Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHOD AND BIOMARKERS FOR IN VITRO DIAGNOSIS OF MENTAL DISORDERS
Document Type and Number:
WIPO Patent Application WO/2018/096141
Kind Code:
A1
Abstract:
The invention relates to a method for in vitro diagnosis of the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder, wherein the mental disorder is associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, the method comprising: a) measuring in a sample of a body tissue or fluid from the individual the expression levels of at least two marker genes, each of which coding for at least one marker protein; b) comparing the measured expression levels to predetermined threshold values representing the expression levels of said marker genes in a healthy population; and c) based on the comparison, determining whether the individual has the mental disorder or a predisposition to the mental disorder, wherein the measured expression levels are indicative to the mental disorder or disposition if the measured expression levels of said marker genes exceed, reach or fall below the predetermined threshold value. For example, the expression level (relative mRNA expression) of each of the marker genes Ifng, Ccl4, Il13ra1, Il12rb2, C3, and Slc27a2 is significantly decreased in a transgenic rat (TG, gray) in relation to non-transgenic littermates (LM, white). That is, the lower expression level of each tested marker gene in the transgenic rat relative to the expression level of the respective marker gene in the non-transgenic control is indicative of dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis.

Inventors:
KORTH CARSTEN (DE)
TROSSBACH SVENJA (DE)
HECHER LAURA (DE)
Application Number:
PCT/EP2017/080504
Publication Date:
May 31, 2018
Filing Date:
November 27, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
KORTH CARSTEN (DE)
International Classes:
C12Q1/68
Domestic Patent References:
WO2003013551A12003-02-20
WO2010097631A12010-09-02
WO2008048639A22008-04-24
WO2009102748A22009-08-20
WO2013186562A12013-12-19
WO2010097631A12010-09-02
Foreign References:
US20110136738A12011-06-09
Other References:
HETTEMA JOHN M: "Genetic association between RGS1 and internalizing disorders", PSYCHIATRIC GENETICS, RAPID COMMUNICATIONS OF OXFORD LTD, XX, vol. 23, no. 2, 1 April 2013 (2013-04-01), pages 56 - 60, XP009193991, ISSN: 0955-8829, DOI: 10.1097/YPG.0B013E32835D7048
S V TROSSBACH ET AL: "Misassembly of full-length Disrupted-in-Schizophrenia 1 protein is linked to altered dopamine homeostasis and behavioral deficits", MOLECULAR PSYCHIATRY, vol. 21, no. 11, 12 January 2016 (2016-01-12), GB, pages 1561 - 1572, XP055441893, ISSN: 1359-4184, DOI: 10.1038/mp.2015.194
BOLSTAD ET AL.: "A comparison of normalization methods for high density oligonucleotide array data based on variance and bias", BIOINFORMATICS, vol. 19, no. 2, 2003, pages 185 - 193, XP008041261, DOI: doi:10.1093/bioinformatics/19.2.185
CHAN ET AL.: "Development of a blood-based molecular biomarker test for identification of schizophrenia before disease onset", TRANSLATIONAL PSYCHIATRY, vol. 5, 2015, pages e601
DEAN: "Dissecting the Syndrome of Schizophrenia: Progress toward Clinically Useful Biomarkers", SCHIZOPHRENIA RESEARCH AND TREATMENT, vol. 2011, 2011
HAMBURG ET AL.: "Simultaneous effects on parvalbumin-interneuron and dopaminergic system development in a transgenic rat model for sporadic schizophrenia", SCIENTIFIC REPORTS, vol. 6, 2016, pages 34946, XP055361170, DOI: doi:10.1038/srep34946
HENNAH; PORTEOUS: "The DISC1 pathway modulates expression of neurodevelopmental, synaptogenic and sensory perception genes", PLOS ONE, vol. 4, no. 3, 2009, pages e4906
HOWES; KAPUR: "The dopamine hypothesis of schizophrenia: version III--the final common pathway", SCHIZOPHR BULL., vol. 35, no. 3, May 2009 (2009-05-01), pages 549 - 62, XP055361164, DOI: doi:10.1093/schbul/sbp006
KORTH: "DISCopathies: brain disorders related to DISC1 dysfunction", REV NEUROSCI, vol. 20, no. 5-6, 2009, pages 321 - 30, XP055003321, DOI: doi:10.1515/REVNEURO.2009.20.5-6.321
KORTH: "Aggregated proteins in schizophrenia and other chronic mental diseases: DISCIopathies", PRION, vol. 6, no. 2, 2012, pages 134 - 41, XP055361166, DOI: doi:10.4161/pri.18989
KORTH: "Aggregated proteins in schizophrenia and other chronic mental diseases", PRION, vol. 6, no. 2, 2012, pages 1 - 8, XP055361166, DOI: doi:10.4161/pri.18989
SOARES ET AL.: "DISC1: Structure, Function, and Therapeutic Potential for Major Mental Illness", ACS CHEM NEUROSCI, vol. 2, no. 11, 16 November 2011 (2011-11-16), pages 609 - 632, XP055030941, DOI: doi:10.1021/cn200062k
TROSSBACH ET AL.: "Peripheral DISC1 protein levels as a trait marker for schizophrenia and modulating effects of nicotine", BEHAV BRAIN RES, vol. 275C, 2014, pages 176 - 182
TROSSBACH ET AL.: "Misassembly of full-length Disrupted-in-Schizophrenia 1 protein is linked to altered dopamine homeostasis and behavioral deficits", MOLECULAR PSYCHIATRY, 2016, pages 1 - 12
WARBRICK ET AL.: "Direction and magnitude of nicotine effects on the fMRI BOLD response are related to nicotine effects on behavioral performance", PSYCHOPHARMACOLOGY (BERL, vol. 215, no. 2, 2011, pages 333 - 344
Attorney, Agent or Firm:
REMUS, Alvaro (DE)
Download PDF:
Claims:
Claims

1 . A method for in vitro diagnosis of the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder, wherein the mental disorder is associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, the method comprising:

a) measuring in a sample of a body tissue or fluid from the individual the expression levels of at least two marker genes, each of which coding for at least one marker protein, wherein said marker genes are selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3; b) comparing the measured expression levels to predetermined threshold values representing the expression levels of said marker genes in a healthy population; and

c) based on the comparison, determining whether the individual has the mental disorder or a predisposition to the mental disorder, wherein the measured expression levels are indicative to the mental disorder or disposition if the measured expression levels of said marker genes exceed, reach or fall below the predetermined threshold value.

2. The method according to claim 1 , wherein a first marker gene is RGS1 and at least one second marker gene is selected from the group consisting of human NKG7, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2,

SLC27A2, and C3.

3. The method according to claim 2, wherein the first marker gene is RGS1 and the second marker gene is NKG7 and/or CCL4.

4. The method according to any of claims 1 to 3, wherein at least one

additional marker gene is selected from the human equivalents of the genes listed in Table 1 .

5. The method according to any of claims 1 to 4, wherein the measured expression levels are indicative to the mental disorder or disposition if each measured expression level is lower than a respective reference expression level and/or reaches or falls below the predetermined threshold value.

6. The method according to any of claims 1 to 5, wherein the expression

levels are measured by quantitative reverse transcription Polymerase Chain Reaction or a high-affinity binding assay, and/or wherein the expression levels are measured by microarray analysis.

7. A combination of at least two marker proteins derived from marker genes, or at least two nucleic acid molecules comprising marker genes coding for marker proteins, said marker genes being selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3, for use in in vitro diagnostics.

8. The combination according to claim 7, wherein a first marker gene is

RGS1 and at least one second marker gene is selected from the group consisting of human NKG7, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3.

9. The combination according claim 7 or 8, wherein at least one additional marker gene is selected from the human equivalents of the genes listed in Table 1 .

10. The combination according to any one of claims 7 to 9 used in an in vitro method of diagnosing the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder, wherein the mental disorder is associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis.

1 1 A kit for diagnosing the presence of a mental disorder in a human

individual or the predisposition of the human individual to the mental disorder in vitro, wherein the mental disorder is associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, the kit comprising:

a) a set of oligonucleotide primers which are suitable to initiate

amplification of the transcripts of at least two marker genes, each of which coding for at least one marker protein, in a Polymerase Chain Reaction and/or microarray, wherein said marker genes are selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3, and/or

at least two first antibodies or molecules, each of which specifically binding to a marker protein in a body tissue or fluid from the individual, wherein the marker proteins are derived from marker genes selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3;

b) at least two reporter probes capable of binding to complementary DNA (cDNA) derived from the transcripts, which are suitable to be detected in a quantitative reverse transcription Polymerase Chain Reaction, and/or

at least two labelled second antibodies, each of which specifically binding to one of the first antibodies or molecules, which are designed to be detected in a high-affinity binding assay; and optionally, c) at least two reference samples.

A method for determining the response to at least one pharmaceutical compound able to correct a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, wherein the expression levels of at least two marker genes are determined and compared according to steps a) and b) of the method according to claim 1 , and wherein the measured expression levels indicate that the response to the pharmaceutical compound is positive if each aberrant expression level of said marker genes is normalized or at least improved.

A nonhuman transgenic animal useful for providing organs, tissues, or cells, which is able to stably express a modified gene coding for human DISC1 protein, wherein the expression level of the modified gene is higher than that of the respective wild-type gene and thus results in the formation of aggregates of the DISC1 protein within the cells, said animal

representing a subset of human subjects having at least one mental disorder, for use in the identification and analysis of marker proteins or genes for diagnosing mental disorders in human individuals.

A method for determining the therapeutic effect of a potentially curative pharmaceutical compound on a mental disorder, or a predisposition of a human individual to the mental disorder, associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, wherein said pharmaceutical compound is administered to a transgenic animal, and wherein it is indicated that the therapeutic effect is positive if aberrant expression levels of at least two marker genes selected from the group consisting of NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3 are normalized in said transgenic animal after administration of said pharmaceutical compound.

The method according to claim 14, wherein the transgenic animal is a nonhuman transgenic animal useful for providing organs, tissues, or cells, which is able to stably express a modified gene coding for human DISC1 protein, wherein the expression level of the modified gene is higher than that of the respective wild-type gene and thus results in the formation of aggregates of the DISC1 protein within the cells, said animal representing a subset of human subjects having at least one mental disorder.

Description:
Method and biomarkers for in vitro diagnosis of mental disorders

Field of the invention

The invention relates to a method for in vitro diagnosis of the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder. The invention further relates to a marker protein, a nucleic acid molecule, or a combination of marker proteins or nucleic acid molecules for use in in vitro diagnostics, and to a kit for diagnosing the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder in vitro. The invention also concerns a nonhuman transgenic animal useful for providing organs, tissues, or cells for use in the identification and analysis of marker proteins for human individuals, as well as a method for determining the therapeutic effect of a potential pharmaceutical compound on a mental disorder, or a predisposition of a human individual to the mental disorder, using the transgenic animal.

Background of the invention

Mental disorders such as schizophrenia or depression are so far solely diagnosed by a clinical approach through an interview based on the patient's self- reported experiences, behaviour reported by relatives or friends, and a mental status exam. There is no reliable and useful objective laboratory test for mental disorders, for example, based on a biological cause. That unmated clinical diagnosis is unsatisfactory by the reason that a small part of subjectivity remains and that the whole test is addicted to an observer. Another issue is the point that the heterogeneous disease course suggests different biological causes. In one case an illness runs with one single episode and has no residuum, in another case multiple episodes with no or minimum residuum are the result, such as a case which is characterised by a residuum after a first episode falling into relapse without the return to normality or a case with progressing residuum after each episode of the disorder without a return to normality. The various ways of a mental disorder makes it harder to correctly diagnose the illness by mere clinical diagnosis only with the aim of finding the right way to treat the patients. However, patients and their affiliated persons such as the pharmaceutical companies are interested on an objective and independent method to diagnose mental disorders like schizophrenia or depression.

It is assumed that there are several biological causes for mental disorders such as schizophrenia or depression, which, however, all converge in a common behavioural pathway which then can give the illusion of a homogeneity of an underlying biology. For example, typical biological symptoms of a schizophrenia patient are the increased striatal dopamine levels. Another possible symptom are neuroanatomical abnormalities such as enlarged ventricles, aberrant interneuron positioning, or biochemical abnormalities such as abnormal proteostasis of some key proteins. Based on this knowledge an objective method to diagnose a mental disorder like schizophrenia should be discovered.

For example, disrupted in schizophrenia 1 (DISCI ) is a protein that has been shown to participate in the regulation of cell proliferation, differentiation, migration, neuronal axon and dendrite outgrowth, mitochondrial transport, fission and/or fusion, and neurotransmitter functions at the synapse. Several studies have shown that unregulated expression may predispose animals to behavioural abnormalities or human individuals to the development of schizophrenia, clinical depression, bipolar disorder, and other psychiatric conditions. It has been hypothesized that unbalanced proteostasis in neurons may lead to DISC1 protein aggregates in a subset of patients with

schizophrenia or other chronic mental disorders, and several findings justify classification of DISC1 -dependent brain disorders as protein conformational disorders which we have tentatively termed DISCI opathies (Korth, 2012). That is, disturbed proteostasis and protein aggregation can be considered as a mechanism of mental disorders. Prior art

Dean (201 1 ) outlines the evolving notion of biomarkers for diagnosing mental disorders, especially schizophrenia, and discloses outcomes from a variety of biomarkers discovery strategies. In particular, the impact of high-throughput screening technologies on biomarker discovery is highlighted and how new or improved technologies may allow the discovery of either diagnostic biomarkers for schizophrenia or biomarkers that will be useful in determining appropriate treatments for people with the disorder. It is suggested that biomarkers can be identified and that these biomarkers will be useful in diagnosing and treating people with schizophrenia. However, although Dean suggests some proteins as potential biomarkers for schizophrenia and bipolar disorders, the author admits that those biomarkers still have to be validated and the development of clinically useful markers may still take a long time.

Chan et al. (2015) describe the development of a serum biomarker test for the identification of individuals at risk of developing schizophrenia based on multiplex immunoassay profiling analysis. A meta-analysis of independent cohorts of first-onset drug-naive schizophrenia patients and controls was conducted. Using least absolute shrinkage and selection operator regression, an optimal panel of biomarkers that best discriminated patients and controls was identified. This biomarker panel was verified using two independent validation cohorts and its predictive performance for identifying patients before onset of psychosis was tested using two cohorts of pre-onset or at-risk individuals. These findings are alleged to represent the first successful step towards a test that could address the clinical need for early intervention in psychiatry. However, this study does not consider underlying biological heterogeneity of schizophrenia as the identification of biomarkers starts in patient cohorts defined by clinical diagnosis.

WO 2013/186562 A1 discloses a biomarker set for diagnosing disorders like depression, anxiety disorder or other psychotic disorders consisting of eleven markers. It is suggested to use one or more analytes selected from Interleukin 10 (IL-10), Interleukin 18 (IL-18), Interleukin 2 (IL-2), Interleukin 8 (IL-8), Monocyte Chemotactic Protein 1 (MCP- 1 ), Macrophage Inflammatory Protein 1 alpha (MIP-la), Macrophage Inflammatory Protein 1 beta (ΜΙΡ-Ιβ), Matrix Metalloproteinase 2 (MMP-2), Tumor Necrosis Factor beta (TNF-β), Interleukin 4 (IL-4), and Interferon gamma (IFN-γ) as a biomarker for such diseases, or predisposition thereto. WO 2013/186562 A1 further discloses a method of diagnosing depression, anxiety disorder or other psychotic disorders in an individual, wherein the amounts of said analyte biomarkers in a biological sample obtained from the individual are quantified and then compared with the amounts present in a normal control sample from a normal subject, such that a difference in the level of the analyte biomarkers in the biological sample is indicative of the disorder, or predisposition thereto. The data provided show that the analytes may be statistically significant biomarkers for the diagnosis of depression and anxiety disorder. In particular, innate immune responsiveness is increased in persons with depressive and anxiety disorders, indicating a possible genetic vulnerability for depression or anxiety.

From WO 2010/097631 A1 it is known to use IL-17, IgA, Cortisol (CORT), Apolipoprotein Al, IL-6, Complement 3 (C3), Factor VII, Serum Amyloid P (SAP or APCS), Beta 2 Microglobulin, ICAM-I, IL-I beta, TNF alpha, MIF,

Angiotensinogen, NrCAM (Neuronal cell adhesion molecule), CD40, Cancer Antigen 125 (CA125), HCC 4 (CCL6; SCYA6), Eotaxin 3 (CCL26 or SCYA26), VEGF, Haptoglobin (HP), IL-I alpha, Apolipoprotein H (Beta-2 Glycoprotein) and TIMP 1 as a specific panel of analyte biomarkers for major depressive disorder, or predisposition thereto. This panel of biomarkers can be used in a method of diagnosing or monitoring major depressive disorder, or predisposition thereto, wherein said analyte biomarkers are detected and/or quantified in a sample from a test subject. The levels of these analyte biomarkers are found to be increased in patients with major depressive disorder.

US 201 1 /0136738 A1 discloses a method for identifying gene targets which are associated with schizophrenia or schizophrenia-like symptoms. Animal models of schizophrenia were utilized, and initiated, and from tissue obtained from at least one of those animals, transcriptional regulation was assessed over time, relative to the onset of the schizophrenia model. It is suggested to measure gene expression from animals at time points after, and, optionally, before the initiation of the model and to compare the gene expression, whether from before or after the initiation of the model, or both, to gene expression at one or more time points from control animals, which are not subject to a schizophrenia model. Transcripts can then be detected which are dysregulated in tissue from animals that are a model of schizophrenia. Any change in gene expression observed in the schizophrenia model, whether relative to other time points in the same model, relative to another schizophrenia model, or relative to the same time point or time points in control animals can be informative with respect to gene targets for schizophrenia or the symptoms of schizophrenia. However, although several genes have been identified, the dysregulation of which seems to be indicative of the presence of schizophrenia or the symptoms thereof, this document does neither provide any clinically suitable biomarkers nor any validated test for diagnosing schizophrenia.

Accordingly, there is a need to identify biological causes involved in mental disorders such as schizophrenia, as well as for methods that can detect such causes so they can be utilized in screening therapeutics, in diagnosing mental disorders, and in developing treatments for individuals with mental disorders. These biological causes may be manifold and consist in genes, protein pathology, or others. There is also a need for new biomarkers and methods for diagnosing mental disorders or detecting susceptibility to mental disorders, and for preventing or following up development of such disorders.

Moreover, patients and their relatives want an ..objective" diagnosis rather than an oral verdict and pharmaceutical companies want an ..objective" test to base 100-million EU clinical trials thereon, rather than a clinical diagnosis. However, although some single biomarkers or rather large biomarker sets for diagnosing mental disorders are already known, there is still no reliable and precise test which would be suitable to replace or even support the common clinical approach. In contrast, the current issue with the known biomarker tests is that they involved only a few and unspecific or too many biomarkers so that they get either inaccurate or too extensive and thus are not reliable and deliver aberrant diagnoses. Yet another problem in current biomarker identification is that this identification starts in patient cohorts defined by clinical diagnosis and not considering underlying biological heterogeneity. As a consequence of this procedure, any possible specific effects in patient subsets are diluted out.

Summary of the invention

It is the objective of the invention to provide a method for in vitro diagnosis of the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder, as well as at least one marker protein, nucleic acid molecule, or combination of marker proteins or nucleic acid molecules for use in such method, which ensure an accurate and reliable diagnosis of mental disorders.

This objective is met by a method for in vitro diagnosis of the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder, wherein the mental disorder is associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, and wherein said method comprises:

a) measuring in a sample of a body tissue or fluid from the individual the

expression levels of at least two marker genes, each of which coding for at least one marker protein, wherein said marker genes are selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2,

IL13RA1, KMO, FPR2, SLC27A2, and C3;

b) comparing the measured expression levels to predetermined threshold values representing the expression levels of said marker genes in a healthy population; and

c) based on the comparison, determining whether the individual has the

mental disorder or a predisposition to the mental disorder, wherein the measured expression levels are indicative to the mental disorder or disposition if the measured expression levels of said marker genes exceed, reach or fall below the predetermined threshold value.

In the method according to the invention the measured expression levels of at least two marker genes are combined in order to determine whether the individual has the mental disorder or a predisposition to the mental disorder in step c). Combining two or more markers significantly increases specificity of the method according to the invention. In some cases, sensitivity of the method may be decreased at the same time. However, in the method according to the invention specificity is more important than sensitivity since the method is provided for a sub-group of patients only and thus low sensitivity relating to the all-comprising clinical diagnosis is expected and can therefore be neglected.

The threshold values are predetermined by empirically determining a reference (standard) expression level for each marker gene, i.e. the average expression level of the marker gene in a healthy population. The measured expression levels of the marker genes in the body tissue or fluid sample of the individual (patient) are each compared to the respective predetermined threshold value. If the measured expression levels of said marker genes exceed, reach or fall below the predetermined threshold values (the direction of the change depends on whether the respective aberrant expression level is higher or lower than the respective normal expression level), it is indicated that the individual has the mental disorder or at least a predisposition thereto. That is, altered expression levels of the marker genes in the body tissue or fluid sample relative to the expression levels of the same marker genes in the normal control (reference or standard expression levels of a healthy population) are indicative of the presence of the mental disorder, or predisposition thereto.

According to the invention the method is focused on a subset of so far purely clinically defined mental disorders that are associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis. That is, the method according to the invention is useful for a subset of patients that are afflicted with mental disorders caused by disturbed homeostasis of DISC1 protein / pathway or dopamine homeostasis. For example, unregulated expression, degradation or altered protein structure of DISC1 may predispose individuals to the development of schizophrenia, recurrent depression, bipolar disorder, and other psychiatric conditions. So-called DISCIopathies seem to be caused by unbalanced proteostasis in neurons leading to DISC1 protein aggregates so that DISC1 -dependent brain disorders may be classified as protein conformational disorders. Accordingly, protein aggregation can be deemed as a biological phenotype for sporadic chronical mental disorders for a subgroup of patients, for example schizophrenia or depression patients.

Sporadic disorders are understood to represent disease entities where no clear and unambiguous genetic cause can be identified.

The extended DISC1 pathway includes many other genes, also involved in either mental illness or neurodegenerative disease (Figure 1 ; Hennah &

Porteous 2009; Soares et al. 201 1 ; Korth 2009 und 2012). The marker genes listed in Table 1 (and their human equivalents), in particular NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2 and C3, belong to a network of genes directly or indirectly regulated by DISC1 , DISC1 -associated proteins, or the DISC1 pathway and are therefore potentially useful as

biomarkers for diagnosing mental disorders that are associated with a

dysfunctional DISC1 protein pathway.

Dopamine also plays a critical role in the genesis of psychosis, the acute symptom of schizophrenia. It is hypothesized that a framework exists which links risk factors, including pregnancy and obstetric complications, stress and trauma, drug use, and genes, to increased presynaptic striatal dopaminergic function. This hypothesis explains how a complex array of pathological, positron emission tomography, magnetic resonance imaging, and other findings, such as frontotemporal structural and functional abnormalities and cognitive

impairments, may converge neurochemically to cause psychosis through aberrant salience and lead to a diagnosis of schizophrenia (Howes & Kapur 2009). Moreover, on a molecular mechanistic level, DISC1 missassembly seems to modulate dopamine homeostasis. DISCI opathies thus define a biology-based category of human mental illnesses with involvement of the dopamine system that can be modeled in animals, i.e. in a transgenic animal (e.g., tgDISCI rat). Accordingly, marker proteins involved in dopamine homeostasis may also be potentially useful as biomarkers for diagnosing mental disorders that are associated with a dysfunctional DISC1 protein pathway. Known tests for diagnosing schizophrenia start biomarker discovery with the clinical diagnosis of the disease, which is doomed to fail because of underlying clinical heterogeneity that dilutes any possible significant biomarkers for subsets of clinically defined mental disorders, and known array tests generated by proteomics use surrogate markers rather than markers based on biological causes, which results in low plausibility. The method according to the invention is based on a biological definition (e.g.,„DISC1 opathy") and a biological (animal) model thereof. Accordingly, the marker genes used therein (NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3) belong to a network of genes directly or indirectly regulated by DISCI - associated proteins. This advantageous approach provides high plausibility as it is based on a subgroup of patients with a defined biological cause and thus avoids to be affected by clinical and biological heterogeneity of the various mental disorders. Using the human equivalents of the marker genes selected from the group consisting of NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3, the method according to the invention therefore ensures an accurate and reliable diagnosis of mental disorders that are associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis. The marker genes and proteins used herein also comprise processed proteins or splice variants, i.e. molecules derived from the original gene or protein indicated.

In an advantageous embodiment of the invention a first marker gene is RGS1 and at least one second marker gene is selected from the group consisting of human NKG7, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3. These marker genes or the transcripts or related marker proteins thereof, alone or in various combinations, are particularly advantageous for diagnosing mental disorders associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis. Preferably, RGS1 is combined with CCL4 and/or NKG7. Especially RGS1 is correlated to cognitive endophenotypes of patients and also clearly decreased in patients. On the other hand, the NK cell markers CCL4 and NKG7 have by themselves already a high diagnostic potential, however they are not expressed in the brain. The decreased NK cell genes NKG7 and CCL4 have no overlap with RGS1 in terms of co-regulation. Therefore, RGS1 in conjunction with an NK cell marker is advantageous to diagnose the subset with credibility, plausibility and specificity. No single marker is likely able to do this because one is not expressed in the brain, the other not specific enough.

In another advantageous embodiment of the invention at least one additional marker gene is selected from the human equivalents of the genes listed in

Table 1. Using additional marker may further increase sensitivity and/or sensitivity of the method according to the invention. The marker genes and proteins indicated in Table 1 also comprise processed proteins or splice variants, i.e. molecules derived from the original gene or protein indicated.

In another advantageous embodiment of the invention the measured expression levels are indicative to the mental disorder or disposition if each measured expression level is lower than a respective reference expression level and/or reaches or falls below the predetermined threshold value. That is, a lower expression level of the marker genes in the body tissue or fluid sample relative to the respective expression levels in the normal controls (reference or standard expression levels of a healthy population) is indicative of the presence of the mental disorder, or predisposition thereto. Surprisingly, the marker genes selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3 are all downregulated, i.e. their transcript level is decreased relative to the reference (standard) level of a healthy population. In contrast, prior art assumes increased marker levels, particularly, with markers for pro-inflammatory cytokines. The decreased expression levels of the marker genes according to the invention therefore seem to represent a subset of cases with decreased inflammatory markers. Surprisingly, this is not in contradiction to previous findings because the decrease of these markers in a subset is diluted out and overcompensated by an increase of the same markers in the other subsets such that when the all- comprising clinical diagnosis is used the markers appear slightly increased.

In another advantageous embodiment of the invention, the body tissue or fluid is selected from whole blood, blood plasma, blood serum, cerebrospinal fluid, urine, saliva, biopsy material, and/or isolated cells and their ex vivo derivatives. For example, the body fluid can also be modelled by taking cells from a patient or human individual and reprogramming those to various cell types (induced pluripotent stem (iPS) cells and differentiation of those).

In another advantageous embodiment of the invention the expression levels of the marker genes (transcript levels) may be measured by quantitative reverse transcription Polymerase Chain Reaction (qRT-PCR), quantitative real-time PCR, or any other method suitable to determine the amount of transcript (mRNA or cDNA) within the body fluid. Alternatively, the expression levels of the marker proteins (protein levels) can be measured by any high-affinity binding assay, for example, an immune-/antibody-based assay such as an Enzyme Linked Immunosorbent Assay (ELISA). The high-affinity binding assay is not limited to antibodies but can also consist of peptides, small nucleic acids called aptamers, even small organic molecules, or others. It is also conceivable to combine both techniques to what is called immuno PCR.

For example, the expression levels may be determined using naturally occurring or chemically synthesized compounds capable of specifically binding to the respective marker protein or the transcripts (mRNA) of the marker genes. Such compounds may be selected from the group comprising a peptide, an antibody or a fragment thereof, an aptamer (peptide or oligonucleotide), and an oligonucleotide. The compound may be labelled with a detectable label, such as a luminescent, fluorescent or radioactive group. Alternatively or additionally, the compound may be labelled with an affinity tag, e.g., a biotin, avidin, streptavidin or His (e.g. hexa-His) tag. For high-throughput applications an array comprising the compound may be used, e.g., a microarray in the form of a biochip.

Most notably, and especially if a high-throughput assay shall be established, the expression level of the marker protein is measured by means of a microarray analysis (biochip technology).

The objective is also met by a combination of at least two marker proteins derived from marker genes, or at least two nucleic acid molecules comprising marker genes coding for marker proteins, said marker genes being selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C31 , for use in in vitro diagnostics.

Preferably, a first marker gene is RGS1 and at least one second marker gene is selected from the group consisting of human NKG7, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3.

As the NK cell markers CCL4 and NKG7 have by themselves already a high diagnostic potential (however, they are not expressed in the brain) and their genes have no overlap with RGS1 in terms of co-regulation, RGS1 in

conjunction with an NK cell marker is advantageous to diagnose the subset with credibility, plausibility and specificity. Therefore, it is particularly advantageous if the first marker gene is RGS1 and the second marker gene is NKG7 and/or CCL4.

The combination of markers according to the invention may further comprise at least one additional marker gene selected from the human equivalents of the genes listed in Table 1. The marker genes and proteins indicated in Table 1 also comprise processed proteins or splice variants, i.e. molecules derived from the original gene or protein indicated.

The combination according to invention can be advantageously used in an in vitro method of diagnosing the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder, wherein the mental disorder is associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis.

The invention further concerns a kit for diagnosing the presence of a mental disorder in a human individual or the predisposition of the human individual to the mental disorder in vitro, wherein the mental disorder is associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, the kit comprising:

a) a set of oligonucleotide primers which are suitable to initiate amplification of the transcripts of at least two marker genes, each of which coding for at least one marker protein, in a Polymerase Chain Reaction and/or microarray, wherein said marker genes are selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3, and/or

at least two first antibodies or molecules, each of which specifically binding to a marker protein in a body tissue or fluid from the individual, wherein the marker proteins are derived from marker genes selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3;

b) at least two reporter probes capable of binding to complementary DNA (cDNA) derived from the transcripts, which are suitable to be detected in a quantitative reverse transcription Polymerase Chain Reaction (qRT-PCR), and/or

at least two labelled second antibodies, each of which specifically binding to one of the first antibodies or molecules, which are designed to be detected in a high-affinity binding assay (immune-/antibody-based assay such as Enzyme Linked Immunosorbent Assay (ELISA)); and optionally, c) at least two reference samples.

For example, the kit according to the invention may comprise naturally occurring or chemically synthesized molecules capable of specifically binding or hybridizing to the marker proteins or the transcripts (mRNA) of the marker genes. Such molecules may be selected from the group comprising a peptide, an antibody or a fragment thereof, an aptamer (peptide or oligonucleotide), and an oligonucleotide (primer). Some molecules may be labelled with a detectable label, such as a luminescent, fluorescent or radioactive group.

In an advantageous embodiment of the invention said kit comprises at least one set of oligonucleotide primers selected from the group consisting of

a) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:1 and SEQ ID NO:2;

b) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:3 and SEQ ID NO:4 c) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:5 and SEQ ID NO:6;

d) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:7 and SEQ ID NO:8;

e) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:9 and SEQ ID NO:10;

f) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:1 1 and SEQ ID NO:12;

g) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:13 and SEQ ID NO:14

h) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:15 and SEQ ID NO:16;

i) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:17 and SEQ ID NO:18;

j) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:19 and SEQ ID NO:20;

k) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:21 and SEQ ID NO:22;

I) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:23 and SEQ ID NO:24;

m) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:25 and SEQ ID NO:26;

n) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:27 and SEQ ID NO:28;

o) a set of oligonucleotide primers comprising the nucleic acid sequences according to SEQ ID NO:29 and SEQ ID NO:30;

p) nucleic acid molecules, the polynucleotide sequence of which is at least

90%, preferably 95%, identical to the nucleotide sequence of a

oligonucleotide primer of any of a) to o), and which is capable of binding to complementary DNA (cDNA) derived from the transcripts of a gene coding for at least one marker protein selected from the group consisting of the proteins listed in Table 1 ;

q) nucleic acid molecules, the complementary strand of which hybridizes to a nucleic acid molecule of any of a) to p) under stringent conditions; r) nucleic acid molecules, the nucleotide sequence of which is

complementary to the nucleotide sequence of a nucleic acid molecule of any of a) to q).

The primer sequences are shown in Table 2 (SEQ ID NOS refer to the primers for amplifying the human marker genes).

The invention further concerns a method for determining the response to at least one pharmaceutical compound able to correct a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, wherein the expression levels of at least two marker genes are determined and compared according to steps a) and b) of the method according to claim 1 , and wherein the measured expression levels indicate that the response to the pharmaceutical compound is positive if each aberrant expression level of said marker genes is normalized or at least improved. Accordingly, a method for monitoring the therapeutic efficacy of a pharmaceutical compound in an individual having a mental disorder is provided, comprising a comparison of a current expression level of the marker genes present in a body tissue or fluid sample of said individual after

administration of the pharmaceutical compound with at least one sample taken earlier from the same individual, e.g., prior to commencement of the therapy, and/or from the same individual at an earlier stage of therapy. If the expression levels of the marker genes are changed by the application of the

pharmaceutical compound in a way that it is at least partially normalized towards the respective average expression level of a healthy population (i.e. reference (standard) expression level), the therapeutic response to the pharmaceutical compound is positive. That is, a therapy is effective if the difference between the current expression levels and the threshold values is decreased in relation to the difference between the earlier expression levels and the threshold values. With this method according to the invention it is possible to analyse the efficacy of existing pharmaceutical compounds or those that are not developed using a transgenic animal as described below.

The protein pathology of a subset of sporadic mental disorders as outlined above can be modeled by means of a transgenic animal (e.g. by modestly overexpressing the non-mutant full length human DISC1 transgene: tgDISCI rat) and presents a novel pharmacological target in mental illness drug discovery. The concept of "reverse translation" assumes the existence of subsets of mental disorders from the outset and thus starts marker search with a biologically defined subset rather than a mix of heterogeneous cases merely defined by clinical diagnosis. Genetic or protein pathology of patients/families can be modeled in animals and then markers can be identified in this animal model. Markers can be "reverse translated" to sporadic patients in order to define those biological subsets ante mortem, i.e. in live patients. Animal model, marker and patient subsets can then be paired in order to develop tailored therapies.

To this end, a nonhuman transgenic animal useful for providing organs, tissues, or cells, which is able to stably express a modified gene coding for human DISC1 protein, wherein the expression level of the modified gene is higher than that of the respective wild-type gene and thus results in the formation of aggregates of the DISC1 protein within the cells, said animal representing a subset of human subjects having at least one mental disorder, is provided for use in the identification and analysis of marker proteins or genes for diagnosing mental disorders in human individuals. The transgenic animal may be a rodent, preferably a rat.

The invention further includes a method for identifying and analysing marker proteins or genes for diagnosing mental disorders in human individuals using said transgenic animal.

A transgenic rat model modestly overexpressing the full-length DISC1 transgene (named "tgDISCI rat"), shows phenotypes consistent with a significant role of DISC1 misassembly in a subset of sporadic mental disorders. The tgDISCI rat displays mainly perinuclear DISC1 aggregates in neurons. Furthermore, the tgDISCI rat shows a robust signature of behavioural phenotypes that includes amphetamine supersensitivity, hyperexploratory behaviour, rotarod deficits, as well as aberrant dopamine neuroanatomy and neurochemistry, all pointing to changes in dopamine (DA) neurotransmission. Elevated cytosolic dopamine causes an increase in DISC1 multimerization, insolubility and complexing with the dopamine transporter, suggesting a physiological mechanism linking DISC1 assembly and dopamine homeostasis. DISC1 protein pathology and its interaction with dopamine homeostasis is a novel cellular mechanism that is relevant for behavioural control and may have a role in mental disorders (Trossbach 2016). Neuroanatomical analysis revealed a reduced density of dopaminergic neurons in the substantia nigra and reduced dopaminergic fibres in the striatum of the transgenic rat. Parvalbumin- positive interneuron occurrence in the somatosensory cortex was shifted from layers ll/lll to VA/I, and the number of calbindin-positive interneurons was slightly decreased. Reduced corpus callosum thickness confirmed trend-level observations from in vivo MRI and voxel-wise tensor based morphometry.

These neuroanatomical changes help explain functional phenotypes of this animal model, some of which resemble changes observed in human

schizophrenia post mortem brain tissues. DISC1 overexpression or

misassembly can account for a variety of seemingly unrelated morphological phenotypes and thus provides a possible explanation for findings observed in sporadic schizophrenia patients (Hamburg et al. 2016).

Accordingly, the tgDISCI rat reflects neuropathological features of a subset of sporadic cases with CMI and has behavioral (amphetamine sensitization) and biochemical (D2R high switch) features very similar to human patients with schizophrenia, and therefore exhibits high face validity. On a molecular mechanistic level, DISC1 missassembly seems to modulate dopamine homeostasis so that the tgDISCI rat is a useful model for a subset of sporadic CMI, advancing biological diagnostics and therapy. Further, DISCI opathies define a biology-based category of human mental disorders with involvement of the dopamine system that can be modeled in animals, i.e. in the tgDISCI rat. The tgDISCI rat represents a subset of patients with schizophrenia (or other mental illnesses), termed "DISCI opathies".

It is conceivable that human induced pluripotent stem (iPS) cells modestly overexpressing a similar DISC1 transgene, and differentiated to various cells or brain organoids, including human PBMC subpopulations, might also be used for the purpose of identifying possible markers and patient-tailored therapies but with the shortcoming that these cell systems are not amenable to behavioural testing paradigms. However, for specific applications, the invention provides at least one human induced pluripotent stem (iPS) cell which is able to stably express a modified gene coding for human DISC1 protein, wherein the expression level of the modified gene is higher than that of the respective wild- type gene and thus results in the formation of aggregates of the DISC1 protein within the cell. Such iPS cell can also be used for in the identification and analysis of marker proteins or genes for diagnosing mental disorders in human individuals. The invention further includes a method for identifying and analysing marker proteins or genes for diagnosing mental disorders in human individuals using such iPS cells.

With the concept of reverse translation, a distinct marker set can be identified, that may not be complete but that is sufficient to allow fairly specific blood diagnostics of DISCI opathies. The blood test may be used to identify patients that may profit from (future) curative pharmacotherapies also effective in the tgDISCI rat. However, they may also profit from existing merely symptomatic pharmacotherapies targeting neurotransmitter systems by indicating which patient subsets are likely to respond to a dopamine homeostasis-modifying drug. For example, a patient that has been tested positive for a DISCI opathy may receive one particular dopamine-homeostasis reinstating drug with priority rather than testing various drugs based on clinical guesses as is current clinical practice.

Curative drugs are defined as drugs that target the biological cause, for example DISC1 protein pathology, of a mental disorder whereas symptomatic drugs are drugs that target one downstream consequence of the biological causes, for example aberrant dopamine homeostasis. Curative drugs are advantageous because they eventually eliminate all downstream aberrances, rather than only one.

The invention further relates to a method for determining the therapeutic effect of a potentially curative pharmaceutical compound on a mental disorder, or a predisposition of a human individual to the mental disorder, associated with a dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis, wherein said pharmaceutical compound is administered to a transgenic animal (the transgenic animal being used as an indicator for a successful or

unsuccessful therapy of the mental disorder), and wherein it is indicated that the therapeutic effect is positive if aberrant expression levels of at least two marker genes selected from the group consisting of human NKG7, RGS1, CCL4, IFNG, IL12RB2, IL13RA1, KMO, FPR2, SLC27A2, and C3 are normalized in said transgenic animal after administration of said pharmaceutical compound. The invention then allows to test a patient for said markers and assign a curative pharmacotherapy to that patient.

In an advantageous embodiment of the invention the transgenic animal is a nonhuman transgenic animal useful for providing organs, tissues, or cells, which is able to stably express a modified gene coding for human DISC1 protein, wherein the expression level of the modified gene is higher than that of the respective wild-type gene and thus results in the formation of aggregates of the DISC1 protein within the cells, said animal representing a subset of human subjects having at least one mental disorder. As explained in detail above, such animal represents a subset of patients with schizophrenia (or other mental illnesses), termed "DISCI opathies".

It is possible that an aberrant DISC1 pathway is also encountered in human individuals that are not considered clinically sick. This may be due to a variety of causes also termed resilience factors. These individuals, however, may reveal subtle cognitive impairments upon closer inspection that may improve with suitable drugs which would then be called cognitive enhancement rather than pharmacotherapy. That is, a potential therapy for clinically sick patients might also be used as cognitive enhancer for healthy individuals.

The term "marker gene" as used herein refers to a distinctive gene coding for a distinctive protein or peptide (herein referred to as "marker protein") suitable to be used as an indicator of a biological, biochemical, or physiological process, event, or condition within a body, tissue, or cell. For example, a marker gene can be used to detect at least one biological, biochemical, or physiological symptom of a disease by detection of its transcript or protein. In this context, "distinctive" means that the marker gene or marker protein is accessible to be detected via a physical, physico-chemical, or electro-physical detection method, either directly or indirectly by means of at least one detectable (labelled) compound or composition. The marker gene can be detected either by detecting the marker protein or by detecting the transcripts (mRNA, or indirectly: cDNA) of the marker gene. If specific marker proteins are named herein, all derivatives thereof comprising processed proteins or splice variants, i.e.

molecules derived from the original gene or protein indicated, shall be included.

The term "marker" as used herein includes both the marker gene and the marker protein.

The term "expression level" (or "expression level of the marker gene") as used herein refers to the amount of transcript (mRNA) of a gene coding for a specific peptide or protein (transcript level), or the amount of specific peptide or protein derived from this gene (protein level), within a body, tissue, cell, or fluid sample. The expression level can be measured, for example, by quantitative

determination of the amount of either mRNA (or cDNA) or translated protein within a sample.

The term "aberrant expression level" as used herein refers to an abnormal expression level of a marker gene, which significantly differs from the average expression level of the same marker gene in a healthy population and is involved in the clinical manifestation of a disease and/or represents a symptom of a disease, e.g., a mental disorder.

The invention is further exemplarily described in detail with reference to the figures.

Brief description of the figures Figure 1 shows two alternative, complementary and selective (incomplete) depictions of the DISC1 pathway, demonstrating that the DISC1 protein interacts with many proteins and signaling pathways that have independently been described for mental illness or other chronic brain disease conditions.

Figure 2 shows bar diagrams representing an independent validation of a selection of markers from Table 1 in the tgDISCI rat (TG, gray) vs. non- transgenic littermates (LM, white) using quantitative polymerase chain reaction (qPCR). Marker names abbreviated, for full name, see Table 1 .

Figure 3 shows bar diagrams representing a screening of a selection of markers from Table 1 in a population of schizophrenia patients vs. controls (n = 20 for each group); cohort of patients with schizophrenia (SCZ, gray) vs. healthy controls (CTRL, white). Marker names abbreviated, for full name, see Table 1 .

Figure 4 shows graphical representations demonstrating the correlation of different markers in the tgDISCI rat and schizophrenia patients. A. Correlation matrix of markers in the rat (left) and human (right). B. Selective depiction of single correlations appearing similar in the transgenic tgDISCI rat (TG) vs. non- transgenic littermates (LM), and schizophrenia patients (SCZ) vs. healthy controls (CTRL).

Figures 5a and 5b show tables of a Spearman correlation (non-parametric) of human markers (see Table 1 ) demonstrating that single correlations either exist between patients and controls, or only for patients or controls

demonstrating the disruption or pathological creation and stabilization of regulated networks of markers (all data analyzed without outliers).

Figure 6 shows a bar diagram demonstrating detection specificity and sensitivity related to the clinical diagnosis of schizophrenia (SCZ, dark grey; healthy controls (HC), light grey) when a selection of single markers from Table 1 is investigated. The threshold was defined at being below 50% of the average of the healthy control group. Figure 7 shows a bar diagram demonstrating detection specificity and sensitivity related to the clinical diagnosis of schizophrenia (SCZ, dark grey; healthy controls (HC), light grey) when a selection of a combination of two or three markers from Table 1 is investigated. The threshold was defined at being below 50% of the average of the healthy control group.

Figure 8 shows a graph representing normalized expression levels (brain) of RGS1 transcripts in controls (CTRL), schizophrenia (SCZ) and Bipolar Disorder (BP) samples.

Description of exemplary and preferred embodiments of the invention Subjects and classifications

Control subjects and patients diagnosed with schizophrenia were part of a clinical study as described by Warbrick et al. (201 1 ) and Trossbach et al.

(2014).

Animals

Animal experiments were executed in conformity with the German Animal Protection Law and were authorized by local authorities (LANUV NRW,

Recklinghausen, Germany). Experiments were performed with transgenic Sprague Dawley rats overexpressing full-length, non-mutant DISC1 carrying the polymorphisms L607F (rs6675281 ) and S704C (rs821616) (tgDISCI rat;

Trossbach et al., 2016) and non-transgenic littermates. Male tgDISCI and control rats were bred at the Heinrich Heine University Dusseldorf, Animal Facility, Germany. Animals were housed three animals per cage under standard laboratory conditions with lights on from 0700 hours to 1900 hours and with water and food provided ad libitum. Blood extraction and preparation of lymphocytes was performed with adult tgDISCI rats and littermate controls at the age of 8-9 months.

Preparation of lymphocytes from blood

Anaesthetized rats underwent a heart puncture to harvest a minimum of 8 mL of blood. Rat lymphocytes were prepared with the Ficoll-Paque Premium 1 .084 solution (GE Healthcare, Little Chalfont, United Kingdom) according to manufacturer's instructions. Preparation of human lymphocytes was performed with Ficoll-Paque Plus (GE Healthcare, Little Chalfont, UK) as described by Trossbach et al. (2014). Lymphocyte samples were snap-frozen in liquid nitrogen and stored at -80°C until further processing.

Preparation of RNA and cDNA

RNA of rat and human lymphocytes was prepared utilizing the RNeasy Mini Kit according to manufacturer's guidelines. Residual genomic DNA was digested on column by the RNase-free DNasel Set (both Qiagen, Hilden, Germany). RNA was diluted to a concentration of 100 ng/ L and 1 g was used as input for the production of cDNA with the RevertAid First Strand Synthesis Kit in a total of 20 μί utilizing the random hexamer primers provided by the kit (Thermo Fisher Scientific, Waltham, MA, USA). The resulting cDNA was diluted 1 :50, 1 :25 or 1 :50 dependent on PCR results as indicated in Table 1 and 5 μί were used as template input.

Gene expression profiling

Total RNA preparations were checked for RNA integrity by Agilent 2100

Bioanalyzer quality control. All samples in this study showed high quality RNA Integrity Numbers (RIN >9). RNA was further analysed by photometric

Nanodrop measurement and quantified by fluorometric Qubit RNA assays (Life Technologies). Synthesis of biotin labeled cDNA was performed on ten replicates of each experimental group (DISC1 transgenic (TG) rats and littermate (LM) controls, respectively) according to the manufacturers ' protocol (WT Plus Reagent Kit; Affymetrix, Inc). Briefly, 100 ng of total RNA were converted to cDNA. After amplification by in vitro transcription and 2nd cycle synthesis, cDNA was fragmented and biotin labeled by terminal transferase. Finally, end labeled cDNA was hybridized to Affymetrix Rat Gene 2.0 ST Gene Expression Microarrays for 16h at 45 °C, stained by strepatavidin/phycoerythrin conjugate and scanned as described in the manufacturers ' protocol.

Three samples (2x TG, 1 x LM) did not pass hybridization quality control, two additional samples (3x TG, 2x LM) had to be excluded from further analyses because of abnormal ventricle volume. Data analyses on 12 Affymetrix CEL files were conducted with GeneSpring GX software (Vers. 12.5; Agilent Technologies). Probes within each probeset were summarized by GeneSprings ' ExonRMA16 algorithm after quantile

normalization of probe level signal intensities across all samples to reduce inter- array variability (Bolstad et al., 2003). Input data pre-processing was concluded by baseline transformation to the median of all samples. To further improve signal-to-noise ratio, a given probeset had to be expressed above background (i.e. fluorescence signal of a probeset was detected within the 20th and 100th percentiles of the raw signal distribution of a given array) in all replicates in at least one of two, or both conditions to be subsequently analysed in pairwise comparison. Differential gene expression was statistically determined by moderated T-test. The significance threshold was set to p = 0.01 .

Quantitative expression analysis

For the verification of differential expression target primers were tested by PCR using the HotStarTaq (Qiagen, Hilden, Germany). Primer sequences, dilutions and PCR supplements are depicted in Table 2. Effective primers were used for quantitative Real Time PCR (qPCR) with the StepOnePlus Real-Time PCR System (Applied Biosystems, Carlsbad, CA, USA) and the Platinum SYBR Green qPCR SuperMix-UDG (Invitrogen, Carlsbad, CA, USA) in MicroAMP Fast Optical 96-Well Reaction Plates (Applied Biosystems, Carlsbad, CA, USA). Depending on the target, 5% Factor Q solution (Qiagen, Hilden, Germany) was added to the mix. QPCR conditions: 10 min at 95°C, 40 cycles of 15 s at 95 °C and 60°C for 1 min. The resulting data were processed with the corresponding StepOne Software v2.3 (Thermo Fisher Scientific, Waltham, MA, USA). The expression of the respective target was normalized to the expression level of the housekeeping gene Actin (rat) or ARF1 (human), as well as against a rat or human control cDNA per plate to minimize variances between runs.

As shown in Figure 2, the expression level (relative mRNA expression) of each of the marker genes Ifng, Ccl4, H13ra1, H12rb2, C3, and Slc27a2 from Table 1 is significantly decreased in the tgDISCI rat (TG, gray) in relation to non- transgenic littermates (LM, white). This result indicates that the decreased expression level of these marker genes observed in the microarray analysis (Table 1 ) can be repeated by an independent detection method (quantitative polymerase chain reaction; qPCR). Marker genes Ifng, Ccl4, H13ra1, H12rb2, C3, and Slc27a2 are all downregulated, i.e. the transcript level is decreased relative to the reference (standard) level of the "healthy" littermates. That is, the lower expression level of each tested marker gene in the transgenic rat relative to the expression level of the respective marker gene in the non-transgenic control is indicative of dysfunctional DISC1 protein pathway or disturbed dopamine homeostasis. The altered expression level can thus be deemed to be indicative to neuropathological and biochemical features very similar to human patients with schizophrenia.

Figure 3 shows a screening of marker genes IFNG, CCL4, IL13RA1, IL12RB2, RGS1, C3, and SLC27A2 from Table 1 in a population of schizophrenia patients (SCZ, gray) vs. healthy controls (CTRL, white), demonstrating that the decreased expression levels (relative mRNA expression) of the same markers in the tgDISCI model are also observed in a cohort of patients with

schizophrenia. Marker genes IFNG, CCL4, IL13RA1, IL12RB2, RGS1, C3, and SLC27A2 are all downregulated, i.e. the transcript level is decreased relative to the reference (standard) level of the healthy controls. Accordingly, the altered expression level can be deemed to be indicative to schizophrenia in human individuals.

Figure 4 shows correlations of different markers in tgDISCI rats and human individuals, demonstrating similarity between the rat system and the human system. The alterations of the expression levels of the tested markers relative to the respective reference expression levels are similar in the tgDISCI rat and schizophrenia patients. Thus, transfer of the principles and mechanisms observed in the rat system to the human system is reasonable.

Figures 5a and 5b show correlation tables of human markers demonstrating that single correlations either exist between patients and controls, or only for patients or controls. The cross-correlations between single markers show that disease can either disrupt existing correlating networks or stabilize new

(pathological) ones and demonstrates that the identified markers according to Table 1 are functionally interconnected.

Figures 6 and 7 demonstrate the detection specificity and sensitivity related to the clinical diagnosis of schizophrenia when a selection of single markers from Table 1 (RGS1 , CCL4, and NKG7; Figure 6) or a selection of a combination of two or three markers from Table 1 (RGS1 + CCL4 and/or NKG7, and CCL4 + NKG7; Figure 7) is investigated. Basically it is demonstrated that specificity of the method according to the invention is very high while sensitivity is rather low. However, in the method according to the invention specificity is more important than sensitivity since the method is provided for a sub-group of patients only and thus low sensitivity relating to the all-comprising clinical diagnosis is expected and can therefore be neglected. Figure 7 shows that the combination of two or more markers can significantly increase specificity of the method according to the invention, cf. marker gene RGS1 (alone: 88% => + CCL4 and/or NKG7: 94% / 97%).

Figure 8 shows that, surprisingly, RGS1 expression in brain is clearly decreased in patients (Schizophrenia and Bipolar Disorder) compared to healthy controls. RGS1 is correlated to cognitive endophenotypes of patients and seems to be the best marker for diagnosing related diseases. RGS1 is significantly correlated to attention and memory in the Digital Symbol Test (DSST) indicating high clinical relevance of this marker to measurable cognitive deficiencies (data not shown). The decrease in RGS1 expression is not due to a decreased number of macrophages where it is expressed and which would correspond to the cell lineage where microglia is also derived from, the only cell type of the brain expressing RGS1 (data not shown). RGS1, preferably in conjunction with an NK cell marker, therefore seems to be advantageous to diagnose the patient subsets with credibility, plausibility and specificity.

Accordingly, RGS1 seems to crystallize as the most important marker compared to the other markers listed in Table 1 . However, RGS1 expression levels seem also changed in diseases like melanoma, multiple sclerosis or others, so that at least a second marker can be beneficial. For example, the NK cell markers NKG7 or CCL4 may be of particular importance (but

interchangeable). In the rat NKG7 and CCL4 indicate a decrease in expression levels, in humans it is not clear whether they are decreased due to a decrease in NK cell number, a decrease in expression per NK cell, or both. In summary, diagnostics could be prioritized to RGS1 and one or two NK cell markers such as NKG7 and CCL4, cf. Figure 7. In patients, NK cell markers seem decreased due to a decrease in NK cell numbers, but a simultaneous decrease in expression levels cannot be excluded (data not shown).

Non-patent literature:

1 . Bolstad et al. (2003): A comparison of normalization methods for high

density oligonucleotide array data based on variance and bias.

Bioinformatics, 19, 2, 185-193, 2003.

2. Chan et al. (2015): "Development of a blood-based molecular biomarker test for identification of schizophrenia before disease onset." Translational Psychiatry (2015) 5, e601 ; doi:10.1038/tp.2015.91 ; published online 14 July 2015.

3. Dean (201 1 ): "Dissecting the Syndrome of Schizophrenia: Progress toward Clinically Useful Biomarkers." Schizophrenia Research and Treatment Volume 201 1 , Article ID 614730, 10 pages, doi:10.1 155/201 1/614730.

4. Hamburg et al. (2016): "Simultaneous effects on parvalbumin-interneuron and dopaminergic system development in a transgenic rat model for sporadic schizophrenia." Scientific Reports | 6:34946 | DOI:

10.1038/srep34946.

5. Hennah & Porteous (2009): "The DISC1 pathway modulates expression of neurodevelopmental, synaptogenic and sensory perception genes." PLoS ONE 2009; 4(3):e4906

6. Howes & Kapur (2009): "The dopamine hypothesis of schizophrenia:

version I II— the final common pathway." Schizophr Bull. 2009 May;

35(3):549-62. doi: 10.1093/schbul/sbp006. Epub 2009 Mar 26.

7. Korth (2009): "DISCopathies: brain disorders related to DISC1 dysfunction." Rev Neurosci 2009; 20(5-6):321 -30

8. Korth (2012): "Aggregated proteins in schizophrenia and other chronic

mental diseases: DISCI opathies." Prion 6(2):134-41 .

9. Korth (2012): "Aggregated proteins in schizophrenia and other chronic

mental diseases." Prion 6:2, 1 -8. April/May/June 2012.

10. Soares et al. (201 1 ): "DISC1 : Structure, Function, and Therapeutic Potential for Major Mental Illness." ACS Chem Neurosci. 201 1 Nov 16;2(1 1 ):609-632. Epub 201 1 Aug 5.

1 1 . Trossbach et al. (2014): "Peripheral DISC1 protein levels as a trait marker for schizophrenia and modulating effects of nicotine." Behav Brain Res 275C: 176-182. Trossbach et al. (2016): "Misassembly of full-length Disrupted-in- Schizophrenia 1 protein is linked to altered dopamine homeostasis and behavioral deficits." Molecular Psychiatry (2016), 1-12.

Warbrick et al. (201 1 ): "Direction and magnitude of nicotine effects on the fMRI BOLD response are related to nicotine effects on behavioral performance." Psychopharmacology (Berl) 215(2): 333-344.

;) BS T I T U T ERUL E 26

Table 1 TG versus LM

Gene symbol [rat] Gene description Genbank Refseq Change FC P -val

1 Rgsl regulatorofG-proteinsignalingl BC098681 NM_019336 2,03 0,006

2 Ccl4 chemokine(C-Cmotif)ligand4 U06434 NM_053858 t 1,67 0,000 formylpeptidereceptor21 formylpeptide-

3 Fpr2 | Fpr2l XM_001073508///; t 1,65 0,005 receptor2-like

4 C3 complementcomponent3 NM_016994 t 1,63 0,004

5 Nkg7 naturalkillercelIgroup7sequenee AF082535 NM_133540 1,60 0,000

6 Ill2rb2 interleukinl2receptor,beta2 NM_001191750 * 1,59 0,009 serine(orcysteine)proteinaseirihibitor,

m 7 Serpinbla BC098686 N _001031642

cz cladeB,memberla * 1,52 0,007

8 Ly49si3 AY653730 NM_001009919 * 1,51 0,006 phospholipaseA2,groupVII(platelet-

9 Pla2g7 BC088457 N _001009353

activatingfaetoracetylhydrolase,plasma) * 1,48 0,006 m similartoH2Ahistonefamily,memberO |

10 Hist2h2aa3 XM_002726027 * 1,47 0,000 m histonecluster2, H2aa3

m

— soluteearrierfamily27(fattyaeid-

11 Slc27a2 D85100 NM_031736 t 1,46 0,009 transporter),member2

12 RGD1559149 simto60SribosomalproteinL7a 1,46 0,003

13 Ill3ral interleukinl3receptor,alphal BC093615 NM_145789 1,45 0,007 cytochromeP450,family4,subfamilyf,

14 Cyp4fl8 BC101918 NM_001033686

polypeptidel8 * 1,44 0,004

15 RpllO ri boso ma I p rote i n L10 BC058467 1,41 0,006

PREDICTED: Rattus norvegicus alkaline

18 Acer3

ceramidase 3 (Acer3), mRNA * 1,38 0,003

19 Ifng interferongamma AF010466 N _138880 * 1,38 0,001

40 Tyrobp Tyroproteintyrosinekinasebindingprotein AY247021 NM_212525 * 1,31 0,001

41 Itm2a integraImembraneproteiri2A BC099174 NM_001025712 1,30 0,002 killercelllectin-

42 Klrgl X79812 NM_031649

likereceptorsubfamilyG,memberl * 1,30 0,005

43 Srgap3 SUT-ROBORhoGTPaseactivatirtgprotein3 NM_001191975 * 1,30 0,004

44 Tlr5 toll-likereceptor5 FJ750588 NM_001145828 * 1,30 0,007

45 Slamf8 SLAMfa mi lymem ber8 IM _001105973 * 1,30 0,002

46 Olrl531 olfactoryreceptorl531 NM_001001102 4 1,30 0,004

47 Clec4a2 C-type lecti ndomai nfa mi iy4, me mber A2 AY494061 NM_001005880 1,30 0,009

StAR-relatedlipidtransfer

48 Stard3

(START)domaincontaining3 * 1,30 0,000 histoneelusterl,H2ac | H2ae-

49 Histlh2ac XM_003751712

like| H2ai | H2an | H4m * 1,29 0,001

50 Eno3 enolase3, beta, muscle BC083566 NM_012949 * 1,29 0,004

51 iflS kinesinfamilymemberlS AY291581 NM_181635 1,29 0,007

54 Sodl superoxidedismutasel,soluble FQ220715 NM_017050 * 1,29 0,004

55 Cd55 BC061869 NM_022269 * 1,29 0,002

56 Ly6c Ly6-Cantigen NM_020103 * 1,29 0,009

57 Tuba3a |Tuba3b tubulin,alpha3A|tubulin,alpha3B BC079395 NM_00104G008 1,28 0,001

58 LOC252890 Z39smallnucleolarRNA NR_002705 * 1,28 0,004

59 Sytl3 synaptotagmin-like3 BC166706 NM_001127560 1,28 0,007 lymphotoxinbetareceptor

60 Ltbr BC085880 NM_001008315

FRsuperfamily,member3) * 1,28 0,003

(TN

61 Zfp580 zincfingerprotein580 XM_218196///XM. 4 1,28 0,000

104 Pspn persephin AF040961 NM . _013014 4 1,21 0,005

105 Tlr2 toll-Iikereceptor2 AY151255 NM. , 198769 * 1,20 0,006 zincfingerprotein4181 similarto

106 Zfp418 FQ212491 NM . _001191620 4 1,20 0,008 zincfingerprotein418

ATPase,H+transporting,lysosomalVl

107 Atp6vlg3 ISIM. _001105991 4 1,20 0,003 subunitG3

108 LOC680549 similartoPbx/knottedlhomeobox2 FQ221048 4 1,20 0,010

109 LOC100363043 ehromosomel4openreadingframell9 XM_ .002725161 1,20 0,006

110 LOC100364650 rCG38872-like 4 1,20 0,009

111 Pgm2 phosphoglucomutase2 BC160893 NM. _001106007 * 1,19 0,005

112 Mospd3 motilespermdomaincontaining3 BC099230 NM . _001025629 * 1,19 0,004 transmembraneandeoiled-

113 Tmccl

eoildomainfamilyl * 1,19 0,008 mesencephalicastrocyte-derived

114 Manf BC166980 NM . _001108183 * 1,19 0,000 neurotrophicfactor

tubulin,gammaeomplexassociated

115 Tubgcp6 NM. _001108748 4 1,19 0,001 protein6

116 Cldn24 claudin24 NM . _001110144 * 1,19 0,007

117 Ccdc90b coi led-coi Idomai ncontaini ng90B BC097480 M. _001024885 * 1,19 0,001

118 Chic2 cysteine-richhydrophobicdomain2 NM . _001105736 * 1,19 0,010

ISJADHdehydrogenasef ubiquinone)

119 NdufsS BC168721 NM. _001030052 4 1,18 0,002

SproteinS

120 Etv6 etsvariant6 BC105773 NM . _001037353 * 1,18 0,004

121 Banp Btg3associatednuclearprotein BC160844 NM. _001106191 * 1,18 0,007

122 GD1560608 similartonovelprotein NM . _001109280 4 1,18 0,008

123 Npm3 nucleophosmin/nucleoplasmin,3 XM_ .577868 1,18 0,005

124 Olr240 olfactoryreceptor240 XM_ . 001076482 4 1,18 0,000

125 Ptgdr2 prostaglandinD2receptor2 AY228550 NM. _001012070 t 1,18 0,003

126 Nkd2 nakedcuticlehomolog2(Drosophila) NM . _001107454 4 1,18 0,008

127 LOC500959 triosephosphateisomerase AY461585 NM. _001033072 * 1,18 0,001

ERdegradationenhancer,mannosidase

128 Edem2 BC079029 NM_001004230 t 1,18 0,005 alpha-like2

129 Fgf4 flbroblastgrowthfactor4 AB079673 |AF260830 NM_053809 1,18 0,003

130 Polk polymerase(DNAdirected)kappa BC166778 NM_138516 * 1,18 0,001

131 Huslb HUSlcheekpointhomoiogb(S.pombe) N _001134846 1,18 0,005

NADHdehydrogenase(ubiquinone)lbeta

132 Ndufb4 FQ217067 NM_001037338

subcomplex4 * 1,18 0,006

133 Arhgap9 RhoGTPaseactivatingprotein9 BC107938 NM_001080789/// * 1,17 0,002

134 Crhrl corticotropinreleasinghormone receptorl L254381 EU0124381 EUO NM_030999 1,17 0,001 m

a 135 LOC100366245 rCG30616-like 4 1,17 0,004 m

H 136 RGD1565819 | Zfp831 similartoC20orfl741 zincfingerprotein831 NM_001171096 1,17 0,004 a

— 137 DefalO defensinalphalO AY623754 N _001033074 4 1,17 0,003 m

m 138 RGD1565819 | Zfp831 similartoC20orfl741 zincfingerprotein831 NM_001171096 4 1,17 0,005 m

m

— 139 Pvrl4 poliovirusreceptor-related4 FQ228642 NM_001109076 4 1,17 0,007

73

a 140 Rapsn receptor-associatedproteinofthesynapse NM_001108584 4 1,17 0,001 m 141 Opa3 opticatrophy3(human} BC168945 1,17 0,007

142 Pnmal paraneoplasticantigenMAl AF335505 NM_130820 4 1,17 0,008 olfactoryreceptorl4711 olfactory

143 Olrl471 | LOC100360028 NM_001000722 4 1,17 0,009 receptorOlrl471-like

144 Arrdcl arrestindomaincontainingl BC158871 NM_001100770 * 1,17 0,002

145 Erp29 endoplasmicreticulumprotein29 BC091129 NM_053961 t 1,16 0,006

146 Zcrbl zincfingerCCHC-typeandRNAbindingmotifl BC099747 NM_001034940 * 1,16 0,009 coiled-coil-helix-coiled-coil-helixdomain

147 LOC100364342 |Chchd4 4 1,16 0,002 containing4-like

148 RGD1559979 similartoAPHlBhomolog(C.elegans) XM_003754686/// t 1,16 0,004

149 Mir3558 microRNAmir-3558 NR_037340 4 1,16 0,003

150 Tulpl tubbylikeproteinl NM 001107642

151 LOC100360334 forminhomology2domaincontaining3-like BC090338

familywithsequencesimilarityl58,

152 Faml58a BC086432

memberA

ADP-ribosylation-likefactor6interaeting

153 Arl6ip6 BC079329 NM_001024310 protein6

succinatedehydrogenasecomplex,

154 Sdhb BC158620 NM_001100539 subunitB, ironsulfur(lp)

sre-relatedkinaselaekingC-terminal

155 Srms regulatorytyrosineandN-terminal BC090006 NM_0G1011961 myristylationsites

aldehydedehydrogenasel6family,

156 Aldhl6al BC101860 NM_001033706 memberAl

157 Id2 inhibitorofDNAbinding2 BC086391 NM_013060

158 LOC688495 NM_001135252

159 Prrx2 paired related homeobox2 NMJ301105739 translocaseofoutermitochondrial

160 Tomm20l XM_001072851 membrane20 homolog(yeast)-like

161 Vtn vitronectin BC105821 MM_019156

162 Lipg lipase,endothelial AY916123 NM_001012741

163 Dpf2 D4,zincanddoublePHDfingersfamily2 NM_001108516

164 Adcy2 adenylatecyclase2(brain) M80550 NM_031007

165 Dclrela DNAcross-linkrepairlA NM_001106201

170 p21protein(Cdc42/Rac)-activatedkinase6 NM_001106498 1,14 0,010 tyrosine3-monooxygenase/tryptophan5-

171 monooxygenaseactivationprotein, BC076502 NM_019377 + 1,14 0,009 betapolypeptide

gamma-aminobutyricacid(GABA)A

172 Gabrd M35162 NM_017289 1,14 0,009 receptor,delta

173 Polg polymerase(DNAdireeted),gamma NM_053528 1,13 0,003 caspaserecruitmentdomainfamily,

on 174 CardlO NM_001130554 1,13 0,006 a memberlO

175 Zic5 Zicf a mi lymem ber5 IMM_001108391 4 1,13 0,007 proline/arginine-richendleucine- c 176 Prelp AF163569 NM_053385 4 1,13 0,003 —\ richrepeatprotein

177 Cldnl8 claudinl8 4 1,13 0,001 proteinphosphatasel,

178 Ppplr36 BC079166 NM_001013944 1,13 0,005 — regulatorysubunit36

179 MGC125002 similartoRIKENcDNA5830433M19 BC105829 1,13 0,001

180 Limchl LIMandcalponinhomologydomainsl FQ226534 NM_001191678 4 1,13 0,009 181 Prrl9 prolinerichl9 BC168703 NM_001173428 4 1,13 0,004 3

186 Olr6 olfactoryreceptor6 NM_001000539 4 1,12 0,008

187 Gpr68 Gprotein-coupledreceptor68 NM_001108049 4 1,12 0,004

188 opticatrophy3(human) BC168945 NM_001107486 4 1,12 0,005

189 transmembraneprotease,serine2 BC061712 NM_130424 4 1,12 0,003

190 Wfdc3 WAPfour-disulfidecoredomain3 NM_001106541 4 1,12 0,010

adisintegrinandmetallopeptidase

191 Adamla BC081807 NM_020078 , ' 1,12 0,005 domainla

FATtumorsuppressorhomolog3

192 Fat3 AB076401 NM_138544 1,12 0,010

(Drosophila)

193 Mmpll matrixmetallopeptidasell BC099781 NM_012980 1,12 0,005

194 Lcat lecithincholesterolacyltransferase BC091155 NM_017024 1,12 0,007

195 Akap5 Akinase(PRKA)anchorprote»5 U67136 ¾ NM_133515 1,12 o,oca

196 Fkbp4 FK506bindingprotein4 NM_001191863 1,11 0,006

197 Jrk jerkyhomolog(mouse) BC152551 NM_001104612 1,11 0,008

198 RGD1311517 similartoRIKENcDNA9430015G10 BC083613 t 1,11 0,006 vacuolarproteinsorting28homolog

199 Vps28 BC168742 NM_001130492 t 1,11 0,009

(S.cerevisiae)

tRNA5-methylarninomethyl-2-

200 Trmu BC161991 NM_001135876 1,11 0,004 thiouridylatemethyltransferase

201 Dtx3 deltexhomolog3(Drosophila) NM_001191989 1,11 0,004

202 Mir3588 microRNAmir-3588 NR_037386 1,10 0,004

203 Calhml calciumhomeostasismodulatorl N _001109168 ::,t ' . 1,10 0,006 fascinhomolog2,actin-

204 Fscn2 bundlingprotein,retinal NM_001107072 1,10 0,006

(Strongylocentrotuspurpuratus)

205 Robol roundabouthomologl(Drosophila) AF041082 NM_022188 1,10 0,004

206 RGD1310212 similartoKIAAllll-likeprotein NM_001106765 1,10 0,002

207 Homer3 homerhomolog3(Drosophila) AB020879 NM_053310 1,10 0,004

208 Neu2 sialidase2(cytosolicsialidase) D16300 NM_017130 1,10 0,007

209 Wispl WNTlinduciblesignalingpathwayproteinl AF228049 NM_031716 1,10 0,010

210 Tdrd5 tudordomaincontaining5 BC168218 ΝΜ_001134739/// · 1,10 0,009

211 Asgr2 asialoglycoproteinreceptor2 AF230645 NM_017189 t 1,10 0,007

212 Speg SPEGcomplexlocus U57097 NM_001108802/// - 1,10 0,009

213 RGD1306782 similartoRIKENcDNA1700029Pll XM_001077385 1,10 0,009

214 Mecom MDSlandEVIlcomplexlocus 1,10 0,002

2ί£ " : Col20al collagen, typeXX,alphal XM p01QS8¾3g ? 1,10 0,006 tyrosine3-monooxygenase/tryptophan5-

216 Ywhae monooxygenaseactivation protein, M84416 NM. _031603 t 1,09 0,003 epsilonpolypeptide

217 Acotl2 acyl-CoAthioesterasel2 AB040609 NM _130747 1,09 0,004

218 LOC100363401 celldivisioncycle26-like XM_ 002729269 1,08 0,007

219 Cldnll claudinll BC070927 NM 053457 1,08 0,008

220 DSTN Destrin CAG46754.1 CR541956.1 t 1,23 0,040

22iE EN03 Beta-Enolase 3 P13929.5 1,12 0,040

222 RN F165/Ark2C E3 ubiquitin protein ligase Arkadia N P_001317260.1 t 1,18 0,040

Ring-fingerand transmembranedomain

223 RN FT2 Q96EX2.2 t 1,33 0,040 containingprotein 2

SlOO calcium binding protein in amniotic

224 S100A12 P80511.2 t 1,46 0,040 fluid

ankyrin repeat domain containing protein

225 AN KRD34C N P 001139813.1 t 1,20 0,040

34C

226 C9 Complement 9 AAB51328.1 t 1,15 0,040

227 CIS Complement Cls subcomponent P09871.1 t 1,22 0,040

228 CD34 Hematopoietic progenitor cell antigen 34 P28906.2 t 1,19 0,040

Tumor necrosis factor receptor

229 CD40 P25942.1 + 1,32 0,040 superfamily member 5

230 CFB Complement factor B P00751.2 V 1,16 0,040 cA MP -activated global trnascriptional

231 CRP P0ACJ8.1 t 1,21 0,040 regulator

232 CXCL1 CXC motif chemokine 1 P09341.1 t 1,45 0,040

233 HIF1A Hypoxia-inducible factor 1-alpha Q16665.1 t 1,23 0,040

234 IL10 Interleukin 10 CAG46790.1 t 1,17 0,040

235 IL6 Interleukin 6 P05231.1 t 1,22 0,040

Potassium voltage-gated channel

236 KCNA2 P16389.2 1,20 0,040 subfamily A member 2

237 Ly6/PLAUR domain-containing protein 1 Q8N2G4.2 * 1,23 0,040

238 OMG Oligodendrocyte-myelin glycoprotein P23515.2 * 1,15 0,040

239 RBF0X1 RNA-binding protein fox-1 homolog 1 Q9JJ43.3 1,24 0,040

240 RBFOX2 RNA-binding protein fox-1 homolog 2 043251.3 * 1,32 0,040

241 TARDBP / TDP-43 TAR-DNA binding protein 43 Q13148.1 t 1,36 0,040

242 CAPN1 Calpain 1 catalytic subunit P07384.1 t 1,26 0,040

σ)

Table 2

target (rat) primer forward 5'-3' primer reverse 5'-3' cDNA dilutior add-ons LM TG

GCCCTCTCTGGCTGTTACTG CTGATGGCCTGGTTGTCTTT

Ifng 1 :25 5% Factor Q

(SEQ ID NO: 31 ) concentration: 50 nM (SEQ ID NO: 32) concentration: 300 nM

CTCTCTCCTCCTGCTTGTGG CACAGATTTGCCTGCCTTTT

Ccl4 1 :25 5% Factor Q

(SEQ ID NO: 33) concentration: 900 nM (SEQ ID NO: 34) concentration: 50 nM

GAAACATGGAGGGTGCAAGT CACTGCGACAAAGACTGGAA

Il13ra1 1 :25 5% Factor Q

(SEQ ID NO: 35) concentration: 300 nM (SEQ ID NO: 36) concentration: 300 nM

AGCCTCTTAACAGCACATCCT TGAAATTCATATTCTGTGAATGGTCT

1 :25 no

(SEQ ID NO: 37) concentration: 300 nM (SEQ ID NO: 38) concentration: 300 nM

GAGAGCTGGTTGTGGACCAT CAGTCGCAGGTCAATGAAGA

C3 1 :25 5% Factor Q

(SEQ ID NO: 39) concentration: 50 nM (SEQ ID NO: 40) concentration: 300 nM

GCAGGAAATACAACGCCACT TCTTCCAACAGCTCCGATTT

Slc27a2 1 :25 5% Factor Q

(SEQ ID NO: 41 ) concentration: 50 nM (SEQ ID NO: 42) concentration: 300 nM

GAGAGGGAAATCGTGCGTG CATGGATGCCACAGGATTCC dependent on

Actin no

(SEQ ID NO: 43) concentration: 300 nM (SEQ ID NO: 44) concentration: 300 nM target

I

target (human) primer forward 5'-3' primer reverse 5'-3' cDNA dilutior add-ons HC SP UR CTRL SCZ

GGCTGTAGATTCTCGAGTGCGG CGCTACATCTGAATGACCTGC

IFNG 1 :50 no

(SEQ ID NO: 1 ) concentration: 300 nM (SEQ ID NO: 2) concentration: 300 nM

CTGAGTTCTGCAGCCTCACC CTGGGATCAGCACAGACTTGC CCL4 1 :10 no

(SEQ ID NO: 3) concentration: 300 nM (SEQ ID NO: 4) concentration: 300 nM

CCACCCGAGGGAGCCAGCTC CTTCTGGGGGTGAGATGC IL13RA1 1 :10 no

(SEQ ID NO: 5) concentration: 50 nM (SEQ ID NO: 6) concentration: 50 nM

GACTGTGGCCTGCACCTG GACAGCAGTAACCTTGGCTGTG IL12RB2 1 :10 no

(SEQ ID NO: 7) concentration: 300 nM (SEQ ID NO: 8) concentration: 300 nM

GCTCCAGACACAGATGACCTG GCGTAGACCTTGACTGCTCCAG

C3 1 :10

(SEQ ID NO: 9) concentration: 50 nM (SEQ ID NO: 10) concentration: 300 nM

CCCTCACGGCCTTTGTTCTC GCCAGAGCATAGCCAGCAATG

C3 1 :10 no

(SEQ ID NO: 11 ) (SEQ ID NO: 12) concentration: 300 nM

CCACGACAGAGTTGGAGATAC GGCCTTGCATAACTAGGTAGG SLC27A2 1 :25 no

(SEQ ID NO: 13) concentration: 300 nM (SEQ ID NO: 14) concentration: 300 nM

GAGTTCTGGCTGGCTTGTGAAG GGCTGTAGATTCTCGAGTGCGG RGS1 1 :10 no

(SEQ ID NO: 15) concentration: 300 nM (SEQ ID NO: 16) concentration: 300 nM

GAATGTCTTGGGATGGCAGTG JAK2 1 :10 no

(SEQ ID NO: 17) concentration: 300 nM (SEQ ID NO: 18) concentration: 300 nM

GAGACATCCG CCCCTACAAG CCR5 1 :10

(SEQ ID NO: 19) concentration: 300 nM (SEQ ID NO: 20) concentration: 300 nM

GTGTCCTATGAGTCTGCTGG FPR2 1 :10

(SEQ ID NO: 21 ) concentration: 300 nM (SEQ ID NO: 22) concentration: 300 nM

CCCGCTTGTCTCAACCACC NKG7 1 :10

(SEQ ID NO: 23) concentration: 300 nM (SEQ ID NO: 24) concentration: 300 nM

GAATGCGGGCTTTGAAGACTG KMO 1 :10

(SEQ ID NO: 25) concentration: 300 nM (SEQ ID NO: 26) concentration: 300 nM

GACCAGAGGTAACACGGCAG SERPINB1 1 :10

(SEQ ID NO: 27) concentration: 300 nM (SEQ ID NO: 28) concentration: 300 nM

GACCACGATCCTCTACAAGC TCCCACACAGTGAAGCTGATG dependent on

ARF1

(SEQ ID NO: 29) concentration: 300 nM (SEQ ID NO: 30) concentration: 300 nM target