ArticlePDF AvailableLiterature Review

Comprehending depression through proteomics

Authors:
TheInternationalJournalofNeuropsychopharmacology
http://journals.cambridge.org/PNP
AdditionalservicesforTheInternationalJournalof
Neuropsychopharmacology:
Emailalerts:Clickhere
Subscriptions:Clickhere
Commercialreprints:Clickhere
Termsofuse:Clickhere
Comprehendingdepressionthroughproteomics
DanielMartinsdeSouza
TheInternationalJournalofNeuropsychopharmacology/Volume15/Issue10/November2012,pp13731374
DOI:10.1017/S146114571200034X,Publishedonline:17April2012
Linktothisarticle:http://journals.cambridge.org/abstract_S146114571200034X
Howtocitethisarticle:
DanielMartinsdeSouza(2012).Comprehendingdepressionthroughproteomics.TheInternationalJournalof
Neuropsychopharmacology,15,pp13731374doi:10.1017/S146114571200034X
RequestPermissions:Clickhere
Downloadedfromhttp://journals.cambridge.org/PNP,IPaddress:145.253.118.83on08Oct2012
Comprehending depression through proteomics
Daniel Martins-de-Souza
Max Planck Institute of Psychiatry, Munich, Germany
Received 14 March 2012; Reviewed 14 March 2012; Revised 15 March 2012 ; Accepted 15 March 2012 ;
First published online 17 April 2012
Even being a severe neuropsychiatric disorder affect-
ing approximately 10 % of the population worldwide,
the molecular mechanisms underlying depression,
unipolar depression, clinical depression or major de-
pressive disorder (MDD) are still to be comprehended
(Fava & Kendler, 2000). One of the main challenges is
to understand how the multifactorial characteristics of
MDD which come from genetic or metabolic predis-
position triggered by environmental factors are con-
nected to each other. But in order to understand their
interaction, it is necessary to characterize each one of
these factors.
Key molecular components of MDD such as gen-
etics (Goltser-Dubner et al. 2010), gene expression
(Mehta et al. 2010) and a range of studies in preclinical
models (Muller & Holsboer, 2006) have been inves-
tigated. But one of the key components of the mol-
ecular basis of MDD the proteome has just started
to be characterized in human tissues. Proteome was
defined around 15 years ago as the total set of ex-
pressed proteins by a cell, tissue or organism at a given
time under a determined condition (Wilkins et al.
1996). This definition led to the science currently
known as proteomics that also included the study of
protein–protein interactions and post-translational
modifications. The identification of sets of differen-
tially expressed proteins or even the differential post-
translational modifications of proteins while studying
samples from MDD patients may lead to two main
outcomes: (1) revealing proteins, and consequently
metabolites and biochemical pathways which may
shed light on the comprehension of pathological
states; (2) revealing biomarker candidates with
potential for therapeutic applications (Martins-
de-Souza et al. 2010). Non-hypothesis-driven pro-
teomic applications offer different insights of MDD by
complementing those provided by the standard tar-
geted methodologies.
Regarding the molecular comprehension of MDD,
post-mortem dorsolateral prefrontal cortex from 24
MDD patients and 12 matched controls were analysed
using label-free proteomics. Distinct proteome finger-
prints between MDD and controls associated with
energy metabolism and synaptic function were ob-
served. Additionally, differential proteome profiles in
MDD with and without psychosis were assessed
showing a marked overlap to proteome changes seen
in schizophrenia brains (Martins-de-Souza et al.
2012a). Complementarily, a shotgun proteomics ap-
proach has been used also to identify differences in the
phosphorylation of brain proteins compared to con-
trols. The majority of phosphorylation differences
were associated with synaptic transmission and cellu-
lar architecture (Martins-de-Souza et al. 2012b). Other
studies in brain tissue from MDD patients were per-
formed previously using anterior cingulate cortex
(ACC) and frontal cortex (FC) (Beasley et al. 2006 ;
Johnston-Wilson et al. 2000). Both showed the altered
expression of carbonic anhydrase (CA2) and Aldolase
C (ALDOC), suggesting effects on energy metabolism.
Additionally, dihydropyrimidinase-related protein-2
(DPYSL2) was also found common to both studies,
but down-regulated in FC and up-regulated in ACC.
However, these studies focused on schizophrenia
using MDD samples as controls for specificity.
Considering the lack of biochemical markers for
MDD that could aid, for instance in patient stratifi-
cation, the proteome of the cerebrospinal fluid
(CSF) from 12 MDD patients and 12 controls were
evaluated recently (Ditzen et al. 2012). Using
traditional proteomic methodologies such as two-
dimensional gel electrophoresis followed by matrix-
assisted laser desorption ionization–time-of-flight–
mass spectrometry (MALDI-TOF-MS), 11 proteins
were observed as differentially expressed including
Address for correspondence : Dr D. Martins-de-Souza, Max Planck
Institute of Psychiatry, Proteomics and Biomarkers, Kraepelinstr.
2, D-80804 Munich, Germany.
Tel. :+49 89 30622 630 Fax :+49 89 30622 200
Email : martins@mpipsykl.mpg.de
See Xu et al. (2012). Comparative proteomic analysis of plasma
from major depressive patients : identification of proteins
associated with lipid metabolism and immunoregulation
(doi :S1461145712000302).
International Journal of Neuropsychopharmacology (2012), 15, 1373–1374. fCINP 2012
doi:10.1017/S146114571200034X
FOCUS
protein players in neuroprotection, neurodevelop-
ment and sleep regulation.
In their recent publication, Xu and colleagues pres-
ent a proteome analysis of the plasma from 21 first-
onset drug-naive MDD patients which were compared
to 21 controls using shotgun proteomics for protein
identification and isobaric tags for relative and absol-
ute quantitation (iTRAQ) for protein quantitation (Xu
et al. 2012). The study focuses on the understanding of
MDD as a systemic disorder, but also mentions the
biomarker potential of these candidates. By using this
proteomic hypothesis-free approach and a further
validation by Western blot and enzyme-linked im-
munoadsorbent assay (ELISA), Xu et al. identified 94
proteins and found nine of those differentially ex-
pressed in MDD patients. These are mostly involved
with lipid metabolism and the immune system and
authors suggest that these are processes that might be
involved in the early stages of MDD pathophysiology.
Proteomic studies mentioned here have indeed
provided understanding of the molecular aspects of
MDD, exploring not only the role of the proteins, but
also the role of biochemical pathways and related
metabolites. However, only the report from Ditzen
et al. (2012) in MDD CSF explored reasonably the dif-
ferentially expressed proteins as biomarkers candi-
dates for MDD. This aspect of proteomic studies is still
open to exploration in order to reveal proteins that can
be useful not only in diagnosis, but in patient stratifi-
cation in accordance with the different types of MDD
(i.e. atypical depression and psychotic depression),
prognosis, treatment monitoring and response, and
potential drug targets.
Acknowledgements
I sincerely thank all tissue donors and their families for
comprehending how important their consent is to
medical research and to the lives of patients. I also
thank Professor Chris W. Turck and his entire group at
the Max Planck Institute of Psychiatry as well as
Professor Peter Falkai and Dr Andrea Schmitt from
the University of Go
¨ttingen for their unconditional
support.
Statement of Interest
None.
References
Beasley CL, Pennington K, Behan A, Wait R, et al. (2006).
Proteomic analysis of the anterior cingulate cortex in the
major psychiatric disorders: evidence for disease-
associated changes. Proteomics 6, 3414–3425.
Ditzen C, Tang N, Jastorff AM, Teplytska L, et al. (2012).
Cerebrospinal fluid biomarkers for major depression
confirm relevance of associated pathophysiology.
Neuropsychopharmacology 37, 1013–1025.
Fava M, Kendler KS (2000). Major depressive disorder.
Neuron 28, 335–341.
Goltser-Dubner T, Galili-Weisstub E, Segman RH (2010).
Genetics of unipolar major depressive disorder. Israel
Journal of Psychiatry and Related Sciences 47, 72–82.
Johnston-Wilson NL, Sims CD, Hofmann JP, Anderson L,
et al. (2000). Disease-specific alterations in frontal cortex
brain proteins in schizophrenia, bipolar disorder, and
major depressive disorder. The Stanley Neuropathology
Consortium. Molecular Psychiatry 5, 142–149.
Martins-de-Souza D, Guest PC, Harris LW,
Vanattou-Saifoudine N, et al. (2012a). Identification
of proteomic signatures associated with depression
and psychotic depression in post-mortem brains
from major depression patients. Translational Psychiatry
2, e87.
Martins-de-Souza D, Guest PC, Vanattou-Saifoudine N,
Rahmoune H, et al. (2012b). Phosphoproteomic
differences in major depressive disorder postmortem
brains indicate effects on synaptic function. European
Archives of Psychiatry and Clinical Neuroscience. Published
online: 21 February 2012. doi :10.1007/s00406-012-0301-3.
Martins-de-Souza D, Harris LW, Guest PC, Turck CW, et al.
(2010). The role of proteomics in depression research.
European Archives of Psychiatry and Clinical Neuroscience 260,
499–506.
Mehta D, Menke A, Binder EB (2010). Gene expression
studies in major depression. Current Psychiatry Reports 12,
135–144.
Muller MB, Holsboer F (2006). Mice with mutations in the
HPA-system as models for symptoms of depression.
Biological Psychiatry 59, 1104–1115.
Wilkins MR, Pasquali C, Appel RD, Ou K, et al. (1996).
From proteins to proteomes : large scale protein
identification by two-dimensional electrophoresis and
amino acid analysis. Biotechnology (NY)14, 61–65.
Xu HB, Zhang RF, Luo D, Zhou Y, et al. (2012).
Comparative proteomic analysis of plasma from major
depressive patients: identification of proteins associated
with lipid metabolism and immunoregulation.
International Journal of Neuropsychopharmacology.
doi:S1461145712000302.
1374 D. Martins-de-Souza
... 15 This includes genegene and gene-environmental interactions, as well as posttranslational modifications. 16 Using proteomics may help determine disease-specific markers by identifying and assessing all of the proteins in a certain physiological condition. 17 In the proteomic approach to depression, the appropriate tissue is brain tissue or cerebrospinal fluid (CSF). ...
... In doing so, a more comprehensive understanding of the pathophysiology of depression could be established, along with the identification of a biomarker for clinical applications. 16 In this study, we compared the same patient' s proteome between the depression status before treatment and during the remission status after antidepressant treatment. Each patient's protein expression was individually analyzed to identify biomarker candidates with accurate diagnostic ability. ...
Article
Full-text available
Currently, there are a few biological markers to aid in the diagnosis and treatment of depression. However, it is not sufficient for diagnosis. We attempted to identify differentially expressed proteins during depressive moods as putative diagnostic biomarkers by using quantitative proteomic analysis of serum. Blood samples were collected twice from five patients with major depressive disorder (MDD) at depressive status before treatment and at remission status during treatment. Samples were individually analyzed by liquid chromatography-tandem mass spectrometry for protein profiling. Differentially expressed proteins were analyzed by label-free quantification. Enzyme-linked immunosorbent assay (ELISA) results and receiver-operating characteristic (ROC) curves were used to validate the differentially expressed proteins. For validation, 8 patients with MDD including 3 additional patients and 8 matched normal controls were analyzed. The quantitative proteomic studies identified 10 proteins that were consistently upregulated or downregulated in 5 MDD patients. ELISA yielded results consistent with the proteomic analysis for 3 proteins. Expression levels were significantly different between normal controls and MDD patients. The 3 proteins were ceruloplasmin, inter-alpha-trypsin inhibitor heavy chain H4 and complement component 1qC, which were upregulated during the depressive status. The depressive status could be distinguished from the euthymic status from the ROC curves for these proteins, and this discrimination was enhanced when all 3 proteins were analyzed together. This is the first proteomic study in MDD patients to compare intra-individual differences dependent on mood. This technique could be a useful approach to identify MDD biomarkers, but requires additional proteomic studies for validation.
... It is also a multifactorial disorder. Studies suggest that MDD is caused by a combination of genetic predisposition (~35%) and environmental factors [181]. Our understanding of MDD's pathophysiology remains incomplete. ...
Article
Full-text available
In the last two decades, our knowledge of synaptic proteomes and their relationship to normal brain function and neuropsychiatric disorders has been expanding rapidly through the use of more powerful neuroproteomic approaches. However, mass spectrometry (MS)-based neuroproteomic studies of synapses still require cell-type, spatial, and temporal proteome information. With the advancement of sample preparation and MS techniques, we have just begun to identify and understand proteomes within a given cell type, subcellular compartment, and cell-type-specific synapse. Here, we review the progress and limitations of MS-based neuroproteomics of synapses in the mammalian CNS and highlight the recent applications of these approaches in studying neuropsychiatric disorders such as major depressive disorder and substance use disorders. Combining neuroproteomic findings with other omics studies can generate an in-depth, comprehensive map of synaptic proteomes and possibly identify new therapeutic targets and biomarkers for several central nervous system disorders.
... It is also a multifactorial disorder. Studies suggest that MDD is caused by a combination of genetic predisposition (~35%) and environmental factors [180]. Our understanding of 12 MDD pathophysiology remains incomplete. ...
Preprint
Full-text available
In the last two decades, our knowledge of synaptic proteomes and their relationship to normal brain function and neuropsychiatric disorders has been expanding rapidly through the use of more powerful neuroproteomic approaches. However, mass spectrometry (MS) based neuroproteomic studies of synapses still need cell-type, spatial, and temporal proteome information. With the advancement of sample preparation and MS techniques, we have just begun to identify and understand proteomes within a given cell type, subcellular compartment, and cell-type-specific synapse. Here, we review the progress and limitations of MS-based neuroproteomics of synapses in the mammalian CNS and highlight the recent applications of these approaches in studying neuropsychiatric disorders such as major depressive disorder and substance use disorders. Combining neuroproteomic findings with other omics studies can generate an in-depth, comprehensive map of synaptic proteomes and possibly identify new therapeutic targets and biomarkers of several central nervous system disorders.
... It is also a multifactorial disorder. Studies suggest that MDD is caused by a combination of genetic predisposition (~35%) and environmental factors [163]. Our understanding of MDD pathophysiology remains incomplete. ...
Preprint
Full-text available
In the last two decades, our knowledge of synaptic proteomes and their relationship to normal brain function and neuropsychiatric disorders has been expanding rapidly through the use of more powerful neuroproteomics approaches. However, mass spectrometry (MS) based neuroproteomics studies of synapses still need cell-type, spatial, and temporal proteome information. With the advancement of sample preparation and MS techniques, we have just begun to identify and understand proteomes within a given cell type, subcellular compartment, and cell-type-specific synapse. Here, we review the progress and limitations of MS-based neuroproteomics of synapses and highlight the recent applications of these approaches in studying neuropsychiatric disorders such as major depressive disorder and substance use disorders. Combining neuroproteomics findings with other omics studies can generate an in-depth, comprehensive map of synaptic proteomes and possibly identify new therapeutic targets and biomarkers of several central nervous system disorders.
... Genomics and the novel omics-based technologies may promote the understanding of gene and protein functions, and offer together a more comprehensive understanding into the pathobiological core of psychiatric illnesses, as well as the possibility of identifying novel diagnostic, therapeutic, and disease progression biomarkers [33,34]. Nevertheless, information obtained through proteomics, metabolomics, epigenetics, and lipidomics-based techniques were complementary rather than opposing, contributing to a better understanding of the biological basis of many diseases such as psychiatric disorders [35][36][37]. ...
Article
Introduction: Currently, the diagnosis of psychiatric illnesses is based upon DSM-5 criteria. Although endophenotype-specificity for a particular disorder is discussed, the identification of objective biomarkers is ongoing for aiding diagnosis, prognosis, or clinical response to treatment. We need to improve the understanding of the biological abnormalities in psychiatric illnesses across conventional diagnostic boundaries. The present review investigates the innovative post-genomic knowledge used for psychiatric illness diagnostics and treatment response, with a particular focus on proteomics. Areas covered: This review underlines the contribution that psychiatric innovative biomarkers have reached in relation to diagnosis and theragnosis of psychiatric illnesses. Furthermore, it encompasses a reliable representation of their involvement in disease through proteomics, metabolomics/pharmacometabolomics and lipidomics techniques, including the possible role that gut microbiota and CYP2D6 polimorphisms may play in psychiatric illnesses. Expert opinion: Etiologic heterogeneity, variable expressivity, and epigenetics may impact clinical manifestations, making it difficult for a single measurement to be pathognomonic for multifaceted psychiatric disorders. Academic, industry, or government's partnerships may successfully identify and validate new biomarkers so that unfailing clinical tests can be developed. Proteomics, metabolomics, and lipidomics techniques are considered to be helpful tools beyond neuroimaging and neuropsychology for the phenotypic characterization of brain diseases.
... Although our findings reinforce the notion that the progression of brain abnormalities may occur both among non-remitted subjects with schizophrenia and among those with affective psychoses, the distinct location of such findings in the FESZ versus FEAP groups, along with the longitudinal GM differences that emerged when we directly compared the non-remitting FESZ and FEAP patients (see Supplementary Material), supports the view that different mechanisms may underlie the progression of brain changes in these disorders (Demjaha et al. 2012). These region-specific abnormalities are in line with postmortem studies on schizophrenia ( Schmitt et al. 2011Schmitt et al. , 2013 and mood disorders (Martins-de-Souza, 2012a). Furthermore, they highlight how important in vivo neuroimaging studies may be to identify which brain structures should be targeted in further contemporary post-mortem proteomic investigations into the expression of proteins that regulate neurodevelopmental processes, glial cell functioning and other biochemical pathways of potential interest to models of disease progression for schizophrenia, affective psychoses, or both ( Honea et al. 2005;Martins-de-Souza, 2012a, b). ...
Article
Background: Magnetic resonance imaging (MRI) studies have shown that brain abnormalities in psychosis might be progressive during the first years of illness. We sought to determine whether first-episode psychosis (FEP) subjects show progressive regional grey matter (GM) changes compared with controls, and whether those changes are associated with diagnosis, illness course or antipsychotic (AP) use. Method: Thirty-two subjects with first-episode schizophrenia-spectrum disorders (FESZ), 24 patients with first-episode affective psychoses (FEAP) and 34 controls recruited using a population-based design underwent structural MRI scanning at baseline and at a 5-year follow-up. Regional GM volumes were assessed with voxel-based morphometry (VBM). Patients were treated at community settings, and about half of them remained mainly untreated. Results: No significant progressive changes in GM regional volumes were observed in either the FESZ or FEAP group overall. However, FESZ subjects with a non-remitting course showed GM decrements in the left superior temporal gyrus (STG) and insula relative to remitted FESZ subjects. Non-remitted FEAP subjects exhibited a GM decrease in the dorsolateral prefrontal cortex (DLPFC) bilaterally in comparison to remitted FEAP subjects. Among FESZ subjects, AP use was associated with regional GM decrements in the right insula and increments in the cerebellum. Conclusions: Our results suggest that the progression of brain abnormalities in FEP subjects is restricted to those with a poor outcome and differs between diagnosis subgroups. AP intake is associated with a different pattern of GM reductions over time.
... Stimulated by the disappointing results of the GWAS for antidepressant response, researchers have started to explore the potential of gene expression and proteomics as sources of predictive biosignatures most recently. As the downstream effects of different predictors of antidepressant response-be they genetic and/or environmental-should ultimately be mediated by changes in gene function, investigating predictors and underlying biological factors associated with response by means of large-scale "omics" approaches (gene expression, i.e., transcriptomics, proteomics, and metabolomics) may provide valuable information [49,50]. The regulation of gene expression has been proposed as one molecular mechanism that could mediate stable adaptations and maladaptations in the brain, as supposed to be involved in both the pathophysiology of depressive disorder as well as in the mechanism of action by which antidepressant treatment works. ...
Article
Full-text available
Major depression, affecting an estimated 350 million people worldwide, poses a serious social and economic threat to modern societies. There are currently two major problems calling for innovative research approaches, namely, the absence of biomarkers predicting antidepressant response and the lack of conceptually novel antidepressant compounds. Both, biomarker predicting a priori whether an individual patient will respond to the treatment of choice as well as an early distinction of responders and nonresponders during antidepressant therapy can have a significant impact on improving this situation. Biosignatures predicting antidepressant response a priori or early in treatment would enable an evidence-based decision making on available treatment options. However, research to date does not identify any biologic or genetic predictors of sufficient clinical utility to inform the selection of specific antidepressant compound for an individual patient. In this review, we propose an optimized translational research strategy to overcome some of the major limitations in biomarker discovery. We are confident that early transfer and integration of data between both species, ideally leading to mutual supportive evidence from both preclinical and clinical studies, are most suitable to address some of the obstacles of current depression research.
Article
Full-text available
Omics technologies emerged as complementary strategies to genomics in the attempt to understand human illnesses. In general, proteomics technologies emerged earlier than those of metabolomics for major depressive disorder (MDD) research, but both are driven by the identification of proteins and/or metabolites that can delineate a comprehensive characterization of MDD's molecular mechanisms, as well as lead to the identification of biomarker candidates of all types-prognosis, diagnosis, treatment, and patient stratification. Also, one can explore protein and metabolite interactomes in order to pinpoint additional molecules associated with the disease that had not been picked up initially. Here, results and methodological aspects of MDD research using proteomics, metabolomics, and protein interactomics are reviewed, focusing on human samples.
Article
Fatigue is the most distressing side effect of radiation therapy, and its progression etiology is unknown. This study describes proteome changes from sera of fatigued men with non-metastatic prostate cancer receiving external beam radiation therapy (EBRT). Fatigue scores, measured by the Functional Assessment of Chronic Illness Therapy-Fatigue, and serum were collected from 12 subjects at baseline (before EBRT) and at midpoint (Day 21) of EBRT. Depleted sera from both time points were analyzed using two-dimensional difference gel electrophoresis, and up/down regulated proteins were identified using liquid chromatography-tandem mass spectrometry. Western blot analyses confirmed the protein changes observed. Results showed that apolipoprotein (Apo)A1, ApoE, and transthyretin (TTR) consistently changed from baseline (Day 0) to midpoint (Day 21). The mean ApoE level of subjects with high change in fatigue (HF: n = 9) increased significantly from baseline to midpoint and were higher than in subjects with no change in fatigue. The mean ApoA1 level was higher in HF subjects at baseline and at midpoint than in no fatigue subjects at both time points. The mean TTR level of no fatigue subjects was higher at baseline and midpoint than in HF subjects. These ApoE, ApoA1, and TTR results may assist in understanding pathways that can explain fatigue progression etiology in this clinical population.
Article
Full-text available
Major depressive disorder (MDD) is a leading cause of disability worldwide and results tragically in the loss of almost one million lives in Western societies every year. This is due to poor understanding of the disease pathophysiology and lack of empirical medical tests for accurate diagnosis or for guiding antidepressant treatment strategies. Here, we have used shotgun proteomics in the analysis of post-mortem dorsolateral prefrontal cortex brain tissue from 24 MDD patients and 12 matched controls. Brain proteomes were pre-fractionated by gel electrophoresis and further analyzed by shotgun data-independent label-free liquid chromatography-mass spectrometry. This led to identification of distinct proteome fingerprints between MDD and control subjects. Some of these differences were validated by Western blot or selected reaction monitoring mass spectrometry. This included proteins associated with energy metabolism and synaptic function and we also found changes in the histidine triad nucleotide-binding protein 1 (HINT1), which has been implicated recently in regulation of mood and behavior. We also found differential proteome profiles in MDD with (n=11) and without (n=12) psychosis. Interestingly, the psychosis fingerprint showed a marked overlap to changes seen in the brain proteome of schizophrenia patients. These findings suggest that it may be possible to contribute to the disease understanding by distinguishing different subtypes of MDD based on distinct brain proteomic profiles.
Article
Full-text available
Major depressive disorder is a common neuropsychiatric disorder contributing to several socio-economic burdens including disability and suicide. As the underlying pathophysiology of major depressive disorder remains unclear, no objective test is yet available for aiding diagnosis or monitoring disease progression. To contribute to a better understanding of its pathogenesis, a comparative proteomic study was performed to identify proteins differentially expressed in plasma samples obtained from first-onset, treatment-naive depressed patients as compared to healthy controls. Samples from the two groups were immunodepleted of seven high-abundance proteins, labelled with isobaric tags for relative and absolute quantitation and then analysed by multi-dimensional liquid chromatography-tandem mass spectrometry. The proteomic results were further validated by immunoblotting or enzyme-linked immunoadsorbent assays and analysed with the MetaCore database. The results demonstrate that the functions of the altered proteins are primarily involved in lipid metabolism and immunoregulation. These findings suggest that early perturbation of lipid metabolism and immunoregulation may be involved in the pathophysiology of major depressive disorder.
Article
Full-text available
There is still a lack in the molecular comprehension of major depressive disorder (MDD) although this condition affects approximately 10% of the world population. Protein phosphorylation is a posttranslational modification that regulates approximately one-third of the human proteins involved in a range of cellular and biological processes such as cellular signaling. Whereas phosphoproteome studies have been carried out extensively in cancer research, few such investigations have been carried out in studies of psychiatric disorders. Here, we present a comparative phosphoproteome analysis of postmortem dorsolateral prefrontal cortex tissues from 24 MDD patients and 12 control donors. Tissue extracts were analyzed using liquid chromatography mass spectrometry in a data-independent manner (LC-MSE). Our analyses resulted in the identification of 5,195 phosphopeptides, corresponding to 802 non-redundant proteins. Ninety of these proteins showed differential levels of phosphorylation in tissues from MDD subjects compared to controls, being 20 differentially phosphorylated in at least 2 peptides. The majority of these phosphorylated proteins were associated with synaptic transmission and cellular architecture not only pointing out potential biomarker candidates but mainly shedding light to the comprehension of MDD pathobiology. Electronic supplementary material The online version of this article (doi:10.1007/s00406-012-0301-3) contains supplementary material, which is available to authorized users.
Article
Full-text available
Individual characteristics of pathophysiology and course of depressive episodes are at present not considered in diagnostics. There are no biological markers available that can assist in categorizing subtypes of depression and detecting molecular variances related to disease-causing mechanisms between depressed patients. Identification of such differences is important to create patient subgroups, which will benefit from medications that specifically target the pathophysiology underlying their clinical condition. To detect characteristic biological markers for major depression, we analyzed the cerebrospinal fluid (CSF) proteome of depressed vs control persons, using two-dimensional polyacrylamide gel electrophoresis and time-of-flight (TOF) mass spectrometry peptide profiling. Proteins of interest were identified by matrix-assisted laser desorption ionization TOF mass spectrometry (MALDI-TOF-MS). Validation of protein markers was performed by immunoblotting. We found 11 proteins and 144 peptide features that differed significantly between CSF from depressed patients and controls. In addition, we detected differences in the phosphorylation pattern of several CSF proteins. A subset of the differentially expressed proteins implicated in brain metabolism or central nervous system disease was validated by immunoblotting. The identified proteins are involved in neuroprotection and neuronal development, sleep regulation, and amyloid plaque deposition in the aging brain. This is one of the first hypothesis-free studies that identify characteristic protein expression differences in CSF of depressed patients. Proteomic approaches represent a powerful tool for the identification of disease markers for subgroups of patients with major depression.
Article
Full-text available
The dramatic technical advances in methods to measure gene expression on a genome-wide level thus far have not been paralleled by breakthrough discoveries in psychiatric disorders-including major depression (MD)-using these hypothesis-free approaches. In this review, we first describe the methodologic advances made in gene expression analysis, from quantitative polymerase chain reaction to next-generation sequencing. We then discuss issues in gene expression experiments specific to MD, ranging from the choice of target tissues to the characterization of the case group. We provide a synopsis of the gene expression studies published thus far for MD, with a focus on studies using mRNA microarray methods. Finally, we discuss possible new strategies for the gene expression studies in MD that circumvent some of the addressed issues.
Article
Introduction Major depressive disorder (MDD) is estimated by the World Health Organization to be a leading cause for loss of disability-adjusted life years. In the USA, the 2001 National Comorbidity Survey showed overall lifetime prevalence estimates for MDD of 16.2% (Kessler et al., 2003). Over 50% of respondents with MDD received some type of treatment in the 12 months before their interview. However, only about one-fifth of patients received “adequate” treatment during that time, indicating that more efforts are required at improving access to and utilization of care. Even for patients who are treated, pharmacotherapy is not uniformly effective. Many episodes do not completely respond to initial antidepressant, response may be delayed for weeks or months, and residual symptoms may cause significant morbidity. In this paper, we review the evidence for first-line treatment of MDD, and strategies for patients with treatment-resistant depression. Methods For the purposes of this review, we searched MEDLINE and PsycINFO for controlled trials published in English between January 1981 and October 2010, in which adults with MDD were randomly assigned to receive medication, placebo, or active comparator drugs, and for all meta-analyses of pharmacotherapy for MDD. The number of published randomized placebo-controlled trials (RCTs) of antidepressants for the acute treatment of MDD is too vast to allow discussion of each study. In order to recommend first-line treatment for MDD, we examined meta-analyses of RCTs of antidepressant drugs for MDD as a statistical means of weighing the relative efficacy of antidepressants that have not been compared directly. Because placebo response rates in antidepressant trials tend to be high, many positive trials have small effect sizes, and meta-analysis of multiple trials may be used to determine whether drug–placebo differences are meaningful.
Article
Separation and identification of proteins by two-dimensional (2-D) electrophoresis can be used for protein-based gene expression analysis. In this report single protein spots, from polyvinylidene difluoride blots of micropreparative E. coli 2-D gels, were rapidly and economically identified by matching their amino acid composition, estimated pI and molecular weight against all E. coli entries in the SWISS-PROT database. Thirty proteins from an E. coli 2-D map were analyzed and identities assigned. Three of the proteins were unknown. By protein sequencing analysis, 20 of the 27 proteins were correctly identified. Importantly, correct identifications showed unambiguous "correct" score patterns. While incorrect protein identifications also showed distinctive score patterns, indicating that protein must be identified by other means. These techniques allow large-scale screening of the protein complement of simple organisms, or tissues in normal and disease states. The computer program described here is accessible via the World Wide Web at URL address (http://expasy.hcuge.ch/).
Article
Severe psychiatric disorders such as schizophrenia, bipolar disorder and major depressive disorder are brain diseases of unknown origin. No biological marker has been documented at the pathological, cellular, or molecular level, suggesting that a number of complex but subtle changes underlie these illnesses. We have used proteomic technology to survey postmortem tissue to identify changes linked to the various diseases. Proteomics uses two-dimensional gel electrophoresis and mass spectrometric sequencing of proteins to allow the comparison of subsets of expressed proteins among a large number of samples. This form of analysis was combined with a multivariate statistical model to study changes in protein levels in 89 frontal cortices obtained postmortem from individuals with schizophrenia, bipolar disorder, major depressive disorder, and non-psychiatric controls. We identified eight protein species that display disease-specific alterations in level in the frontal cortex. Six show decreases compared with the non-psychiatric controls for one or more diseases. Four of these are forms of glial fibrillary acidic protein (GFAP), one is dihydropyrimidinase-related protein 2, and the sixth is ubiquinone cytochrome c reductase core protein 1. Two spots, carbonic anhydrase 1 and fructose biphosphate aldolase C, show increase in one or more diseases compared to controls. Proteomic analysis may identify novel pathogenic mechanisms of human neuropsychiatric diseases.
Article
Abnormalities of the anterior cingulate cortex have previously been described in schizophrenia, major depressive disorder and bipolar disorder. In this study 2-DE was performed followed by mass spectrometric sequencing to identify disease-specific protein changes within the anterior cingulate cortex in these psychiatric disorders. The 2-DE system comprised IPGs 4–7 and 6–9 in the first, IEF dimension and SDS-PAGE in the second dimension. Resultant protein spots were compared between control and disease groups. Statistical analysis indicated that 35 spots were differentially expressed in one or more groups. Proteins comprising 26 of these spots were identified by mass spectroscopy. These represented 19 distinct proteins; aconitate hydratase, malate dehydrogenase, fructose bisphosphate aldolase A, ATP synthase, succinyl CoA ketoacid transferase, carbonic anhydrase, α- and β-tubulin, dihydropyrimidinase-related protein-1 and -2, neuronal protein 25, trypsin precursor, glutamate dehydrogenase, glutamine synthetase, sorcin, vacuolar ATPase, creatine kinase, albumin and guanine nucleotide binding protein β subunit. All but three of these proteins have previously been associated with the major psychiatric disorders. These findings provide support for the view that cytoskeletal and mitochondrial dysfunction are important components of the neuropathology of the major psychiatric disorders.
Article
Major depressive disorder (MDD) is a heterogeneous, highly prevalent, and moderately heritable disorder. A complex and diverse genetic-environmental interplay converges to set apart a significant minority that is susceptible to MDD, from among those who experience shorter lived and less recurrent intensive and incapacitating forms of sadness. The major technological advances of deciphering the human genome reference sequence and its common gene variations are beginning to allow cost effective genetic studies of unprecedented scale, applying increasingly denser genome wide mapping to increasingly larger case control samples. This effort is now at the initial stages of unraveling the genetic architecture of several complex phenotypes. Despite a tardy beginning, MDD genetic research is maturing from modest scale candidate gene association studies to include family-based linkage studies, and will soon allow genome wide case control association studies. Replicated risk conferring gene variants discovered so far exert a modest effect size that appears to contribute to overt phenotype expression in the context of a highly intricate concert of interrelated epigenetic and epistatic modifiers. The unraveling of additional previously unimplicated MDD risk conferring genes, that will throw light on molecular mechanisms mediating such susceptibilities, is necessary for progressing beyond current generation monoamine modulating antidepressant drugs. The review outlines basic concepts and current progress briefly overviews major replicated gene findings that to date mostly stem from hypotheses driven candidates, and ends with a discussion of current directives, including sample size and phenotype considerations and advancement of systematic studies of the functional significance of implicated gene variants, beyond their current exploratory stage.