schizophrenia against CSF samples from subjects free from Schizophrenia.
  • Subjects free from Schizophrenia include subjects with no known disease or condition (normal subjects) and subjects with diseases (including neurological and neurodegenerative diseases) other than Schizophrenia.
  • the first group consists of SFs that are decreased in the CSF of subjects having Schizophrenia as compared with the CSF of subjects free from Schizophrenia. These SFs can be described by apparent molecular weight (MW) and isoelectric point (pi) as provided in Table I.
  • MW apparent molecular weight
  • pi isoelectric point
  • the second group consists of SFs that are increased in the CSF of subjects having Schizophrenia as compared with the CSF of subjects free from Schizophrenia.
  • These SFs can be described by MW and pi as follows:
  • the signal obtained upon analyzing CSF from subjects having Schizophrenia relative to the signal obtained upon analyzing CSF from subjects free from Schizophrenia 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 SF in subjects free from Schizophrenia according to the analytical protocol and detection technique in use, as is conventional in the diagnostic art.
  • at least one control positive CSF sample from a subject known to have Schizophrenia or at least one control negative CSF sample from a subject known to be free from Schizophrenia (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 SF 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 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 an SF or SPI is typically less than 3% and variation in the measured mean MW of an SF or SPI is typically less than 5%.
  • calibration experiments should be performed to compare the MW and pi for each SF or protein isoform as detected (a) by the Reference Protocol and (b) by the deviant protocol.
  • SFs can be used for detection, prognosis, diagnosis, or monitoring of Schizophrenia, or for identifying patients most likely to respond to a specific therapeutic treatment, or for drug development.
  • CSF from a subject e.g., a subject suspected of having Schizophrenia
  • 2D electrophoresis for quantitative detection of one or more of the following SFs: SF-14, SF-16, SF-17, SF-18, SF-19, SF-20, SF-21, SF-22, SF-23, SF-24, SF-25, SF-26, SF-27, SF-28, SF-29, SF-30, SF-31, SF-32, SF- 33, SF-34, SF-35, SF-36, SF-37, SF-38, SF-39, SF-40, SF-41, SF-42, SF-43, SF-44, SF-45,
  • a decreased abundance of said one or more SFs in the CSF from the subject relative to CSF from a subject or subjects free from Schizophrenia (e.
  • CSF from a subject is analyzed by 2D electrophoresis for quantitative detection of one or more of the following SFs: SF-80, SF-81, SF-82, SF-83, SF-84, SF-85, SF-86, SF-87, SF-88, SF-89, SF-90, SF-91, SF-92, SF-93, SF-
  • SF-106 SF-107, SF-108, SF-109, SF-110, SF-111, SF-112, SF-113, SF-114, SF-115, SF-116,
  • An increased abundance of said one or more SFs in the CSF from the subject relative to CSF from a subject or subjects free from Schizophrenia indicates the presence of Schizophrenia.
  • CSF from a subject is analyzed by 2D electrophoresis for quantitative detection of (a) one or more SFs or any combination of them, whose decreased abundance indicates the presence of Schizophrenia, i.e., SF-14, SF-16, SF-17, SF-18, SF-19, SF-20, SF-21, SF-22, SF-23, SF-24, SF-25, SF-26, SF-27, SF-28, SF-29, SF-30, SF-31, SF- 32, SF-33, SF-34, SF-35, SF-36, SF-37, SF-38, SF-39, SF-40, SF-41, SF-42, SF-43, SF-44, SF-45, SF-46, SF-47, SF-48, SF-49, SF-51, SF-52, SF-53, SF-55, SF-56, SF-57,
  • SF-220 SF-221, SF-222, SF-226, SF-227, SF-228, SF-229, SF-230, SF-231, SF-232, SF-233,
  • CSF from a subject is analyzed by 2D electrophoresis for quantitative detection of one or more of the following SFs: SF-14, SF-16, SF-17, SF-18, SF-19, SF-20, SF-21, SF-22, SF-23, SF-24, SF-25, SF-26, SF-27, SF-28, SF- ' 29, SF-30, SF-31, SF-32, SF-33, SF-34, SF-35, SF-36, SF-37, SF-38, SF-39, SF-40, SF-41, SF-42, SF-43, SF-44, SF-45, SF-46, SF-47, SF-48, SF-49, SF-51, SF-52, SF-53, SF-55, SF- 56, SF-57, SF-58, SF-80, SF-81, SF-82, SF
  • a decrease in one or more SF/ERF ratios in a test sample relative to the SF/ERF ratios in a control sample or a reference range indicates the presence of Schizophrenia; SF-14, SF-16, SF-17, SF-18, SF-19, SF-20, SF-21, SF-22,.
  • an increase in one or more SF/ERF ratios in a test sample relative to the SF/ERF ratios in a control sample or a reference range indicates the presence of Schizophrenia; SF-80, SF-81, SF-82, SF-83, SF-84, SF-85, SF-86, SF-87, SF-88, SF-89, SF-90, SF-91, SF-92, SF- 93, SF-94, SF-95, SF-96, SF-97, SF-98, SF-99, SF-100, SF-101, SF-102, SF-103, SF-104, SF-105, SF-106, SF-107, SF-108, SF-109, SF-110, SF-111, SF-112, SF-113, SF-114, SF-115, SF-116, SF-117, SF-118, SF-119, SF
  • CSF from a subject is analyzed by 2D electrophoresis for quantitative detection of (a) one or more SFs, or any combination of them, whose decreased SF/ERF ratio(s) in a test sample relative to the SF/ERF ratio(s) in a control sample indicates the presence of Schizophrenia, i. e.
  • CSF from a subject is analyzed for quantitative detection of a plurality of SFs.
  • CSF from a subject is analyzed for quantitative detection of one or more Schizophrenia-Associated Protein Isoforms (SPIs) for screening or diagnosis of Schizophrenia, to determine the prognosis of a subject having Schizophrenia, to monitor the effectiveness of Schizophrenia therapy, for identifying patients most likely to respond to a particular therapeutic treatment or for drug development.
  • SPIs Schizophrenia-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.
  • the term "Schizophrenia- Associated Protein Isoform” refers to a protein isoform that is differentially present in CSF from a subject having Schizophrenia compared with CSF from a subject free from Schizophrenia.
  • the term “isoform” also refers to a protein that exists in only a single form, i.e., it is not expressed as several variants.
  • SPIs Two groups of SPIs have been identified by amino acid sequencing of SFs. SPIs 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 SPIs was performed primarily using the SEQUEST search program (Eng et al, J. Am. Soc.
  • the first group consists of SPIs that are decreased in the CSF of subjects having Schizophrenia as compared with the CSF of subjects free from Schizophrenia, where the differential presence is significant.
  • the amino acid sequences of tryptic digest peptides of these SPIs identified by tandem mass spectrometry and database searching as described in the Examples, infra are listed in Table IV in addition to the pis and MWs of these SPIs.
  • SPI- 238 and SPI- 140 the " partial sequence information for these SPIs derived from tandem mass spectrometry was not found to be described in any known public database. These SPIs are listed as 'NOVEL' in Table IV, and further described below.
  • the second group comprises SPIs that are increased in the CSF of subjects having Schizophrenia as compared with the CSF of subjects free from Schizophrenia, where .the differential presence is significant.
  • the amino acid sequences of tryptic digest peptides of these SPIs identified by tandem mass spectrometry and database searching are listed in Table V in addition to the pis and MWs of these SPIs.
  • SPI- 206 the partial sequence information derived from tandem mass spectrometry was not found to be described in any known public database. This SPI is listed as 'NOVEL' in Table V, and further described below.
  • the SPI is a protein comprising a peptide sequence described for that SPI (preferably comprising a plurality of, more preferably all of, ' the peptide sequences described for that SPI) and has a pi of about the value stated for that SPI (preferably within 10%, more preferably within 5% still more preferably within 1% of the stated value) and has a MW of about the value stated for that SPI (preferably within 10%, more preferably within 5%, still more preferably within 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
  • NCBI National Center for Biotechnology Information
  • NCBI National Center for Biotechnology Information
  • BLAST is maintained at the National Center for Biotechnology Information (NCBI) (http://www.ncbi.nlm.nih.gov) and is based on a statistical theory developed by Samuel Karlin and Steven Altschul (Proc. Natl Acad. Sci. USA (1990) 87:2284-2268), later modified as in Karlin and Altschul (Proc.
  • BLASTP can be used to search a protein sequence against a protein database.
  • TBLASTN can be used to search a Protein Sequence against a Nucleotide Database, by translating each database Nucleotide sequence in all 6 reading frames.
  • FASTA as described in Pearson and Lipman (1988) Proc. Natl. Acad. Sci. 85:2444-8. See also Pearson Methods Enzymol. (1990) 183:63-98 and Pearson Genomics (1991) l l(3):635-50. Examples of available protein sequence databases include:
  • 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).
  • PIR Protein Information Resource
  • MIPS Munich Information Center for Protein Sequences
  • JIPID Japanese International Protein Sequence Database
  • the Protein Identification Resource (PIR) is a division of the National Biomedical Research Foundation (NBRF) which is affiliated with Georgetown University Medical
  • 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 SPIs: SPI-6, SPI-7, SPI-8, SPI-9, SPI-10, SPI-11, SPI-13, SPI-15 SPI-16, SPI-17, SPI-18, SPI-19, SPI-20, SPI-21, SPI-23, SPI-24, SPI-25, SPI-26, SPI-28 SPI-29, SPI-30, SPI-32, SPI-33, SPI-34, SPI-35, SPI-36, SPI-38, SPI-39, SPI-41, SPI-42 SPI-43, SPI-44, SPI-231, SPI-232, SPI-233, SPI-234, SPI-235, SPI-236, SPI-237, SPI-238 SPI-239, SPI-240, SPI-241, SPI-312, SPI-352, SPI-353, SPI-354, SPI-355, SPI-357, SPI-401 SPI-6,
  • CSF from a subject is analyzed for quantitative detection of one or more of the following SPIs: SPI-54, SPI-56, SPI-57, SPI-58, SPI-59, SPI-60, SPI-62, SPI-63, SPI-65, SPI-67, SPI-69, SPI-73, SPI-74, SPI-75, SPI-76, SPI-77, SPI-78, SPI-80,, SPI-81, SPI-82, SPI-83, SPI-85, SPI-87, SPI-88, SPI-91, SPI-92, SPI-93, SPI-95, SPI-96, SPI-97, SPI-99, SPI-100, SPI-101, SPI-105, SPI-107, SPI-113, SPI- 114, SPI-115, SPI-118, SPI-122, SPI-123, SPI-124, SPI-127, SPI-129, SPI-130
  • CSF from a subject is analyzed for quantitative detection of (a) one or more SPIs, or any combination of them, whose decreased abundance indicates the presence of Schizophrenia, i.e., SPI-6, SPI-7, SPI-8, SPI-9, SPI-10, SPI-11, SPI-13, SPI-15, SPI-16, SPI-17, SPI-18, SPI-19, SPI-20, SPI-21, SPI-23, SPI-24, SPI-25, SPI-26, SPI-28, SPI-29, SPI-30, SPI-32, SPI-33, SPI-34, SPI-35, SPI-36, SPI-38, SPI-39, SPI-41, SPI-42, SPI-43, SPI-44, SPI-231, SPI-232, SPI-233, SPI-234, SPI-235, SPI-236, SPI-237, SPI-238, SPI-239, SPI-240, SPI-241, SPI-3
  • CSF from a subject is analyzed for quantitative detection of one or more SPIs and one or more previously known biomarkers of Schizophrenia (e.g., candidate markers such as hypersensitive platelet glutamate receptors (Berk et al, Int Clin Psychopharmacol (1999) 14:199-122)).
  • candidate markers such as hypersensitive platelet glutamate receptors (Berk et al, Int Clin Psychopharmacol (1999) 14:199-122)
  • the abundance of each SPI and known biomarker relative to a control or reference range indicates whether a subject has Schizophrenia.
  • 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 SPIs described herein include previously unknown proteins, as well as isoforms of known proteins where the isoforms were not previously known to be associated with Schizophrenia.
  • the present invention additionally provides: (a) a preparation comprising the isolated SPI; (b) a preparation comprising one or more fragments of the SPI; and (c) antibodies that bind to said SPI, to said fragments, or both to said SPI and to said fragments.
  • an SPI 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 SPI, as determined by 2D electrophoresis.
  • a "significantly different" pi or MW is one that permits the contaminating protein to be resolved from the SPI 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 an SPI, 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 SPI.
  • the SPIs 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 SPIs are separated on a 2-D gel by virtue of their MWs and pis and visualized by staining the gel.
  • the SPIs 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.
  • SPIs can be detected in an immunoassay.
  • an immunoassay is performed by contacting a sample from a subject to be tested with an anti- SPI antibody under conditions such that immunospecific binding can occur if the SPI is present, and detecting or measuring the amount of any immunospecific binding by the antibody.
  • Anti-SPI 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 SPI is itself a family member.
  • the anti-SPI antibody preferentially binds to the SPI rather than to other isoforms of the same protein.
  • the anti-SPI antibody binds to the SPI 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.
  • SPIs 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-SPI antibodies as described herein, e.g., the antibodies identified in Table VII, or others raised against the SPIs of interest.
  • the immunoblots can be used to identify those anti-SPI antibodies displaying the selectivity required to immuno-specifically differentiate an SPI from other isoforms encoded by the same gene.
  • binding of antibody in tissue sections can be used to detect aberrant SPI localization or an aberrant level of one or more SPIs.
  • antibody to an SPI can be used to assay a tissue sample (e.g., a brain biopsy) from a subject for the level of the SPI where an aberrant level of SPI is indicative of Schizophrenia.
  • a 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 Schizophrenia 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 Schizophrenia.
  • 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 protem 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 immunoa
  • an SPI 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-SPI antibody
  • the capture reagent can optionally be immobilized on a solid phase.
  • a directly or indirectly labelled detection reagent is used to detect the captured SPI.
  • the detection reagent is a lectin.
  • any lectin can be used for this purpose that preferentially binds to the SPI rather than to other isoforms that have the same core protein as the SPI or to other proteins that share the antigenic determinant recognized by the antibody.
  • the chosen lectin binds to the SPI 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 SPI or to said other proteins that share the antigenic determinant recognized by the antibody.
  • a lectin that is suitable for detecting a given SPI 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 SPI in a 2D gel, in a replica of a 2D gel following transfer 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.: PI 1230-050/P11230-150; PI 1120; P38820; P39020), those that bind to phosphoserine (Zymed Laboratories Inc., South San Francisco, CA, catalog no. 61- 8100) and those that bind to phosphothreonine (Zymed Laboratories Inc., South San Francisco, CA, catalog nos. 71-8200, 13-9200).
  • phosphotyrosine BD Transduction Laboratories, catalog nos.: PI 1230-050/P11230-150; PI 1120; P38820; P39020
  • those that bind to phosphoserine Zymed Laboratories Inc., South San Francisco, CA, catalog no. 61- 8100
  • phosphothreonine Zymed Laboratories Inc., South San Francisco, CA, catalog nos. 71-8200, 13-9200
  • a gene encoding an SPI, a related gene, or related nucleic acid sequences or subsequences, including complementary sequences can also be used in hybridization assays.
  • a nucleotide encoding an SPI, 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 encodmg SPIs, or for differential diagnosis of subjects with signs or symptoms suggestive of Schizophrenia.
  • 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 an SPI, under conditions such that hybridization can occur, and detecting or measuring any resulting hybridization.
  • Nucleotides can be used for therapy of subjects having Schizophrenia, as described below. The methods and compositions for clinical screening, diagnosis and prognosis of Schizophrenia in a mammalian subject may be diagnostic of Schizophrenia or indicative of Schizophrenia.
  • Diagnostic methods and compositions are based on Schizophrenia- Associated Features (SFs) and Schizophrenia- Associated Protein Isoforms (SPIs) which are specifically and particularly associated with Schizophrenia and are generally not associated with other diseases or conditions.
  • SFs Schizophrenia- Associated Features
  • SPIs Schizophrenia- Associated Protein Isoforms
  • Such diagnostic SFs or SPis, which are specifically associated with Schizophrenia are useful in screening, diagnosis and prognosis as indicators of
  • Schizophrenia The administration of therapeutic compositions which are directed against or lead to modulation of diagnostic markers may have therapeutic value particularly in Schizophrenia.
  • Schizophrenia but may not be specific only for Schizophrenia, and may be associated with one or more other diseases or conditions.
  • Such indicative SFs or SPis which are associated with Schizophrenia, but not only with Schizophrenia, are useful in screening, diagnosis and prognosis as indicators of Schizophrenia.
  • 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 Schizophrenia, is thereby provided. Additional assessment utilizing diagnostic or particular Schizophrenia SFs or SPIs 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 Schizophrenia 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, SF-329, is provided below in Table VIII.
  • Example of a diagnostic marker for Schizophrenia An indicative marker changes (increases, decreases or otherwise alters form or character) significantly in more than one condition, particularly in Schizophrenia and one or more other distinct diseases or conditions.
  • One such indicative marker, SF-255 is found to increase in Schizophrenia and is provided in Table IX.
  • This same marker, identified or characterised by the same pi and MW, is noted as DF-155 as similarly found to be increased in Bipolar Affective Disorder (BAD) and Unipolar Depression.
  • the SF-255/DF-155 marker is therefore indicative of Schizophrenia and/or Depression.
  • Table IX Example of an indicative marker for Schizophrenia:
  • kits comprising an anti-SPI antibody.
  • a kit may optionally comprise one or more of the following: (1) instructions for using the anti-SPI antibody for diagnosis, prognosis, therapeutic monitoring or any combination of these applications; (2) a labelled binding partner to the antibody; (3) a solid phase (such as a reagent strip) upon which the anti-SPI antibody is immobilized; and (4) a label or insert indicating regulatory approval for diagnostic, prognostic or therapeutic use or any combination thereof.
  • the anti- SPI antibody itself can be labelled 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 an SPI.
  • 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 an SPI, such as by polymerase 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.
  • primers e.g., each in the size range of 6-30 nucleotides, more preferably 10-30 nucleotides and still more preferably 10-20 nucleotides
  • Kits are also provided which allow for the detection of a plurality of SPIs or a plurality of nucleic acids each encoding an SPI.
  • a kit can optionally further comprise a predetermined amount of an isolated SPI protein or a nucleic acid encoding an SPI, e.g., for use as a standard or control.
  • the uni-variate differential analysis tools are useful in identifying individual SFs or SPIs that are diagnostically associated with Schizophrenia or in identifying individual SPIs that regulate the disease process.
  • the disease process is associated with a combination of SFs or SPIs (and to be regulated by a combination of SPIs), rather than individual SFs and SPIs in isolation.
  • the strategies for discovering such combinations of SFs and SPIs differ from those for discovering individual SFs and SPIs. In such cases, each individual SF and SPI 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 SFs or SPIs that individually show significant association with Schizophrenia.
  • the association between the identified SFs or SPIs and Schizophrenia need not be as highly significant as is desirable when an individual SF or SPI 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 Schizophrenia.
  • LDA Linear Discriminant Analysis
  • SFs or SPIs variables
  • Schizophrenia a set of weights is associated with each variable (i.e., SF or SPI) so that the linear combination of weights and the measured values of the variables can identify the disease state by discriminating between subjects having Schizophrenia and subjects free from Schizophrenia.
  • 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 SFs or SPIs which can be used, without limitation, for diagnosis, prognosis, therapy or drug development.
  • LDA Flexible Discriminant Analysis
  • Other enhanced variations of LDA permit the use of non-linear combinations of variables to discriminate a disease state from a normal state.
  • the 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 SFs or SPIs can be identified by qualitative measures by comparing the percentage feature presence of an SF or SPI of one group of samples (e.g., samples from diseased subjects) with the percentage feature presence of an SF or SPI in another group of samples (e.g., samples from control subjects).
  • the "percentage feature presence" of an SF or SPI is the percentage of samples in a group of samples in which the SF or SPI is detectable by the detection method of choice. For example, if an SF is detectable in 95 percent of samples from diseased subjects, the percentage feature presence of that SF in that sample group is 95 percent. If only 5 percent of samples from non-diseased subjects have detectable levels of the same SF, detection of that SF in the sample of a subject would suggest that it is likely that the subject suffers from Schizophrenia.
  • the diagnostic methods and compositions of the present invention can assist in monitoring a clinical study, e.g. to evaluate drugs for therapy of Schizophrenia.
  • candidate molecules are tested for their ability to restore SF or SPI levels in a subject having Schizophrenia to levels found in subjects free from Schizophrenia or, in a treated subject (e.g.
  • SF or SPI levels at or near non-Schizophrenia values.
  • the levels of one or more SFs or SPIs can be assayed.
  • the methods and compositions of the present invention are used to screen candidates for a clinical study to identify individuals having Schizophrenia; such individuals can then be either excluded from or included in 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 Schizophrenia; procedures for these screens are well known in the art.
  • the invention provides isolated mammalian SPIs, preferably human SPIs, 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) SPI, e.g., binding to an SPI substrate or SPI binding partner, antigenicity (binding to an anti-SPI antibody), immunogenicity, enzymatic activity and the like.
  • the invention provides fragments of an SPI 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 an SPI are also provided, as are proteins (e.g., fusion proteins) comprising such fragments. Nucleic acids encoding the foregoing are provided. Once a recombinant nucleic acid which encodes the SPI, a portion of the SPI, or a precursor of the SPI is identified, 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.
  • SPIs 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 SPI 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,
  • native SPIs can be purified from natural sources, by standard methods such as those described above (e.g., immunoaffinity purification).
  • SPIs 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 SPI 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 SPI in a single run.
  • a zoom gel can be used in any separation strategy that employs gel isoelectric focusing.
  • the invention thus provides an isolated SPI, an isolated SPI-related polypeptide, and an isolated derivative or fragment of an SPI or an SPI-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 an SPI or a fragment thereof, or an SPI-related polypeptide, may be synthesized using methods known in the art, such as using conventional chemical approaches or polymerase chain reaction (PCR) amplification.
  • the nucleotide sequences of the present invention also permit the identification and cloning of the gene encoding an SPI homolog or SPI ortholog including, for example, by screening cDNA libraries, genomic libraries or expression libraries.
  • oligonucleotides can be designed for all SPI 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 SPI peptide fragments, using as a template a genomic library or cDNA library pools.
  • Anchored degenerate oligonucleotides can be designed for all SPI 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 an SPI or SPI 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 an SPI.
  • 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 an SPI.
  • 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.
  • 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.
  • 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).
  • YAC yeast artificial chromosome
  • genomic library may be screened by nucleic acid hybridization to labelled probe (Benton and Davis, Science (1977) 196:180; Grunstein and Hogness, Proc. Nat Acad. Sci. USA (1975) 72:3961).
  • the genomic libraries may be screened with labelled degenerate oligonucleotide probes corresponding to the amino acid sequence of any peptide of the SPI 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.
  • SPIs disclosed herein were found to correspond to isoforms of previously identified proteins encoded by genes whose sequences are publicly known. (Sequence analysis and protein identification of SPIs was carried out using the methods described in Section 6.1.14). 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.
  • 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 (NIH) which is available at http://www.ncbi.nlm.nih.gov/GenBank/) provide protein sequences for the SPIs listed in Tables IV and V under the following accession numbers and each sequence is incorporated herein by reference:
  • SPI-206, SPI-238 and SPI-240 the partial sequence information derived from tandem mass spectrometry was not found to be described as a transcribed protein in any known public database.
  • SPI-206, SPI-238 and SPI-240 are therefore listed as 'NOVEL' in Table X.
  • SPI-206, SPI-238 and SPI-240 have been cloned, and are 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 SPI.
  • 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.
  • the probe is 10 nucleotides or longer, more preferably 15 nucleotides or longer.
  • 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 SPI, a fragment of an SPI, an SPI-related polypeptide, or a fragment of an SPI-related polypeptide 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 SPI or SPI-related polypeptides.
  • the various anti- SPI 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 IV. Colonies or plaques from the- library are brought into contact with the antibodies to identify those clones that bind antibody.
  • colonies or plaques containing DNA that encodes an SPI, a fragment of an SPI, an SPI-related polypeptide, or a fragment of an SPI-related polypeptide can be detected using DYNA Beads according to Olsvick et al, 29th ICAAC, Houston, Tex. 1989, incorporated herein by reference.
  • Anti-SPI antibodies are crosslinked to tosylated DYNA Beads M280, and these antibody-containing beads are then contacted with colonies or plaques expressing recombinant polypeptides. Colonies or plaques expressing an SPI or SPI- related polypeptide are identified as any of those that bind the beads.
  • the anti-SPI 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 SPI protein or SPI-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 SPI 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 SPIs 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 polymerase (Gene Amp® or AmpliTaq DNA polymerase).
  • a Perkin-Elmer Cetus thermal cycler and Taq polymerase Gene Amp® or AmpliTaq DNA polymerase.
  • After successful amplification of a segment of the sequence encoding an SPI that segment may be molecularly cloned and sequenced, and utilized as a probe to isolate a complete genomic clone. This, in turn, will permit the determination of the gene's complete nucleotide sequence, the analysis of its expression, and the production of its
  • the gene encoding an SPI 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 an SPI 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 an SPI.
  • a radiolabelled cDNA encoding an SPI 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 an SPI from among other genomic DNA fragments.
  • RNA for cDNA cloning of the gene encoding an SPI can be isolated from cells that express the SPI.
  • Any suitable eukaryotic cell can serve as the nucleic acid source for the molecular cloning of the gene encoding an SPI.
  • the nucleic acid sequences encoding the SPI 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 retroviruses.
  • 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 an SPI 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 SPI, 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.
  • the micleotide sequences of the present invention include nucleotide sequences encoding amino acid sequences with substantially the same amino acid sequences as native SPIs, nucleotide sequences encoding amino acid sequences with functionally equivalent amino acids, nucleotide sequences encoding SPIs, a fragments of SPIs, SPI- related polypeptides, or fragments of SPI-related polypeptides.
  • an isolated nucleic acid molecule encoding an SPI- related polypeptide can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of an SPI 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.
  • SPI-206 was isolated, subjected to proteolysis, and analyzed by mass spectrometry using the methods and apparatus of the Preferred Technology.
  • SEQUEST search program as described in the Examples, infra, uninterpreted tandem mass spectra of tryptic digest peptides were searched against a database of public domain proteins constructed of protein entries in the non-redundant database held by the National Centre for Biotechnology Info ⁇ nation (NCBI) which is accessible at http://www.ncbi.nlm.nih.gov/.
  • 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 the possible amino acid sequence is listed for a single 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 ⁇ l 000000).
  • 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 (H 2 O) and a single proton (H ).
  • the "mass to the 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 the 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 (H 2 O), and a single proton (H 1" ).
  • the partial amino acid sequence and masses listed in Table XI were found to correspond to a peptide within the same putative human protein identified using SEQUEST, (i.e. protein ID CAA15430.1, accessible at http://www.ncbi.nlm.nih.gov/entrez/).
  • SEQUEST i.e. protein ID CAA15430.1, accessible at http://www.ncbi.nlm.nih.gov/entrez/.
  • the full amino acid sequence of the peptide listed in Table XI as a result of the match was found to be : GILILGQEQDTLGGR (shown in Figure 2).
  • the human protein of database accession number CAAl 5430.1 the neuronal pentraxin receptor, whose gene is located on chromosome 22ql2.3-13.2 (accession ID AL008583), is an ortholog of rat neuronal pentraxin receptor (Kirkpatrick LL, Matzuk MM, Dodds DC, Perin MS. Biochemical interactions of the neuronal pentraxins.
  • Neuronal pentraxin (NP) receptor binds to taipoxin and taipoxin-associated calcium-binding protein 49 via NP1 and NP2. f Biol Chem.
  • NPR Neuronal pentraxin receptor
  • a nucleotide sequence ( Figure 2B) encoding a peptide ( Figure 2A) comprising the above two peptides can be cloned by using the following primers:
  • the peptide of Figure 2A has 90% homology with the rat polypeptide (AAB62885).
  • the rat protein a putative integral membrane pentraxin, has 49 and 48% identity to neuronal pentraxin 1 (NP1) and neuronal pentraxin 2 (NP2), respectively (Dodds DC et al, supra; Hsu, et al, 1995, Genomics 28:2, 220-227). Theses proteins are suggested to be constituents of a pathway involved in the clearance of synaptic debris. Addition of NP1 to glial cultures renders them susceptible to a neurotoxin toxicity (Dodds DC et al, supra).
  • NPR is expressed on the cell membrane and can form heteropentamers with NPl and NP2 that can be released from cell membranes (Kirkpatrick et al, supra).
  • NPl has homology to previously identified pentraxins, such as serum amyloid P protein (Dodds DC et al, supra). Serum amyloid P protein has been widely described for its role in amyloid (Botto M, et al, Nat Med. 1997 Aug;3(8):855-9; International Patent Publication WO9505394), and in particular its implication in Alzheimer disease progression (Tennent GA, Lovat LB, Pepys MB.
  • Serum amyloid P component prevents proteolysis of the amyloid fibrils of Alzheimer disease and systemic amyloidosis. Proc Natl Acad Sci U S A. 1995 May 9;92(10):4299-303) and diagnostic (Hawkins PN, Rossor MN, Gallimore JR, Miller B, Moore EG, Pepys MB. Concentration of serum amyloid P component in the CSF as a possible marker of cerebral amyloid deposits in Alzheimer's disease. Biochem Biophys Res Commun. 1994 Jun 15;201(2):722-6) as well as other degenerative disorders (Kalaria RN, Galloway PG, Perry G.
  • the peptide of Figure 2A which has 48% homology with serum amyloid P, and which is increased by 1.92 fold in Schizophrenia-affected patients, can be used for screening diagnosis, prognosis of Schizophrenia according to the methods of the invention.
  • Any of the activities described in the above references concerning amyloid P, NARP, NPR, NPl and NP2 can be used as the basis for functional assays for compounds capable of inhibiting or stimulating the. relevant activity of the peptide of Figure 2A.
  • compounds capable of modulating the activities of amyloid P, NARP, NPR, NPl and NP2 are candidate compounds for the treatment of Alzheimer's disease.
  • assays for such compounds may also be performed, by, for example, recombinantly expressing amyloid P, NARP, NPR, NPl or NP2 and testing for compounds capable of modulating the activity of the expressed protein.
  • the peptide of Figure 2A may be recombinantly expressed by constructing an expression vector comprising the nucleic acid sequence of Figure 2B, or portions thereof.
  • the expressed recombinant protein may be used in assays for compounds capable of modulating the activity of the recombinant protein.
  • assays for compounds capable of inhibiting or. stimulating the cleavage of the peptide of Figure 2A may be performed.
  • only a portion of the peptide is recombinantly produced.
  • the portion of the peptide produced comprises a cleavage site.
  • the portion of the peptide comprises amino acid residues 26 to 46, or residues 237 to 247.
  • a truncated peptide comprising or consisting of the carboxyl terminus is produced, e.g., from amino acid residue 28, 36, 237, 243, 264 or 327 to the carboxyl terminus of the peptide.
  • SPI-238 and SPI-240 were each isolated, subjected to proteolysis, and analyzed by mass spectrometry using the methods and apparatus of the Preferred Technology.
  • SEQUEST search program as described in the Examples, infra, uninterpreted tandem mass spectra of tryptic digest peptides were searched against a database of public domain proteins 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/ and also constructed of Expressed Sequence Tags entries
  • 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.
  • the amino acid sequence P[L/I]A indicates the amino acid sequences PLA and PIA.
  • a sequence containing n designated pairs indicates 2" amino acid sequences. For both SPI-238 and SPI- 240 the same partial sequence was determined by mass spectrometry and is listed in Table XII.
  • 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 (H 2 O) 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 (H 2 O), and a single proton (H + ).
  • H 2 O water molecular
  • H + single proton
  • 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. Biol.215; 403-410. ).
  • This EST sequence does not overlap with EST AA326679 so that the possibility remained that the partial amino acid sequence and masses listed in Table XII could be encoded by the no-overlapping region of these 2 ESTs.
  • PCR primers (1 & 2 from Table XIII) from EST AA326679 and EST AI589129 were used in a PCR reaction (1 ml of Advantage 2 cDNA polymerase mix (Clontech) in a buffer containing 50mM KC1, 10 mM Tris-HCl, 1.5 mM MgC12, pH8.3; 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 Gene Amp Systems 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 XIII (1,2, 3 & 4). This generated overlapping sequence for the entire product.
  • Figure 4B shows the DNA sequence.
  • Figure 4A show the protein sequence of the open reading frame (ORF), demonstrating the presence of the two peptides identified by mass spectrometry.
  • gcc ate cat eta gac eta gaa gaa tac egg Ala lie His Leu Asp Leu Glu Glu Tyr Arg
  • SPI-238 and SPI-240 was also found to be differentially present in samples of CSF from subjects having BAD compared with samples of CSF from subjects free from BAD, being decreased 2.27 fold (in the copending US patent application number 60/254830 which is incorporated herein by reference).
  • the patent WO99/58660 disclosed 97 human secreted proteins. These included a sequence, identified as Gene No: 21, which corresponds to SPI-238 and SPI-240 discussed herein.
  • the nucleotide sequence coding for an SPI, an SPI analog, an SPI-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 SPI or its flanking regions, or the native gene encoding the SPI- 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 SPI
  • a fragment of an SPI comprising a domain of the SPI 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 an SPI or fragment thereof may be regulated by a second nucleic acid sequence so that the SPI or fragment is expressed in a host transformed with the recombinant DNA molecule. For example, expression of an SPI 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 an SPI or an SPI- related polypeptide include, but are not limited to, the SV40 early promoter region (Bernoist and Chambon Nature (1981) 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al, Cell (1980) 22:787- 797), the herpes thymidine kinase promoter (Wagner et al, Proc. Natl. Acad. Sci.
  • 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, Cell (1984) 38:639-646; Ornitz et al, Cold Spring Harbor Symp.
  • mice mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al, Cell (1986) 45:485-495), albumin gene control region which is active in liver (Pinkert et al, Genes and Devel. (1987) 1:268-276), .alpha-fetoprotein gene control region which is active in liver (Krumla ⁇ f et al, Mol. Cell. Biol. (1985) 5:1639-1648; Hammer et al, Science (1987) 235:53- 58; alpha 1-antitrypsin gene control region which is active in the liver (Kelsey et al, Genes and Devel.
  • beta-globin gene control region which is active in myeloid cells (Mogram et al, Nature (1985) 315:338-340; Kollias et al, Cell (1986) 46:89-94; myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al, Cell (1987) 48:703-712); myosin light chain-2 gene control region which is active in skeletal muscle (Sani, Nature (1985) 314:283-286); neuronal-specific enolase (NSE) which is active in neuronal cells (Morelli et al, Gen. Virol.
  • NSE neuronal-specific enolase
  • BDNF brain-derived neurotrophic factor
  • GFAP glial fibrillary acidic protein
  • a vector in a specific embodiment, comprises a promoter operably linked to an SPI-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 an SPI or an SPI-related polypeptide coding sequence into the EcoRI restriction site of each of the three pGEX vectors (Glutathione S-Transferase expression vectors; Smith and Johnson, Gene (1988) 7:31-40). This allows for the expression of the SPI product or SPI-related polypeptide from the subclone in the correct reading frame.
  • a number of viral-based expression systems may be utilized.
  • the SPI coding sequence or SPI- 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, Proc. Natl. Acad. Sci. USA (1984) 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. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al, Methods in Enzymol. (1987) 153:51-544).
  • Expression vectors containing inserts of a gene encoding an SPI or an SPI- 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 an SPI inserted in an expression vector can be detected by nucleic acid hybridization using probes comprising sequences that are homologous to an inserted gene encoding an SPI.
  • 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 an SPI 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., SPI) expressed by the recombinant.
  • SPI gene product expressed by the recombinant.
  • assays can be based, for example, on the physical or functional properties of the SPI in in vitro assay systems, e.g., binding with anti- SPI 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 SPI or SPI-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, J. Natl. Cancer Inst. (1984) 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, Cell (1977) 11:223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Nat Acad. Sci. USA (1962) 48:2026), and adenine phosphoribosyltransferase (Lowy, et al, Cell (1980) 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 methotrexate (Wigler, et al, Proc. Natl. Acad. Sci. USA (1980) 77:3567; O'Hare, et al, Proc. Natl. Acad. Sci. USA (1981) 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci. USA (1981) 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre- Garapin, et al, J. Mol. Biol. (1981) 150:1); and hygro, which confers resistance to hygromycin (Santerre, et al, Gene (1984) 30:147) genes.
  • the SPI, 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.
  • immunoglobulins IgA, IgE, IgG, IgM
  • CHI constant domain of immunoglobulins
  • 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.
  • Nucleic acids encoding an SPI, a fragment of an SPI, an SPI-related polypeptide, or a fragment of an SPI-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, Proc. Natl. Acad. Sci. USA (1991) 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 SPIs are known in the art and have been described in the scientific literature. Moreover, domains of an SPI can be identified using techniques known to those of skill in the art. For example, one or more domains of an SPI 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., Nucleic Acids Res., (1999) 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, Proc. Natl. Acad. Sci. USA (1992) 89: 2002-2006).
  • the skilled artisan can identify domains of an SPI 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 an SPI fragment that retains the enzymatic or binding activity of the SPI.
  • the skilled artisan can identify domains of an SPI 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
  • an SPI 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.
  • An SPI domain can be assessed for its function using techniques well known to those of skill in the art. For example, 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 an SPI is determined using an assay described in one or more of the references identified in Table XIV, infra.
  • an SPI, SPI analog, SPI-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 SPIs are publicly available.
  • antibodies that recognize these SPIs and/or their isoforms include antibodies recognizing: SPI-6, SPI-8, SPI-9, SPI-10, SPI-15, SPI-16, SPI-18, SPI-23, SPI-32, SPI-33, SPI-34, SPI-35, SPI-41, SPI-42, SPI-43, SPI-54, SPI-57, SPI-60, SPI-62, SPI-63, SPI-67, SPI-73, SPI-74, SPI-75, SPI-76, SPI-77, SPI-78, SPI-80, SPI-82, SPI-91, SPI-92, SPI-93, SPI-96, SPI-97, SPI-99, SPI-100, SPI-101, SPI-105, SPI-107, SPI- 113, SPI-114, SPI-115, SPI-122, SPI-
  • antibodies to a specific domain of an SPI are produced.
  • hydrophilic fragments of an SPI 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 SPI homolog but which does not specifically bind to (or binds less avidly to) a second SPI homolog one can select on the basis of positive binding to the first SPI homolog and a lack of binding to (or reduced binding to) the second SPI homolog.
  • the present invention provides an 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 an SPI than to a different isoform or isoforms (e.g., glycoforms) of the SPI.
  • Polyclonal antibodies that 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 an SPI, a fragment of an SPI, an SPI-related polypeptide, or a fragment of an SPI-related polypeptide. In a particular embodiment, rabbit polyclonal antibodies to an epitope of an SPI or an SPI- related polypeptide can be obtained.
  • various host animals can be immunized by injection with the native or a synthetic (e.g., recombinant) version of an SPI, a fragment of an SPI, an SPI-related polypeptide, or a fragment of an SPI-related polypeptide, including but not limited to rabbits, mice, rats, etc.
  • the Preferred Technology described herein provides isolated SPIs suitable for such immunization. If the SPI is purified by gel electrophoresis, the SPI 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 adjuvant, 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 s ⁇ ch 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 (Nature (1975) 256:495-497), as well as the trioma technique, the human B-cell hybridoma technique (Kozbor et al, Immunology Today (1983) 4:72), and the EBV- hybridoma technique to produce human monoclonal antibodies (Cole 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 (PCTVUS90/02545, 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, Proc. Natl. Acad. Sci. USA (1987) 84:3439-3443; Liu et al, J. Immunol. (1987) 139:3521-3526; Sun et al, Proc.
  • 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 an SPI 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.
  • Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a mouse antibody
  • the antibodies of the present invention can also be generated using various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles that carry the polynucleotide sequences encodmg them.
  • such 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 labelled 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 disulf ⁇ de 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 (1995) 182:41-50 ; Ames et al, J. Immunol. Methods (1995) 184:177-186 ; Kettleborough et al, Eur. J. Immunol. (1994) 24:952-958 ; Persic et al, Gene (1997) 187 9-18 ; Burton et al, Advances in Immunology (1994) 57:191-280 ; 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.
  • 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, Nature (1983) 305:537-539). Because of the random 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, EMBOJ. (1991) 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.
  • the invention provides functionally active fragments, derivatives or analogs of the anti-SPI immunoglobulin molecules.
  • Functionally active means that the fragment, derivative or analog is able to elicit anti-anti-idiotype antibodies (z ' .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.
  • synthetic peptides containing the CDR sequences can be used in binding assays with the antigen by any binding assay method known in the art.
  • 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, Science
  • 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. coli may be used
  • 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.
  • 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.
  • the 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.
  • 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 SPIs of the invention, e.g., for imaging these proteins, measuring levels thereof in appropriate physiological samples, in diagnostic methods, etc.
  • 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, BioTechniques (1994) 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, Science (1989) 246:1275-1281) for clones of Fab fragments that bind the specific antigen or by screening antibody libraries (See, e.g., Clackson et al, Nature (1991) 352:624; Hane et al, Proc. Natl. Acad. Sci. USA (1997) 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 chain 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, J. Biol. Chem. (1978) 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.
  • 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, Gene (1986) 45:101; Cockett et al, Bio/Technology (1990) 8:2).
  • host-expression vector systems may 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., metallothionein promoter) or from ma
  • 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, EMBO J. (1983) 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, Nucleic Acids Res.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • 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 control of an AcNPV promoter (for example the polyhedrin promoter).
  • an AcNPV promoter for example the polyhedrin promoter.
  • a number of viral-based expression systems e.g., an adenovirus expression system may be utilized.
  • 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.
  • cells lines that stably express an antibody of interest can be produced by transfecting 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
  • Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.
  • 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 (Grouse 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, Nature (1986) 322:52; Kohler, Proc. Natl. Acad. Sci. USA (1980) 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.
  • any fusion protein may be readily purified by utilizing an antibody specific for the fusion protein being expressed. For example, 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, Proc. Natl.
  • 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-SPI 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. Examples of 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 acetylcholinesterase; 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-SPI 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 heteroconjugate 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 Schizophrenia can be used for diagnosis or monitoring.
  • CSF cerebrospinal fluid
  • serum, plasma or urine obtained from a subject suspected of having or known to have Schizophrenia can be used for diagnosis or monitoring.
  • a decreased abundance of one or more SFs or SPIs (or any combination of them) in a test sample relative to a control sample (from a subject or subjects free from Schizophrenia) or a previously determined reference range indicates the presence of Schizophrenia; SFs and SPIs suitable for this purpose are identified in Tables I and IV, respectively, as described in detail above.
  • an increased abundance of one or more SFs or SPIs (or any combination of them) in a test sample compared to a control sample or a previously determined reference range indicates the presence of Schizophrenia; SFs and SPIs suitable for this purpose are identified in Tables II and V, respectively, as described in detail above.
  • the relative abundance of one or more SFs or SPIs (or any combination of them) in a test sample compared to a control sample or a previously determined reference range indicates a subtype of Schizophrenia (e.g., familial or sporadic Schizophrenia).
  • the relative abundance of one or more SFs or SPIs (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 Schizophrenia.
  • detection of one or more SPIs described herein may optionally be combined with detection of one or more additional biomarkers for Schizophrenia including, but not limited to.
  • any suitable method in the art can be employed to measure the level of SFs and SPIs, including but not limited to the Preferred Technology described herein, kinase assays, immunoassays to detect and/or visualize the SPI (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 SPI expression.
  • a decreased abundance of mRNA encoding one or more SPIs identified in Table IV (or any combination of them) in a test sample relative to a control sample or a previously determined reference range indicates the presence of Schizophrenia.
  • an increased abundance of mRNA encoding one or more SPIs 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 Schizophrenia.
  • Any suitable hybridization assay can be used to detect SPI expression by detecting and/or visualizing mRNA encoding the SPI (e.g., Northern assays, dot blots, in situ hybridization, etc.).
  • labelled antibodies, derivatives and analogs thereof, which specifically bind to an SPI can be used for diagnostic purposes to detect, diagnose, or monitor Schizophrenia.
  • Schizophrenia 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 an SPI or have a stimulatory or inhibitory effect on the expression or activity of an SPI.
  • the invention also provides methods of identifying agents, candidate compounds or test compounds that bind to an SPI-related polypeptide or an SPI fusion protein or have a stimulatory or inhibitory effect on the expression or activity of an SPI-related polypeptide or an SPI 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.
  • biological libraries include 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, Anticancer Drug Des. (1997) 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).
  • agents that interact with (i.e., bind to) an SPI, an SPI fragment (e.g. a functionally active fragment), an SPI-related polypeptide, a fragment of an SPI-related polypeptide, or an SPI fusion protein are identified in a cell-based assay system.
  • cells expressing an SPI, a fragment of an SPI, an SPI- related polypeptide, a fragment of an SPI-related polypeptide, or an SPI fusion protein are contacted with a candidate compound or a control compound and the ability of the ' candidate compound to interact with the SPI is determined.
  • this assay may be used to screen a plurality (e.g. a library) of candidate compounds.
  • 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 SPI, fragment of the SPI, SPI-related polypeptide, a fragment of the SPI- related polypeptide, or an SPI fusion protein endogenously or be genetically engineered to express the SPI, fragment of the SPI, SPI-related polypeptide, a fragment of the SPI- related polypeptide, or an SPI fusion protein.
  • prokaryotic origin e.g., E. coli
  • eukaryotic origin e.g., yeast or mammalian.
  • the cells can express the SPI, fragment of the SPI, SPI-related polypeptide, a fragment of the SPI- related polypeptide, or an SPI fusion protein endogenously or be genetically engineered to express the SPI, fragment of the SPI, SPI-related polypeptide, a fragment of the
  • the SPI, fragment of the SPI, SPI- related polypeptide, a fragment of the SPI-related polypeptide, or an SPI fusion protein or the candidate compound is labelled, for example with a radioactive label (such as P, S or I) or a fluorescent label (such as fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde or fluorescamine) to enable detection of an interaction between an SPI and a candidate compound.
  • a radioactive label such as P, S or 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 an SPI, a fragment of an SPI, an SPI-related polypeptide, a fragment of an SPI- related polypeptide, or ah SPI fusion protein can be determined by methods known to those of skill in the art.
  • the interaction between a candidate compound and an SPI, a fragment of an SPI, an SPI-related polypeptide, a fragment of an SPI- related polypeptide, or an SPI fusion protein can be determined by flow cytometry, a scintillation assay, immunoprecipitation or western blot analysis.
  • agents that interact with (i.e., bind to) an SPI, an SPI fragment (e.g., a functionally active fragment) an SPI-related polypeptide, a fragment of an SPI-related polypeptide, or an SPI fusion protein are identified in a cell-free assay system.
  • a native or recombinant SPI or fragment thereof, or a native or recombinant SPI-related polypeptide or fragment thereof, or an SPI-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 SPI or SPI-related polypeptide, or SPI fusion protein is determined.
  • this assay may be used to screen a plurality (e.g. a library) of candidate compounds.
  • the SPI, SPI fragment, SPI-related polypeptide, a fragment of an SPI-related polypeptide, or an SPI-fusion protein is first immobilized, by, for example, contacting the SPI, SPI fragment, SPI-related polypeptide, a fragment of an SPI- related polypeptide, or an SPI fusion protein with an immobilized antibody which specifically recognizes and binds it, or by contacting a purified preparation of the SPI, SPI fragment, SPI- related polypeptide, fragment of an SPI-related polypeptide, or an SPI fusion protein with a surface designed to bind proteins.
  • the SPI, SPI fragment, SPI-related polypeptide, a fragment of an SPI-related polypeptide, or an SPI 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 SPI, SPI fragment, SPI-related polypeptide, a fragment of an SPI-related polypeptide may be a fusion protein comprising the SPI or a biologically active portion thereof, or SPI-related polypeptide and a domain such as glutathionine- S-transferase.
  • the SPI, SPI fragment, SPI-related polypeptide, fragment of an SPI-related polypeptide or SPI 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 an SPI, SPI fragment, SPI-related polypeptide, a fragment of an SPI-related polypeptide, or an SPI 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 an SPI or is responsible for the post- translational modification of an SPI.
  • a protein such as an enzyme, or a biologically active portion thereof, which is responsible for the production or degradation of an SPI or is responsible for the post- translational modification of an SPI.
  • a plurality e.g., a library
  • cells that naturally or recombinantly express: (i) an SPI, an isoform of an SPI, an SPI homolog an SPI-related polypeptide, an SPI fusion protein, or a biologically active fragment of any of the foregoing; and (ii) a protein that is responsible for processing of the SPI, SPI isoform, SPI homolog, SPI-related polypeptide, SPI fusion protein, or fragment in order to identify compounds that modulate the production, degradation, or post-translational modification of the SPI, SPI isoform, SPI homolog, SPI-related polypeptide, SPI 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 SPI of interest.
  • the ability of the candidate compound to modulate the production, degradation or post-translational modification of an SPI, isoform, homolog, SPI- related polypeptide, or SPI 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) an SPI, SPI fragment, SPI-related polypeptide, a fragment of an SPI-related polypeptide, or an SPI fusion protein are identified in a competitive binding assay.
  • cells expressing an SPI, SPI fragment, SPI-related polypeptide, a fragment of an SPI-related polypeptide, or an SPI fusion protein are contacted with a candidate compound and a compound known to interact with the SPI, SPI fragment, SPI-related polypeptide, a fragment of an SPI-related polypeptide or an SPI fusion protein; the ability of the candidate compound to competitively interact with the SPI, SPI fragment, SPI-related polypeptide*, fragment of an SPI-related polypeptide, or an SPI fusion protein is then determined.
  • agents that competitively interact with (i.e., bind to) an SPI, SPI fragment, SPI- related polypeptide or fragment of an SPI-related polypeptide are identified in a cell-free assay system by contacting an SPI, SPI fragment, SPI-related polypeptide, fragment of an SPI-related polypeptide, or an SPI fusion protein with a candidate compound and a compound known to interact with the SPI, SPI-related polypeptide or SPI fusion protein.
  • the ability of the candidate compound to interact with an SPI, SPI fragment, SPI- related polypeptide, a fragment of an SPI-related polypeptide, or an SPI 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 an SPI, or an SPI-related polypeptide are identified by contacting cells (e.g., cells of prokaryotic origin or eukaryotic origin) expressing the SPI, or SPI- related polypeptide with a candidate compound or a control compound (e.g., phosphate buffered saline (PBS)) and determining the expression of the SPI, SPI-related polypeptide, or SPI fusion protein, mRNA encoding the SPI, or mRNA encoding the SPI-related polypeptide.
  • a candidate compound or a control compound e.g., phosphate buffered saline (PBS)
  • the level of expression of a selected SPI, SPI-related polypeptide, mRNA encoding the SPI, or mRNA encoding the SPI-related polypeptide in the presence of the candidate compound is compared to the level of expression of the SPI, SPI-related polypeptide, mRNA encoding the SPI, or mRNA encoding the SPI-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 SPI, or an SPI-related polypeptide based on this comparison.
  • the candidate compound when expression of the SPI 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 SPI or mRNA.
  • the candidate compound when expression of the SPI 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 SPI or mRNA.
  • the level of expression of an SPI 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 an SPI, or an SPI- related polypeptide are identified by contacting a preparation containing the SPI or SPI-related polypeptide, or cells (e.g., prokaryotic or eukaryotic cells) expressing the SPI or SPI-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 SPI or SPI-related polypeptide.
  • the activity of an SPI or an SPI-related polypeptide can be assessed by detecting induction of a cellular signal transduction pathway of the SPI or SPI-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 an SPI or an SPI-related polypeptide and is operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cellular differentiation, or cell proliferation.
  • a reporter gene e.g., a regulatory element that is responsive to an SPI or an SPI-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 an SPI or SPI-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 an SPI or SPI-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 Schizophrenia (e.g.,Phencyclidine treated rodents (Sams-Dodd Rev
  • the 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 SPI or SPI-related polypeptide is determined. Changes in the expression of an SPI or SPI-related polypeptide can be assessed by the methods outlined above.
  • an SPI or SPI-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 an SPI or SPI-related polypeptide (see, e.g., U.S. Patent No.
  • binding proteins are also likely to be involved in the propagation of signals by the SPIs of the invention as, for example, upstream or downstream elements of a signaling pathway involving the SPIs of the invention.
  • Table XIV enumerates scientific publications describing suitable assays for detecting or quantifying enzymatic or binding activity of an SPI, an SPI analog, an SPI- related polypeptide, or a fragment of any of the foregoing. Each such reference is hereby incorporated in its entirety.
  • as assay referenced in Table XIV 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) an SPI, SPI analog, or SPI-related polypeptide, a fragment of any of the foregoing.
  • 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: SPIs, SPI analogs, SPI-related polypeptides and derivatives (including fragments) thereof; antibodies to the foregoing; nucleic acids encoding SPIs, SPI analogs, SPI-related polypeptides and fragments thereof; antisense nucleic acids to a gene encoding an SPI or SPI- related polypeptide; and modulator (e.g., agonists and antagonists) of a gene encoding an SPI or SPI-related polypeptide.
  • An important feature of the present invention is the identification of genes encoding SPIs involved in Schizophrenia. Schizophrenia can be treated (e.g.
  • one or more antibodies each specifically binding to an SPI are administered alone or in combination with one or more additional therapeutic compounds or treatments.
  • additional therapeutic compounds or treatments include, but are not limited to, Sertindole, Haloperidol, Pirenzepine, Perazine, Risperdal, Famotidine, Clozaril, Mesoridazine, Quetiapine, atypical anti-psychotic medications of Risperidone, Zyperexa (Olanzapine) and Clozapine and any other Dibenzothiazepines.
  • the compounds of the invention may be given in combination with any other compound, including Sertindole, Haloperidol, Pirenzepine, Perazine, Risperdal, Famotidine, Clozaril, Mesoridazine, Quetiapine, atypical anti-psychotic medications of Risperidone, Zyperexa (Olanzapine) and Clozapine and any other Dibenzothiazepines.
  • a biological product such as an antibody is allogeneic to the subject to which it is administered.
  • a human SPI or a human SPI- related polypeptide, a nucleotide sequence encoding a human SPI or a human SPI- related polypeptide, or an antibody to a human SPI or a human SPI-related polypeptide is administered to a human subject for therapy (e.g. to ameliorate symptoms or to retard onset or progression) or prophylaxis.
  • Schizophrenia is treated or prevented by administration to a subject suspected of having or known to have Schizophrenia or to be at risk of developing Schizophrenia of a compound that modulates (i.e., increases or decreases) the level or activity (i.e., function) of one or more SPIs or the level of one or more SFs that are differentially present in the CSF of subjects having Schizophrenia compared with CSF of subjects free from Schizophrenia.
  • Schizophrenia is treated or prevented by administering to a subject suspected of having or known to have Schizophrenia or to be at risk of developing Schizophrenia a compound that upregulates (i.e., increases) the level or activity (i.e., function) of one or more SPIs or the level of one or more SFs that are decreased in the CSF of subjects having Schizophrenia.
  • a compound is administered that downregulates the level or activity (i.e., function) of one or more SPIs or the level of one or more SFs that are increased in the CSF of subjects having Schizophrenia.
  • Examples of such a compound include but are not limited to: SPIs, SPI fragments and SPI-related polypeptides; nucleic acids encoding an SPI, an SPI fragment and an SPI-related polypeptide (e.g., for use in gene therapy); and, for those SPIs or SPI-related polypeptides with enzymatic activity, compounds or molecules known to modulate that enzymatic activity.
  • Schizophrenia is also treated or prevented by administration to a subject suspected of having or known to have Schizophrenia or to be at risk of developing Schizophrenia of a compound that downregulates the level or activity of one or more SPIs or the level of one or more SFs that are increased in the CSF of subjects having Schizophrenia.
  • a compound is administered that upregulates the level or activity of one or more SPIs or the level of one or more SFs that are decreased in the CSF of subjects having Schizophrenia.
  • Examples of such a compound include, but are not limited to, SPI antisense oligonucleotides, ribozymes, antibodies directed against SPIs, and compounds that inhibit the enzymatic activity of an SPI.
  • SPI antisense oligonucleotides e.g., SPI antisense oligonucleotides, ribozymes, antibodies directed against SPIs, and compounds that inhibit the enzymatic activity of an SPI.
  • Other useful compounds e.g., SPI antagonists and small molecule SPI 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 SPIs, or the level of one or more SFs are therapeutically or prophylactically administered to a subject suspected of having or known to have Schizophrenia, in whom the levels or functions of said one or more SPIs, or levels of said one or more SFs, are absent or are decreased relative to a control or normal reference range.
  • compounds that promote the level or function of one or more SPIs, or the level of one or more SFs are therapeutically or prophylactically administered to a subject suspected of having or known to have Schizophrenia in whom the levels or functions of said one or more SPIs, or levels of said one or more SFs, are increased relative to a control or to a reference range.
  • compounds that decrease the level or function of one or more SPIs, or the level of one or more SFs are therapeutically or prophylactically administered to a subject suspected of having or known to have Schizophrenia in whom the levels or functions of said one or more SPIs, or levels of said one or more SFs, are increased relative to a control or to a reference range.
  • compounds that decrease the level or function of one or more SPIs, or the level of one or more SFs are therapeutically or prophylactically administered to a subject suspected of having or known to have Schizophrenia in whom the levels or functions of said one or more SPIs, or levels of said one or more SFs, are decreased relative to a control or to a reference range.
  • the change in SPI function or level, or SF 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 SFs or the levels or activities of said SPIs, or the levels of mRNAs encoding said SPIs. 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.
  • nucleic acids comprising a sequence encoding an SPI, an SPI fragment, SPI-related polypeptide or fragment of an SPI-related polypeptide, are administered to promote SPI 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 SPI function.
  • the compound comprises a nucleic acid encoding an SPI or fragment or chimeric protein thereof, said nucleic acid being part of an expression vector that expresses an SPI or fragment or chimeric protein thereof in a suitable host.
  • a nucleic acid has a promoter operably linked to the SPI coding region, said promoter being inducible or constitutive (and, optionally, tissue-specific).
  • a nucleic acid molecule is used in which the SPI 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 SPI nucleic acid (Koller and Smithies, Proc. Natl. Acad. Sci. USA (1989) 86:8932-8935; Zijlstra et al, Nature (1989) 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. 4,980,286); by
  • microparticle bombardment e.g., a gene gun; Biolistic, Dupont
  • lipids, cell-surface receptors or transfecting agents e.g., lipids, cell-surface receptors or transfecting agents
  • encapsulation in liposomes, microparticles or microcapsules by administering it in linkage to a peptide which is known to enter the nucleus; or by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see, e.g., Wu and W ⁇ , J. Biol. Chem. (1987) 262:4429- 4432), which can be used to target cell types specifically expressing the receptors.
  • 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,); W093/14188 dated July 22, 1993 (Clarke et al,), WO 93/20221 dated October 14, 1993 (Young)).
  • the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Roller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al, Nature (1989) 342:435-438).
  • a viral vector that contains a nucleic acid encoding an SPI is used.
  • a retroviral vector can be used (see Miller et al, Meth. Enzymol. (1993) 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 SPI 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, Biotherapy (1994) 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, J. Clin. Invest. (1994) 93:644-651; Kiem et al, Blood (1994) 83:1467-1473; Salmons and Gunzberg, Human Gene Therapy (1993) 4:129-141; and Grossman and Wilson, Curr. Opin. in Genetics and Devel. (1993) 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, Current Opinion in Genetics and Development (1993) 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, Proc. Soc. Exp. Biol. Med. (1993) 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, Meth. Enzymol. (1993) 217:599-618; Cohen et al, 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, granulocytes; 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.
  • the cell used for gene therapy is autologous to the subject that is treated.
  • a nucleic acid encoding an SPI 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, Meth. Cell Bio. (1980) 21A:229; and Pittelkow and Scott, Mayo Clinic Proc. (1986) 61:771).
  • 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 the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
  • Direct injection of a DNA coding for an SPI may also be performed according to, for example, the techniques described in United States Patent No. 5,589,466. These techniques involve the injection of "naked DNA", i.e., isolated DNA molecules in the absence of liposomes, cells, or any other material besides a suitable carrier.
  • naked DNA comprising (a) DNA encoding an SPI and (b) a promoter are injected into a subject to elicit an immune response to the SPI.
  • Schizophrenia is treated or prevented by administration of a compound that antagonizes (inhibits) the level(s) and/or function(s) of one or more SPIs which are elevated in the CSF of subjects having Schizophrenia as compared with CSF of subjects free from Schizophrenia.
  • Compounds useful for this purpose include but are not limited to anti-SPI antibodies (and fragments and derivatives containing the binding region thereof), SPI antisense or ribozyme nucleic acids, and nucleic acids encoding dysfunctional SPIs that are used to "knockout" endogenous SPI function by homologous recombination (see, e.g., Capecchi, Science (1989) 244:1288-1292).
  • Other compounds that inhibit SPI 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 an SPI to another protein or a binding partner, or to inhibit a known SPI 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 SPI before and after the administration 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 an SPI function is administered therapeutically or prophylactically to a subject in whom an increased CSF level or functional activity of the SPI (e.g., greater than the normal level or desired level) is detected as compared with CSF of subjects free from Schizophrenia or a predetermined reference range.
  • Preferred SPI 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.
  • SPI expression is inhibited by use of SPI 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 an SPI or a portion thereof.
  • an SPI "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 an SPI.
  • the antisense nucleic acid may be complementary to a coding and/or noncoding region of an mRNA encoding an SPI.
  • Such antisense nucleic acids have utility as compounds that inhibit SPI expression, and can be used in the treatment or prevention of Schizophrenia.
  • 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 SPI antisense nucleic acids of the invention in a pharmaceutically acceptable carrier, as described infra.
  • the invention provides methods for inhibiting the expression of an SPI nucleic acid sequence in a prokaryotic or eukaryotic cell comprising providing the cell with an effective amount of a composition comprising an SPI antisense nucleic acid of the invention.
  • SPI antisense nucleic acids and their uses are described in detail below.
  • the SPI 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, Proc. Natl. Acad. Sci. USA (1989) 86:6553-6556; Lemaitre et al, Proc. Natl. Acad. Sci. USA (1987) 84:648-652; PCT Publication No. WO 88/09810, published December 15, 1988) or blood-brain barrier (see, e.g., PCT Publication No.
  • an SPI 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 SPI 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-carboxymethylaminomethyluracil, 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-mannosylqueosine, 5'-
  • 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.
  • Ah ⁇ -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, (Nucl. Acids Res. (1988) 16:3209), and methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al, Proc. Natl. Acad. Sci. USA (1988) 85:7448-7451).
  • the SPI 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 SPI 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 SPI 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 an SPI, preferably a human gene encoding an SPI.
  • 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 SPI 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
  • 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 an SPI 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 SPI antisense nucleic acids can be used to treat or prevent Schizophrenia when the target SPI is overexpressed in the CSF of subjects suspected of having or suffering from Schizophrenia.
  • a single-stranded DNA antisense SPI oligonucleotide is used.
  • Cell types which express or overexpress RNA encoding an SPI 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 an SPI-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 an SPI, immunoassay, etc. In a preferred aspect, primary tissue from a subject can be assayed for SPI expression prior to treatment, e.g., by immunocytochemistry or in situ hybridization.
  • leukocytes e.g., neutrophils, macrophages, monocytes
  • resident cells e.g., astrocytes, glial cells, neuro
  • compositions of the invention comprising an effective amount of an SPI antisense nucleic acid in a pharmaceutically acceptable carrier, can be administered to a subject having Schizophrenia.
  • SPI antisense nucleic acid which will be effective in the treatment of Schizophrenia can be determined by standard clinical techniques.
  • compositions comprising one or more SPI antisense nucleic acids are administered via liposomes, microparticles, or microcapsules.
  • such compositions may be used to achieve sustained release of the SPI antisense nucleic acids.
  • symptoms of Schizophrenia may be ameliorated by decreasing the level of an SPI or SPI activity by using gene sequences encoding the SPI in conjunction with well-known gene "knock-out,” ribozyme or triple helix methods to decrease gene expression of an SPI.
  • ribozyme or triple helix molecules are used to modulate the activity, expression or synthesis of the gene encoding the SPI, and thus to ameliorate the symptoms of Schizophrenia.
  • 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 an SPI 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 an SPI
  • 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'.
  • the construction and production of hammerhead ribozymes is well known in the art and is described more fully in
  • the ribozyme is engineered so that the cleavage recognition site is located near the 5' end of the mRNA encoding the SPI, 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, Science, (1984) 224:574-578; Zaug and Cech, Science, (1986) 231, 470-475; Zaug, et al, Nature, (1986) 324, 429-433; published International patent application No. WO 88/04300 by University Patents Inc.; Been and Cech, Cell, (1986) 47:207-216).
  • 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, Science, (1984) 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 SPI.
  • the ribozymes can be composed of modified oligonucleotides (e.g., for improved stability, targeting, etc.) and should be delivered to cells that express the SPI in vivo.
  • a preferred method of delivery involves using a DNA construct
  • ribozyme under the control 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 SPI and inhibit translation. Because ribozymes, unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficacy.
  • Endogenous SPI expression can also be reduced by inactivating or "knocking out” the gene encoding the SPI, or the promoter of such a gene, using targeted homologous recombination (e.g., see Smithies, et al, Nature (1985) 317:230-234; Thomas and Capecchi, Cell (1987) 51:503-512; Thompson et al, Cell (1989) 5:313-321; and Zijlstra et al, Nature (1989) 342:435-438, each of which is incorporated by reference herein in its entirety).
  • targeted homologous recombination e.g., see Smithies, et al, Nature (1985) 317:230-234; Thomas and Capecchi, Cell (1987) 51:503-512; Thompson et al, Cell (1989) 5:313-321; and Zijlstra et al, Nature (1989) 342:435-438, each of which is incorporated by reference herein in its entirety).
  • a mutant gene encoding a non-functional SPI (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 SPI) 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).
  • this approach can be adapted for use in humans provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors.
  • the endogenous expression of a gene encoding an SPI 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 SPI in target cells in the body.
  • deoxyribonucleotide sequences complementary to the regulatory region of the gene i.e., the gene promoter and/or enhancers
  • 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 stretches 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 an SPI that the situation may arise wherein the concentration of SPI 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 SPI that exhibit normal gene activity and that do not contain sequences susceptible to whatever antisense, ribozyme, or triple helix treatments are being utilized.
  • normal SPI can.be co- administered in order to maintain the requisite level of SPI activity.
  • 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 oligodeoxyribonucleotides and oligoribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis.
  • RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
  • 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 Schizophrenia.
  • Test compounds can be assayed for their ability to restore SF or SPI levels in a subject having Schizophrenia towards levels found in subjects free from Schizophrenia or to produce similar changes in experimental animal models of Schizophrenia.
  • Compounds able to restore SF or SPI levels in a subject having Schizophrenia towards levels found in subjects free from Schizophrenia or to produce similar changes in experimental animal models of Schizophrenia can be used as lead compounds for further drug discovery, or used therapeutically.
  • SF and SPI expression can' be assayed by the Preferred Technology, immunoassays, gel electrophoresis followed by visualization, detection of SPI 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 an SF or SPI 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.
  • mice 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.
  • any animal model system known in the art may be used
  • animal models of Schizophrenia include, but are not limited to, Phencyclidine treated rodents (Sams- Dodd Rev Neurosci (1999) 10:59-90), an animal model of deficient sensorimotor gating (Swerdlow and Geyer Schizophr Bull (1998) 24(2):285-301), neonatal insult to the hippocampal region (Beauregard and Bachevalier Can J Psychiatry (1996) Sep 41(7):446-56), models based on neonatal excitotoxic hippocampal damage (Lillrank et al, Clin Neurosci (1995) 3(2):98-104), attention deficit models (Feldon et al, JPsychiatr Res 4:345-66) and NM
  • transgenic animals can be produced with "knock-out” mutations of the gene or genes encoding one or more SPIs.
  • 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 an SPI are identified in non-human animals (e.g., mice, rats, monkeys, rabbits, and guinea pigs), preferably non-human animal models for Schizophrenia, expressing the SPI.
  • 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 SPIs is determined.
  • a test compound that alters the expression of an SPI can be identified by comparing the level of the selected SPI or SPIs (or rnRNA(s) encoding the same) in an animal or group of animals treated with a test compound with the.
  • test compounds that modulate the activity of an SPI 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 Schizophrenia, expressing the SPI.
  • 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 a test compound on the activity of an SPI is determined.
  • a test compound that alters the activity of an SPI can be identified by assaying animals treated with a control compound and animals treated with the test compound.
  • the activity of the SPI can be assessed by detecting induction of a cellular second messenger of the SPI (e.g., intracellular Ca2+, diacylglycerol, IP3, etc.), detecting catalytic or enzymatic activity of the SPI or binding partner thereof, detecting the induction of a reporter gene (e.g., a regulatory element that is responsive to an SPI 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 an SPI 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 an SPI are identified in human subjects having Schizophrenia, preferably those having Schizophrenia and most preferably those having severe Schizophrenia.
  • a test compound or a control compound is administered to the human subject, and the effect of a test compound on SPI expression is determined by analyzing the expression of the SPI or the mRNA encoding the same in a biological sample (e.g., CSF, serum, plasma, or urine).
  • a test compound that alters the expression of an SPI can be identified by comparing the level of the SPI 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 treated with a test compound.
  • alterations in the expression of an SPI can be identified by comparing the level of the SPI 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 an SPI.
  • test compounds that modulate the activity of an SPI are identified in human subjects having Schizophrenia, preferably those having Schizophrenia and most preferably those with severe Schizophrenia.
  • a test compound or a control compound is administered to the human subject, and the effect of a test compound on the activity of an SPI is determined.
  • a test compound that alters the activity of an SPI 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 an SPI can be identified by comparing the activity of an SPI in a subject or group of subjects before and after the administration of a test compound.
  • the activity of the SPI 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 SPI (e.g., intracellular Ca2+, diacylglycerol, IP3, etc.), catalytic or enzymatic activity of the SPI 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 SPI e.g., intracellular Ca2+, diacylglycerol, IP3, etc.
  • catalytic or enzymatic activity of the SPI 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 an SPI towards levels detected in control subjects is selected for further testing or therapeutic use.
  • a test compound that changes the activity of an SPI 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 Schizophrenia are identified in human subjects, having Schizophrenia, preferably subjects having Schizophrenia and most preferably subjects with severe Schizophrenia.
  • 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 Schizophrenia is determined.
  • a test compound that reduces one or more symptoms can be identified by comparing the subjects treated with a control compound to the subjects treated with the test compound. Techniques known to physicians familiar with Schizophrenia can be used to determine whether a test compound reduces one or more symptoms associated with Schizophrenia.
  • a test compound that enhances memory or reduces confusion in a subject having Schizophrenia will be beneficial for treating subjects having Schizophrenia.
  • a test compound that reduces the severity of one or more symptoms associated with Schizophrenia in a human having Schizophrenia 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 administration are described below.
  • a compound of the invention e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. (1987) 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, inttaperitoneal, intravenous, subcutaneous, intranasal, 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 administration 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 liposome (see Langer, Science (1990) 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. 14:201; Buchwald et al, 1980, Surgery 88:507; Saudek et al, 1989, N. Engl. J. Med. 321:574).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol.
  • 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 intracellularly 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. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • 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.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • 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 lidocaine 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.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • 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 Schizophrenia can be determined by standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • suitable dosage ranges for intravenous administration are generally about 20- 500 micrograms of active compound per kilogram body weight.
  • suitable dosage ranges for intranasal 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.
  • 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 ultrafiltration 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: 8M urea (BDH 452043w ) 4% CHAPS (Sigma C3023) 65mM dithiotheitol (DTT)
  • 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) strips (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 current limit was set to 10mA for 12 gels, and the wattage limit to 5W.
  • the temperature was held at 20 °C throughout the run.
  • 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-1503).
  • 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 I- 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: 23 cm wide x 24cm long (back plate); 23cm wide x 24cm long with a 2cm deep notch in the central 19cm (front plate).
  • back plate 23 cm wide x 24cm long
  • front plate 23cm wide x 24cm long with a 2cm deep notch in the central 19cm
  • back plate was treated with a 0.4% solution of ⁇ -methacryl-oxypropyltrimethoxysilane in ethanol (BindSilaneTM;
  • 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 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 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 concentration of 1.2 mg/1; 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% (v/v)
  • 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, M4 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 PAGE ⁇ analysis program (Release 2.2, 1997, BioRad Laboratories, Hercules, California, Cat. # 170- 7566) to autodetect the registration points, Ml, M2, M3 and 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 CSF1 to CSF 12, 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 XV.
  • 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.
  • 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 of the remaining study gel images was individually matched to the primary master image such that common protein features were paired between the primary master image and each individual study gel image as described below.
  • 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 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.
  • 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. In that case, 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.
  • 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 centroids adjusted to the master geometry using the flow field correction.
  • MCI molecular cluster index
  • An MCI identifies a set of matched features on different images.
  • an MCI represents a protein or proteins eluting at equivalent positions in the 2D separation in different samples.
  • LIMS Laboratory Information Management System
  • a third non-overlapping selection strategy is based on qualitative presence or ' absence alone. Using this procedure, a percentage feature presence was calculated across the control samples and Schizophrenia samples for each MCI which was a potential SF based on such qualitative criteria alone, i.e. presence or absence. The MCIs which recorded a percentage feature presence of 80% or more on Schizophrenia samples and a percentage feature presence of 20% or less on control samples, were selected as the qualitative differential SFs with 80% selectivity. A second group of qualitative differential SFs with 80% selectivity were formed by those MCIs which recorded a percentage feature presence of 80% or more on control samples and a percentage feature presence of 20% or less on Schizophrenia samples.
  • SFs to be selected on the basis of: (a) statistical significance as measured by the Wilcoxon Rank-Sum test, (b) a significant fold change threshold with a chosen selectivity, or (c) qualitative differences with a chosen selectivity.
  • 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 electrospray Z-spray source.
  • MS/MS Mass spectrometry
  • MS/MS Micromass Quadrupole Time-of-Flight
  • Q-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 SPI of the invention for screening or diagnosis of Schizophrenia, determining the prognosis of a Schizophrenia patient, or monitoring the effectiveness of Schizophrenia therapy.
  • the following example also illustrates the use of modulators (e.g., agonist or antagonists) of a SPI of the invention to treat or prevent Schizophrenia.
  • Neuronal pentraxins are expressed predominantly in the nervous system and belong to the pentraxin protein family* that also includes serum amyloid P component (AP) and C-reactive protein (CRP) (Schlimgen et al, (1995) Neuron 3, 519-526; Omeis LA. et al, (1996) Genomics 36,543-545) and neuronal activity-regulated pentraxin (NARP) (Tsui et al, (1996) J Neurosci 16, 2463-2478).
  • AP serum amyloid P component
  • CRP C-reactive protein
  • NARP neuronal activity-regulated pentraxin
  • NP-1 and NP-2 have a putative role in the uptake of synaptic.
  • NP-1 cerebrospinal fluid
  • SPI-206 the neuronal pentraxin receptor (hNPR), whose gene is located on chromosome 22ql2.3-13.2 (accession ID AL008583), an ortholog of the rat neuronal pentraxin receptor (rNPR) (Kirkpatrick LL et al, (2000) J Biol Chem. 275, 17786-92; Dodds DC et al, supra).
  • SPI-206 is 87% identical at the peptide sequence level to rNPR.
  • the rat protein, a putative integral membrane pentraxin has 49 and 48% identity to neuronal pentraxin 1 and neuronal pentraxin 2, respectively (Dodds DC et al, supra).
  • rNPR is expressed on the cell membrane and can form heteropentamers with NPl and NP2 that can be released from cell membranes (Kirkpatrick et al, supra).
  • hNPR (SPI-206) has 48% homology with serum amyloid P, a protein that has been described for its role in amyloid deposition, which is delayed in mice with targeted deletion of the serum amyloid P component gene (Botto et al, (1997) Nat Med. 3(8), 855-9).
  • the expression of hNPR (SF-342, SPI-206) has been shown herein to be significantly increased in the cerebrospinal fluid (CSF) of subjects having Schizophrenia as compared with the CSF of subjects free from Schizophrenia (see Table II).
  • CSF cerebrospinal fluid
  • Table II quantitative detection of hNPR in CSF can be used to diagnose Schizophrenia, determine the progression of Schizophrenia or monitor the effectiveness of a therapy for Schizophrenia.
  • compounds that modulate e.g., upregulate or downregulate
  • the expression, activity or both the expression and activity of NP-1 or hNPR are administered to a subject in need of treatment or for prophylaxis of Schizophrenia.
  • Antibodies that modulate the expression, activity or both the expression and activity of NP-1 or hNPR are suitable for this purpose.
  • nucleic acids coding for all or a portion of NP-1 or hNPR, or nucleic acids complementary to all or a portion of NP-1 or hNPR may be administered.
  • NP-1 or hNPR, or fragments of the NP-1 or hNPR polypeptide may also be administered.
  • the invention also provides screening assays to identify additional compounds that modulate the expression of NP-1 or activity of NP-1.
  • Compounds that modulate the expression of NP-1 in vitro can be identified by comparing the expression of NP-1 in cells treated with a test compound to the expression of NP-1 in cells treated with a control compound (e.g., saline).
  • Methods for detecting expression of NP-1 are known in the art and include measuring the level of NP-1 RNA (e.g., by northern blot analysis or RT-PCR) and measuring NP-1 protein (e.g., by immunoassay or western blot analysis).
  • Compounds that modulate the activity of NP-1 can be identified by comparing the ability of a test compound to agonize or antagonize a function of NP-1, such as its affects on synaptogenesis or plasticity or its binding to hNPR, to the ability of a control compound (e.g., saline) to inhibit the same function of NP-1.
  • a control compound e.g., saline
  • Compounds capable of modulating NP-1 binding to its receptor or NP-1 activity are identified as compounds suitable for further development as a compound useful for the treatment of Schizophrenia.
  • Binding between NP-1 and its receptor can be determined by, for example, contacting NP-1 with cells known to express the hNPR and assaying the extent of binding between NP-1 and the hNPR cell surface receptor, or by contacting NP-1 with hNPR in a cell-free assay, i.e., an assay where the NP-1 and hNPR are isolated, and, preferably, recombinantly produced, and assaying the extent of binding between NP-1 and hNPR.
  • candidate compounds may be tested for their ability to agonize or antagonize the binding of NP-1 to hNPR.
  • the primers used for PCR were taken from the 3' untranslated region of Genbank entry AL 162057, and were as follows: sense, 5' acacccaaacatcttggcatcc 3', antisense, 5' tcagggagtggagatagggaac 3'. Reactions containing lOng cDNA, SYBR green sequence detection reagents (PE Biosystems) and sense and antisense primers were assayed on an ABI7700 sequence detection system (PE Biosystems). The PCR conditions were 1 cycle at 50°C for 2 min, 1 cycle at 95°C for 10 min, and 40 cycles of 95°C for 15s, 55°C for lmin.
  • PCR product was measured in real time as the increase in SYBR green fluorescence, and the data were analyzed using the Sequence Detector program vl.6.3 (PE Biosystems). Standard curves relating initial template copy number to fluorescence and amplification cycle were generated using the amplified PCR product as a template, and were used to calculate SPI-206 copy number in each sample.
  • Figure 3 clearly shows the brain specificity of SPI-206 (up to 2300 copy number/ng cDNA), with low systemic levels (maximum 91 copy number/ng cDNA in fetal lung tissue).
  • SPI 238/240 mRNA The distribution of SPI 238/240 mRNA was restricted in the body and elevated in all parts of the brain.
  • Figure 5 clearly shows the brain specificity of SPI 238/240 (up to 4100 copy number/ng cDNA), with low systemic levels (maximum 83 copy number/ng cDNA in liver tissue).

Abstract

La présente invention concerne des méthodes et des compositions destinées au criblage, au diagnostic et aux prévisions en matière de schizophrénie, ainsi qu'au suivi de l'efficacité d'un traitement de la schizophrénie, à l'identification de patients susceptibles de réagir à un traitement thérapeutique particulier et au développement de médicaments. L'invention concerne également les caractéristiques associées à la schizophrénie (SF), pouvant être détectées par électrophorèse bidimensionnelle du liquide céphalo-rachidien, du sérum ou du plasma. L'invention concerne en outre des isoformes de protéines associées à la schizophrénie (SPI) que l'on peut détecter dans le liquide céphalo-rachidien, le sérum ou le plasma, des préparations contenant ces isoformes isolées, des anticorps immunospécifiques aux SPI et des kits les comprenant.
PCT/GB2001/000783 2000-02-24 2001-02-23 Proteines et genes et leur utilisation dans le diagnostic et le traitement de la schizophrenie WO2001063293A2 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP01905991A EP1259818A2 (fr) 2000-02-24 2001-02-23 Diagnostic et traitement de la schizophrenie
AU2001233954A AU2001233954A1 (en) 2000-02-24 2001-02-23 Proteins, genes and their use for diagnosis and treatment of schizophrenia

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB0004415A GB0004415D0 (en) 2000-02-24 2000-02-24 Proteins and their use for diagnosis and treatment of schizophrenia
GB0004415.6 2000-02-24
US75039500A 2000-12-28 2000-12-28
US09/750,395 2000-12-28

Publications (3)

Publication Number Publication Date
WO2001063293A2 true WO2001063293A2 (fr) 2001-08-30
WO2001063293A8 WO2001063293A8 (fr) 2001-10-18
WO2001063293A3 WO2001063293A3 (fr) 2002-04-11

Family

ID=26243725

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/GB2001/000792 WO2001062785A2 (fr) 2000-02-24 2001-02-23 Proteines, genes et leur utilisation dans le diagnostic et le traitement de la schizophrenie
PCT/GB2001/000783 WO2001063293A2 (fr) 2000-02-24 2001-02-23 Proteines et genes et leur utilisation dans le diagnostic et le traitement de la schizophrenie

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/GB2001/000792 WO2001062785A2 (fr) 2000-02-24 2001-02-23 Proteines, genes et leur utilisation dans le diagnostic et le traitement de la schizophrenie

Country Status (3)

Country Link
EP (2) EP1259607A2 (fr)
AU (2) AU2001233954A1 (fr)
WO (2) WO2001062785A2 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1300588C (zh) * 2005-02-28 2007-02-14 上海交通大学 用于精神分裂症诊断的血浆特异性蛋白获取方法及应用
US7202211B2 (en) 2002-02-21 2007-04-10 Astellas Pharma Inc. Methods of preventing or treating brain ischemia or brain injury
WO2008090319A2 (fr) * 2007-01-22 2008-07-31 Psynova Neurotech Limited Procédés et biomarqueurs permettant de diagnostiquer et de surveiller des troubles psychotiques
US7736654B2 (en) 2001-04-10 2010-06-15 Agensys, Inc. Nucleic acids and corresponding proteins useful in the detection and treatment of various cancers

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003028543A2 (fr) * 2001-10-03 2003-04-10 Pfizer Products Inc. Molecules d'acide nucleique, polypeptides et utilisations associees comprenant le diagnostic et le traitement de la maladie d'alzheimer
US7052849B2 (en) 2001-11-23 2006-05-30 Syn X Pharma, Inc. Protein biopolymer markers predictive of insulin resistance
US20030100012A1 (en) * 2001-11-23 2003-05-29 George Jackowski Plasma protease C1 biopolymer markers predictive of alzheimers disease
CA2748449A1 (fr) 2008-12-27 2010-07-15 Pawan Saharan Nanopeptides derives d'un colostrum mammalien pour infections virales et recurrentes a large spectre avec leur procede d'isolement

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995033480A1 (fr) * 1994-06-07 1995-12-14 The Government Of The United States Of America, Re Facteur derive de l'epithelium pigmentaire: caracterisation, organisation genomique et sequence de son gene
WO1998011222A1 (fr) * 1996-09-13 1998-03-19 The Mount Sinai School Of Medicine Of The City University Of New York Methodes de selection de composes utilisables pour traiter la schizophrenie
WO1998040748A1 (fr) * 1997-03-14 1998-09-17 Neuromark Diagnostic de troubles neurologiques
WO1998042723A1 (fr) * 1997-03-27 1998-10-01 Millennium Pharmaceuticals, Inc. Procedes et compositions s'appliquant au diagnostique et au traitement des troubles neuropsychiatriques
WO1999011796A1 (fr) * 1997-09-05 1999-03-11 Acorda Therapeutics Sulfotransferase hnk-1 et ses methodes d'utilisation
WO1999014328A2 (fr) * 1997-09-17 1999-03-25 Genentech, Inc. Polypeptides secretes et transmembranaires et acides nucleiques les codant
WO2000023784A2 (fr) * 1998-10-21 2000-04-27 Chen, Hong Methode de detection du risque de diabete de type ii sur la base de mutations trouvees dans une carboxypeptidase e
WO2000055173A1 (fr) * 1999-03-12 2000-09-21 Human Genome Sciences, Inc. Sequences et polypeptides geniques associes au cancer des ovaires et du sein
WO2001025268A1 (fr) * 1999-10-04 2001-04-12 Schrotz King Petra Proteines humaines associees a l'attaque
EP1101820A1 (fr) * 1999-11-15 2001-05-23 MERCK PATENT GmbH Pentraxin I et recepteur de la pentraxin, inhibiteurs de ces proteines et compositions pharmaceutiques comprenant ces composés

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5767252A (en) * 1996-04-08 1998-06-16 The Johns Hopkins University School Of Medicine Neuronal cell growth factor, Narp

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995033480A1 (fr) * 1994-06-07 1995-12-14 The Government Of The United States Of America, Re Facteur derive de l'epithelium pigmentaire: caracterisation, organisation genomique et sequence de son gene
WO1998011222A1 (fr) * 1996-09-13 1998-03-19 The Mount Sinai School Of Medicine Of The City University Of New York Methodes de selection de composes utilisables pour traiter la schizophrenie
WO1998040748A1 (fr) * 1997-03-14 1998-09-17 Neuromark Diagnostic de troubles neurologiques
WO1998042723A1 (fr) * 1997-03-27 1998-10-01 Millennium Pharmaceuticals, Inc. Procedes et compositions s'appliquant au diagnostique et au traitement des troubles neuropsychiatriques
WO1999011796A1 (fr) * 1997-09-05 1999-03-11 Acorda Therapeutics Sulfotransferase hnk-1 et ses methodes d'utilisation
WO1999014328A2 (fr) * 1997-09-17 1999-03-25 Genentech, Inc. Polypeptides secretes et transmembranaires et acides nucleiques les codant
WO2000023784A2 (fr) * 1998-10-21 2000-04-27 Chen, Hong Methode de detection du risque de diabete de type ii sur la base de mutations trouvees dans une carboxypeptidase e
WO2000055173A1 (fr) * 1999-03-12 2000-09-21 Human Genome Sciences, Inc. Sequences et polypeptides geniques associes au cancer des ovaires et du sein
WO2001025268A1 (fr) * 1999-10-04 2001-04-12 Schrotz King Petra Proteines humaines associees a l'attaque
EP1101820A1 (fr) * 1999-11-15 2001-05-23 MERCK PATENT GmbH Pentraxin I et recepteur de la pentraxin, inhibiteurs de ces proteines et compositions pharmaceutiques comprenant ces composés

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
DISCIPIO, RICHARD G. ET AL: "The structure of human complement component C7 and the C5b-7 complex" J. BIOL. CHEM. (1988), 263(1), 549-60, XP001009867 *
FURLEY A J ET AL: "THE AXONAL GLYCOPROTEIN TAG-1 IS AN IMMUNOGLOBULIN SUPERFAMILY MEMBER WITH NEURITE OUTGROWTH-PROMOTING ACTIVITY" CELL, CELL PRESS, CAMBRIDGE, NA, US, vol. 61, no. 1, 6 April 1990 (1990-04-06), pages 157-170, XP001016334 ISSN: 0092-8674 *
GEBHARD W ET AL: "CDNA CLONING AND COMPLETE PRIMARY STRUCTURE OF THE SMALL, ACTIVE SUBUNIT OF HUMAN CARBOXYPEPTIDASE N (KININASE 1)" EUROPEAN JOURNAL OF BIOCHEMISTRY, BERLIN, DE, vol. 178, no. 3, January 1989 (1989-01), pages 603-607, XP001025505 ISSN: 0014-2956 *
HASLER T H ET AL: "CDNA CLONING, STRUCTURAL FEATURES, AND EUCARYOTIC EXPRESSION OF HUMAN TAG-1/AXONIN-1" EUROPEAN JOURNAL OF BIOCHEMISTRY, BERLIN, DE, vol. 211, no. 1/2, January 1993 (1993-01), pages 329-339, XP001025504 ISSN: 0014-2956 *
KOZAKI K ET AL: "ISOLATION, PURIFICATION AND CHARACTERIZATION OF A COLLAGEN-ASSOCIATED SERPIN, CASPIN, PRODUCED BY MURINE COLON ADENOCARCINOMA CELLS" JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, MD, US, vol. 273, no. 24, 1998, pages 15125-15130, XP001023973 ISSN: 0021-9258 *
KRUPNIK V E ET AL: "Functional and structural diversity of the human Dickkopf gene family" GENE, ELSEVIER BIOMEDICAL PRESS. AMSTERDAM, NL, vol. 238, no. 2, 1 October 1999 (1999-10-01), pages 301-313, XP002140768 ISSN: 0378-1119 *
OMEIS I A ET AL: "MOUSE AND HUMAN NEURONAL PENTRAXIN 1 (NPTX1): CONSERVATION, GENOMICSTRUCTURE, AND CHROMOSOMAL LOCALIZATION" GENOMICS, ACADEMIC PRESS, SAN DIEGO, US, vol. 36, no. 3, 1996, pages 543-545, XP000909636 ISSN: 0888-7543 *
SCHLIMGEN A K ET AL: "NEURONAL PENTRAXIN, A SECRETED PROTEIN WITH HOMOLOGY TO ACUTE PHASEPROTEINS OF THE IMMUNE SYSTEM" NEURON, CAMBRIDGE, MA, US, vol. 14, 1995, pages 519-526, XP000909700 *
TSIOTRA P ET AL: "ISOLATION OF THE CDNA AND CHROMOSOMAL LOCALIZATION OF THE GENE (TAX1) ENCODING THE HUMAN AXONAL GLYCOPROTEIN TAG-1" GENOMICS, ACADEMIC PRESS, SAN DIEGO, US, vol. 18, no. 3, 1994, pages 562-567, XP001025514 ISSN: 0888-7543 *
TSUJI, T. ET AL.: "A REIC Gene Shows Down-Regulation in Human Immortalized Cells and Human Tumor-Derived Cell Lines" BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 268, 5 February 2000 (2000-02-05), pages 20-24, XP001016362 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7736654B2 (en) 2001-04-10 2010-06-15 Agensys, Inc. Nucleic acids and corresponding proteins useful in the detection and treatment of various cancers
US7202211B2 (en) 2002-02-21 2007-04-10 Astellas Pharma Inc. Methods of preventing or treating brain ischemia or brain injury
CN1300588C (zh) * 2005-02-28 2007-02-14 上海交通大学 用于精神分裂症诊断的血浆特异性蛋白获取方法及应用
WO2008090319A2 (fr) * 2007-01-22 2008-07-31 Psynova Neurotech Limited Procédés et biomarqueurs permettant de diagnostiquer et de surveiller des troubles psychotiques
WO2008090319A3 (fr) * 2007-01-22 2008-12-31 Psynova Neurotech Ltd Procédés et biomarqueurs permettant de diagnostiquer et de surveiller des troubles psychotiques
JP2010517007A (ja) * 2007-01-22 2010-05-20 サイノヴァ ニューロテック リミテッド 精神病性障害を診断およびモニターするための方法およびバイオマーカー
US7981684B2 (en) 2007-01-22 2011-07-19 Psynova Neurotech Limited Methods and biomarkers for diagnosing and monitoring psychotic disorders

Also Published As

Publication number Publication date
EP1259818A2 (fr) 2002-11-27
WO2001063293A3 (fr) 2002-04-11
AU2001233954A1 (en) 2001-09-03
WO2001062785A3 (fr) 2002-01-31
EP1259607A2 (fr) 2002-11-27
AU2001237543A1 (en) 2001-09-03
WO2001063293A8 (fr) 2001-10-18
WO2001062785A2 (fr) 2001-08-30

Similar Documents

Publication Publication Date Title
US20040110938A1 (en) Proteins, genes and their use for diagnosis and treatment of schizophrenia
US20070015217A1 (en) Nucleic acid molecules, polypeptides and uses therefor, including diagnosis and treatment of Alzheimer's disease
US20040053309A1 (en) Proteins, genes and their use for diagnosis and treatment of kidney response
EP1354199A2 (fr) Diagnostic et traitement de la sclerose en plaques
EP1408333A2 (fr) Diagnostic et traitement de la maladie d'Alzheimer
JP2009506301A (ja) Pifファミリーの新たなタンパク質アイソフォーム及びその使用
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
EP1379879A2 (fr) Diagnostic et traitement de la maladie d'alzheimer
US20020142303A1 (en) Proteins, genes and their use for diagnosis and treatment of Schizophrenia
EP1259819A2 (fr) Diagnostic de maladies affectives bipolaires (bad) et de depressions unipolaires
EP1259818A2 (fr) Diagnostic et traitement de la schizophrenie
US20030032773A1 (en) Proteins, genes and their use for diagnosis and treatment of bipolar affective disorder (BAD) and unipolar depression
EP1264183A2 (fr) Le diagnostic et le traitement de la demence vasculaire
US20040203022A1 (en) Proteins and genes for diagnosis and treatment of ErbB2-related cancer
US20040022794A1 (en) Nucleic acid molecules, polypeptides and uses therefor, including diagnosis and treatment of Alzheimer's disease
WO2002054080A2 (fr) Proteines, genes et leur utilisation pour le diagnostic et le traitement de la reponse vasculaire
US20040043425A1 (en) Proteins, genes and their use for diagnosis and treatment of chronic asthma
US20030092197A1 (en) Proteins, genes and their use for diagnosis and treatment of cardiac response
US20030077603A1 (en) Methods and compositions for diagnosis of hepatoma
WO2002054079A2 (fr) Proteines, genes et leur utilisation pour le diagnostic et le traitement de la reponse cardiaque

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

AK Designated states

Kind code of ref document: C1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: C1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: PAT. BUL. 35/2001 UNDER (30) REPLACE "28.02.2000" BY "28.12.2000"

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2001905991

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001905991

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2001905991

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: JP