1
|
Smith BJ, Guest PC, Martins-de-Souza D. Maximizing Analytical Performance in Biomolecular Discovery with LC-MS: Focus on Psychiatric Disorders. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:25-46. [PMID: 38424029 DOI: 10.1146/annurev-anchem-061522-041154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
In this review, we discuss the cutting-edge developments in mass spectrometry proteomics and metabolomics that have brought improvements for the identification of new disease-based biomarkers. A special focus is placed on psychiatric disorders, for example, schizophrenia, because they are considered to be not a single disease entity but rather a spectrum of disorders with many overlapping symptoms. This review includes descriptions of various types of commonly used mass spectrometry platforms for biomarker research, as well as complementary techniques to maximize data coverage, reduce sample heterogeneity, and work around potentially confounding factors. Finally, we summarize the different statistical methods that can be used for improving data quality to aid in reliability and interpretation of proteomics findings, as well as to enhance their translatability into clinical use and generalizability to new data sets.
Collapse
Affiliation(s)
- Bradley J Smith
- 1Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, São Paulo, Brazil;
| | - Paul C Guest
- 1Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, São Paulo, Brazil;
- 2Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- 3Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Daniel Martins-de-Souza
- 1Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, São Paulo, Brazil;
- 4Experimental Medicine Research Cluster, University of Campinas, São Paulo, Brazil
- 5National Institute of Biomarkers in Neuropsychiatry, National Council for Scientific and Technological Development, São Paulo, Brazil
- 6D'Or Institute for Research and Education, São Paulo, Brazil
- 7INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D), São Paulo, Brazil
| |
Collapse
|
2
|
Bernstein HG, Smalla KH, Keilhoff G, Dobrowolny H, Kreutz MR, Steiner J. The many "Neurofaces" of Prohibitins 1 and 2: Crucial for the healthy brain, dysregulated in numerous brain disorders. J Chem Neuroanat 2023; 132:102321. [PMID: 37524128 DOI: 10.1016/j.jchemneu.2023.102321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are proteins that are nearly ubiquitously expressed. They are localized in mitochondria, cytosol and cell nuclei. In the healthy CNS, they occur in neurons and non-neuronal cells (oligodendrocytes, astrocytes, microglia, and endothelial cells) and fulfill pivotal functions in brain development and aging, the regulation of brain metabolism, maintenance of structural integrity, synapse formation, aminoacidergic neurotransmission and, probably, regulation of brain action of certain hypothalamic-pituitary hormones.With regard to the diseased brain there is increasing evidence that prohibitins are prominently involved in numerous major diseases of the CNS, which are summarized and discussed in the present review (brain tumors, neurotropic viruses, Alzheimer disease, Down syndrome, Fronto-temporal and vascular dementia, dementia with Lewy bodies, Parkinson disease, Huntington disease, Multiple sclerosis, Amyotrophic lateral sclerosis, stroke, alcohol use disorder, schizophrenia and autism). Unfortunately, there is no PHB-targeted therapy available for any of these diseases.
Collapse
Affiliation(s)
- Hans-Gert Bernstein
- Department of Psychiatry, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Karl-Heinz Smalla
- Leibniz Institute for Neurobiology, RG Neuroplasticity, D-39118 Magdeburg, Germany; Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany, Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Michael R Kreutz
- Leibniz Institute for Neurobiology, RG Neuroplastcity, D-39118 Magdeburg, Germany; University Medical Center Hamburg Eppendorf, Leibniz Group "Dendritic Organelles and Synaptic Function" ZMNH, Hamburg, Germany
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany
| |
Collapse
|
3
|
Khan A, Zahid S, Hasan B, Asif AR, Ahmed N. Mass Spectrometry based identification of site-specific proteomic alterations and potential pathways underlying the pathophysiology of schizophrenia. Mol Biol Rep 2023; 50:4931-4943. [PMID: 37076706 DOI: 10.1007/s11033-023-08431-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND Schizophrenia (SZ) is a complex multifactorial disorder that affects 1% of the population worldwide with no available effective treatment. Although proteomic alterations are reported in SZ however proteomic expression aberrations among different brain regions are not fully determined. Therefore, the present study aimed spatial differential protein expression profiling of three distinct regions of SZ brain and identification of associated affected biological pathways in SZ progression. METHODS AND RESULTS Comparative protein expression profiling of three distinct autopsied human brain regions (i.e., substantia nigra, hippocampus and prefrontal cortex) of SZ was performed with respective healthy controls. Using two-dimensional electrophoresis (2DE)-based nano liquid chromatography tandem mass spectrometry (Nano-LC MS /MS) analysis, 1443 proteins were identified out of which 58 connote to be significantly dysregulated, representing 26 of substantia nigra,14 of hippocampus and 18 of prefrontal cortex. The 58 differentially expressed proteins were further analyzed using Ingenuity pathway analysis (IPA). The IPA analysis provided protein-protein interaction networks of several proteins including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kb), extracellular signal regulated kinases 1/2 (ERK1/2), alpha serine / Threonine-protein kinase (AKT1), cellular tumor antigen p53 (TP53) and amyloid precursor protein (APP), holding prime positions in networks and interacts with most of the identified proteins and their closely interacting partners. CONCLUSION These findings provide conceptual insights of novel SZ related pathways and the cross talk of co and contra regulated proteins. This spatial proteomic analysis will further broaden the conceptual framework for schizophrenia research in future.
Collapse
Affiliation(s)
- Ayesha Khan
- Neurochemistry Research Laboratory, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Saadia Zahid
- Neurochemistry Research Laboratory, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Beena Hasan
- Neurochemistry Research Laboratory, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Abdul R Asif
- Institute of Clinical Chemistry, University Medical Center, Robert-Koch-Str. 40, 37075, Göttingen, Göttingen, Germany
| | - Nikhat Ahmed
- Neurochemistry Research Laboratory, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
4
|
Meta-analysis of brain samples of individuals with schizophrenia detects down-regulation of multiple ATP synthase encoding genes in both females and males. J Psychiatr Res 2023; 158:350-359. [PMID: 36640659 DOI: 10.1016/j.jpsychires.2023.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 10/05/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Schizophrenia is a chronic and debilitating mental disorder, with unknown pathophysiology. Converging lines of evidence suggest that mitochondrial functioning may be compromised in schizophrenia. Postmortem brain samples of individuals with schizophrenia showed dysregulated expression levels of genes encoding enzyme complexes comprising the mitochondrial electron transport chain (ETC), including ATP synthase, the fifth ETC complex. However, there are inconsistencies regarding the direction of change, i.e., up- or down-regulation, and differences between female and male patients were hardly examined. We have performed a systematic meta-analysis of the expression of 16 ATP synthase encoding genes in postmortem brain samples of individuals with schizophrenia vs. healthy controls of three regions: Brodmann Area 10 (BA10), BA22/Superior Temporal Gyrus (STG) and the cerebellum. Eight independent datasets were integrated (overall 294brain samples, 145 of individuals with schizophrenia and 149 controls). The meta-analysis was applied to all individuals with schizophrenia vs. the controls, and also to female and male patients vs. age-matched controls, separately. A significant down-regulation of two ATP synthase encoding genes was detected in schizophrenia, ATP5A1 and ATP5H, and a trend towards down-regulation of five further ATP synthase genes. The down-regulation tendency was shown for both females and males with schizophrenia. Our findings support the hypothesis that schizophrenia is associated with reduced ATP synthesis via the oxidative phosphorylation system, which is caused by reduced cellular demand of ATP. Abnormal cellular energy metabolism can lead to alterations in neural function and brain circuitry, and thereby to the cognitive and behavioral aberrations characteristic of schizophrenia.
Collapse
|
5
|
Proteomics and Schizophrenia: The Evolution of a Great Partnership. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1400:129-138. [DOI: 10.1007/978-3-030-97182-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
6
|
Doostparast Torshizi A, Duan J, Wang K. Cell-Type-Specific Proteogenomic Signal Diffusion for Integrating Multi-Omics Data Predicts Novel Schizophrenia Risk Genes. PATTERNS (NEW YORK, N.Y.) 2020; 1:100091. [PMID: 32984858 PMCID: PMC7518509 DOI: 10.1016/j.patter.2020.100091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/01/2020] [Accepted: 07/28/2020] [Indexed: 12/25/2022]
Abstract
Accumulation of diverse types of omics data on schizophrenia (SCZ) requires a systems approach to model the interplay between genome, transcriptome, and proteome. We introduce Markov affinity-based proteogenomic signal diffusion (MAPSD), a method to model intra-cellular protein trafficking paradigms and tissue-wise single-cell protein abundances. MAPSD integrates multi-omics data to amplify the signals at SCZ risk loci with small effect sizes, and reveal convergent disease-associated gene modules in the brain. We predicted a set of high-confidence SCZ risk loci followed by characterizing the subcellular localization of proteins encoded by candidate SCZ risk genes, and illustrated that most are enriched in neuronal cells in the cerebral cortex as well as Purkinje cells in the cerebellum. We demonstrated how the identified genes may be involved in neurodevelopment, how they may alter SCZ-related biological pathways, and how they facilitate drug repurposing. MAPSD is applicable in other polygenic diseases and can facilitate our understanding of disease mechanisms.
Collapse
Affiliation(s)
- Abolfazl Doostparast Torshizi
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, North Shore University Health System, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neurosciences, University of Chicago, Chicago, IL 60637, USA
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Behavioral abnormalities and phosphorylation deficits of extracellular signal-regulated kinases 1 and 2 in rat offspring of the maternal immune activation model. Physiol Behav 2020; 217:112805. [DOI: 10.1016/j.physbeh.2020.112805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/23/2019] [Accepted: 01/14/2020] [Indexed: 12/31/2022]
|
8
|
Borgmann-Winter KE, Wang K, Bandyopadhyay S, Torshizi AD, Blair IA, Hahn CG. The proteome and its dynamics: A missing piece for integrative multi-omics in schizophrenia. Schizophr Res 2020; 217:148-161. [PMID: 31416743 PMCID: PMC7500806 DOI: 10.1016/j.schres.2019.07.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/10/2019] [Accepted: 07/13/2019] [Indexed: 01/08/2023]
Abstract
The complex and heterogeneous pathophysiology of schizophrenia can be deconstructed by integration of large-scale datasets encompassing genes through behavioral phenotypes. Genome-wide datasets are now available for genetic, epigenetic and transcriptomic variations in schizophrenia, which are then analyzed by newly devised systems biology algorithms. A missing piece, however, is the inclusion of information on the proteome and its dynamics in schizophrenia. Proteomics has lagged behind omics of the genome, transcriptome and epigenome since analytic platforms were relatively less robust for proteins. There has been remarkable progress, however, in the instrumentation of liquid chromatography (LC) and mass spectrometry (MS) (LCMS), experimental paradigms and bioinformatics of the proteome. Here, we present a summary of methodological innovations of recent years in MS based proteomics and the power of new generation proteomics, review proteomics studies that have been conducted in schizophrenia to date, and propose how such data can be analyzed and integrated with other omics results. The function of a protein is determined by multiple molecular properties, i.e., subcellular localization, posttranslational modification (PTMs) and protein-protein interactions (PPIs). Incorporation of these properties poses additional challenges in proteomics and their integration with other omics; yet is a critical next step to close the loop of multi-omics integration. In sum, the recent advent of high-throughput proteome characterization technologies and novel mathematical approaches enable us to incorporate functional properties of the proteome to offer a comprehensive multi-omics based understanding of schizophrenia pathophysiology.
Collapse
Affiliation(s)
- Karin E Borgmann-Winter
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403, United States of America; Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States of America
| | - Kai Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States of America
| | - Sabyasachi Bandyopadhyay
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403, United States of America
| | - Abolfazl Doostparast Torshizi
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States of America
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Chang-Gyu Hahn
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403, United States of America.
| |
Collapse
|
9
|
Chan ST, McCarthy MJ, Vawter MP. Psychiatric drugs impact mitochondrial function in brain and other tissues. Schizophr Res 2020; 217:136-147. [PMID: 31744750 PMCID: PMC7228833 DOI: 10.1016/j.schres.2019.09.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 09/14/2019] [Accepted: 09/16/2019] [Indexed: 12/31/2022]
Abstract
Mitochondria have been linked to the etiology of schizophrenia (SZ). However, studies of mitochondria in SZ might be confounded by the effects of pharmacological treatment with antipsychotic drugs (APDs) and other common medications. This review summarizes findings on relevant mitochondria mechanisms underlying SZ, and the potential impact of psychoactive drugs including primarily APDs, but also antidepressants and anxiolytics. The summarized data suggest that APDs impair mitochondria function by decreasing Complex I activity and ATP production and dissipation of the mitochondria membrane potential. At the same time, in the brains of patients with SZ, antipsychotic drug treatment normalizes gene expression modules enriched in mitochondrial genes that are decreased in SZ. This indicates that APDs may have both positive and negative effects on mitochondria. The available evidence suggests three conclusions i) alterations in mitochondria functions in SZ exist prior to APD treatment, ii) mitochondria alterations in SZ can be reversed by APD treatment, and iii) APDs directly cause impairment of mitochondria function. Overall, the mechanisms of action of psychiatric drugs on mitochondria are both direct and indirect; we conclude the effects of APDs on mitochondria may contribute to both their therapeutic and metabolic side effects. These studies support the hypothesis that neuronal mitochondria are an etiological factor in SZ. Moreover, APDs and other drugs must be considered in the evaluation of this pathophysiological role of mitochondria in SZ. Considering these effects, pharmacological actions on mitochondria may be a worthwhile target for further APD development.
Collapse
Affiliation(s)
- Shawna T Chan
- Functional Genomics Laboratory, Department of Human Behavior and Psychiatry, University of California, Irvine, USA; School of Medicine University of California, Irvine, USA
| | - Michael J McCarthy
- Psychiatry Service VA San Diego Healthcare System, Department of Psychiatry, University of California, San Diego, USA
| | - Marquis P Vawter
- Functional Genomics Laboratory, Department of Human Behavior and Psychiatry, University of California, Irvine, USA.
| |
Collapse
|
10
|
Energization by multiple substrates and calcium challenge reveal dysfunctions in brain mitochondria in a model related to acute psychosis. J Bioenerg Biomembr 2019; 52:1-15. [PMID: 31853754 DOI: 10.1007/s10863-019-09816-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/26/2019] [Indexed: 12/23/2022]
Abstract
Schizophrenia etiology is unknown, nevertheless imbalances occurring in an acute psychotic episode are important to its development, such as alterations in cellular energetic state, REDOX homeostasis and intracellular Ca2+ management, all of which are controlled primarily by mitochondria. However, mitochondrial function was always evaluated singularly, in the presence of specific respiratory substrates, without considering the plurality of the electron transport system. In this study, mitochondrial function was analyzed under conditions of isolated or multiple respiratory substrates using brain mitochondria isolated from MK-801-exposed mice. Results showed a high H2O2 production in the presence of pyruvate/malate, with no change in oxygen consumption. In the condition of multiple substrates, however, this effect is lost. The analysis of Ca2+ retention capacity revealed a significant change in the uptake kinetics of this ion by mitochondria in MK-801-exposed animals. Futhermore, when mitochondria were exposed to calcium, a total loss of oxidative phosphorylation and an impressive increase in H2O2 production were observed in the condition of multiple substrates. There was no alteration in the activity of the antioxidant enzymes analyzed. The data demonstrate for the first time, in an animal model of psychosis, two important aspects (1) mitochondria may compensate deficiencies in a single mitochondrial complex when they oxidize several substrates simultaneously, (2) Ca2+ handling is compromised in MK-801-exposed mice, resulting in a loss of phosphorylative capacity and an increase in H2O2 production. These data favor the hypothesis that disruption of key physiological roles of mitochondria may be a trigger in acute psychosis and, consequently, schizophrenia.
Collapse
|
11
|
Hoffman JL, Faccidomo S, Kim M, Taylor SM, Agoglia AE, May AM, Smith EN, Wong LC, Hodge CW. Alcohol drinking exacerbates neural and behavioral pathology in the 3xTg-AD mouse model of Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:169-230. [PMID: 31733664 PMCID: PMC6939615 DOI: 10.1016/bs.irn.2019.10.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that represents the most common cause of dementia in the United States. Although the link between alcohol use and AD has been studied, preclinical research has potential to elucidate neurobiological mechanisms that underlie this interaction. This study was designed to test the hypothesis that nondependent alcohol drinking exacerbates the onset and magnitude of AD-like neural and behavioral pathology. We first evaluated the impact of voluntary 24-h, two-bottle choice home-cage alcohol drinking on the prefrontal cortex and amygdala neuroproteome in C57BL/6J mice and found a striking association between alcohol drinking and AD-like pathology. Bioinformatics identified the AD-associated proteins MAPT (Tau), amyloid beta precursor protein (APP), and presenilin-1 (PSEN-1) as the main modulators of alcohol-sensitive protein networks that included AD-related proteins that regulate energy metabolism (ATP5D, HK1, AK1, PGAM1, CKB), cytoskeletal development (BASP1, CAP1, DPYSL2 [CRMP2], ALDOA, TUBA1A, CFL2, ACTG1), cellular/oxidative stress (HSPA5, HSPA8, ENO1, ENO2), and DNA regulation (PURA, YWHAZ). To address the impact of alcohol drinking on AD, studies were conducted using 3xTg-AD mice that express human MAPT, APP, and PSEN-1 transgenes and develop AD-like brain and behavioral pathology. 3xTg-AD and wild-type mice consumed alcohol or saccharin for 4 months. Behavioral tests were administered during a 1-month alcohol-free period. Alcohol intake induced AD-like behavioral pathologies in 3xTg-AD mice including impaired spatial memory in the Morris Water Maze, diminished sensorimotor gating as measured by prepulse inhibition, and exacerbated conditioned fear. Multiplex immunoassay conducted on brain lysates showed that alcohol drinking upregulated primary markers of AD pathology in 3xTg-AD mice: Aβ 42/40 ratio in the lateral entorhinal and prefrontal cortex and total Tau expression in the lateral entorhinal cortex, medial prefrontal cortex, and amygdala at 1-month post alcohol exposure. Immunocytochemistry showed that alcohol use upregulated expression of pTau (Ser199/Ser202) in the hippocampus, which is consistent with late-stage AD. According to the NIA-AA Research Framework, these results suggest that alcohol use is associated with Alzheimer's pathology. Results also showed that alcohol use was associated with a general reduction in Akt/mTOR signaling via several phosphoproteins (IR, IRS1, IGF1R, PTEN, ERK, mTOR, p70S6K, RPS6) in multiple brain regions including hippocampus and entorhinal cortex. Dysregulation of Akt/mTOR phosphoproteins suggests alcohol may target this pathway in AD progression. These results suggest that nondependent alcohol drinking increases the onset and magnitude of AD-like neural and behavioral pathology in 3xTg-AD mice.
Collapse
Affiliation(s)
- Jessica L Hoffman
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sara Faccidomo
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michelle Kim
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Seth M Taylor
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Abigail E Agoglia
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ashley M May
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Evan N Smith
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - L C Wong
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Clyde W Hodge
- Department of Psychiatry, Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
12
|
S Cassoli J, Brandão-Teles C, G Santana A, H M F Souza G, Martins-de-Souza D. Ion Mobility-Enhanced Data-Independent Acquisitions Enable a Deep Proteomic Landscape of Oligodendrocytes. Proteomics 2017; 17. [PMID: 28861932 DOI: 10.1002/pmic.201700209] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/05/2017] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes are a type of neuroglia that provide trophic support and insulation to axons in the central nervous system. The genesis and maturation of oligodendrocytes are essential processes for myelination and the course of CNS development. Using ion mobility-enhanced, data-independent acquisitions and 2D-nanoUPLC fractionation operating at nanoscale flow rates, we established a comprehensive data set of proteins expressed by the human oligodendroglia cell line MO3.13. The final dataset incorporating all fractions comprised 223 531 identified peptides assigned to 10 390 protein hits, an improvement of 4.5 times on identified proteins described previously by our group using the same cell line. Identified proteins play pivotal roles in many biological processes such as cell growth and development and energy metabolism, providing a rich resource for future studies on oligodendrocyte development, myelination, axonal support, and the regulation of such process. Our results can help further studies that use MO3.13 cells as a tool of investigation, not only in relation to oligodendrocyte maturation, but also to diseases that have oligodendrocytes as key players. All MS data have been deposited in the ProteomeXchange with identifier PXD004696.
Collapse
Affiliation(s)
- Juliana S Cassoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Caroline Brandão-Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Aline G Santana
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Gustavo H M F Souza
- Mass Spectrometry Research and Development Laboratory, Health Sciences Department, Waters Corporation, São Paulo, SP, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico, Brazil
| |
Collapse
|
13
|
Schmitt A, Martins-de-Souza D, Akbarian S, Cassoli JS, Ehrenreich H, Fischer A, Fonteh A, Gattaz WF, Gawlik M, Gerlach M, Grünblatt E, Halene T, Hasan A, Hashimoto K, Kim YK, Kirchner SK, Kornhuber J, Kraus TFJ, Malchow B, Nascimento JM, Rossner M, Schwarz M, Steiner J, Talib L, Thibaut F, Riederer P, Falkai P. Consensus paper of the WFSBP Task Force on Biological Markers: Criteria for biomarkers and endophenotypes of schizophrenia, part III: Molecular mechanisms. World J Biol Psychiatry 2017; 18:330-356. [PMID: 27782767 DOI: 10.1080/15622975.2016.1224929] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Despite progress in identifying molecular pathophysiological processes in schizophrenia, valid biomarkers are lacking for both the disease and treatment response. METHODS This comprehensive review summarises recent efforts to identify molecular mechanisms on the level of protein and gene expression and epigenetics, including DNA methylation, histone modifications and micro RNA expression. Furthermore, it summarises recent findings of alterations in lipid mediators and highlights inflammatory processes. The potential that this research will identify biomarkers of schizophrenia is discussed. RESULTS Recent studies have not identified clear biomarkers for schizophrenia. Although several molecular pathways have emerged as potential candidates for future research, a complete understanding of these metabolic pathways is required to reveal better treatment modalities for this disabling condition. CONCLUSIONS Large longitudinal cohort studies are essential that pair a thorough phenotypic and clinical evaluation for example with gene expression and proteome analysis in blood at multiple time points. This approach might identify biomarkers that allow patients to be stratified according to treatment response and ideally also allow treatment response to be predicted. Improved knowledge of molecular pathways and epigenetic mechanisms, including their potential association with environmental influences, will facilitate the discovery of biomarkers that could ultimately be effective tools in clinical practice.
Collapse
Affiliation(s)
- Andrea Schmitt
- a Department of Psychiatry and Psychotherapy , LMU Munich , Germany.,b Laboratory of Neuroscience (LIM27) , Institute of Psychiatry, University of Sao Paulo , Sao Paulo , Brazil
| | - Daniel Martins-de-Souza
- b Laboratory of Neuroscience (LIM27) , Institute of Psychiatry, University of Sao Paulo , Sao Paulo , Brazil.,c Laboratory of Neuroproteomics, Department of Biochemistry , Institute of Biology University of Campinas (UNICAMP), Campinas , SP , Brazil
| | - Schahram Akbarian
- d Division of Psychiatric Epigenomics, Departments of Psychiatry and Neuroscience , Mount Sinai School of Medicine , New York , USA
| | - Juliana S Cassoli
- c Laboratory of Neuroproteomics, Department of Biochemistry , Institute of Biology University of Campinas (UNICAMP), Campinas , SP , Brazil
| | - Hannelore Ehrenreich
- e Clinical Neuroscience , Max Planck Institute of Experimental Medicine, DFG Centre for Nanoscale Microscopy & Molecular Physiology of the Brain , Göttingen , Germany
| | - Andre Fischer
- f Research Group for Epigenetics in Neurodegenerative Diseases , German Centre for Neurodegenerative Diseases (DZNE), Göttingen , Germany.,g Department of Psychiatry and Psychotherapy , University Medical Centre Göttingen , Germany
| | - Alfred Fonteh
- h Neurosciences , Huntington Medical Research Institutes , Pasadena , CA , USA
| | - Wagner F Gattaz
- b Laboratory of Neuroscience (LIM27) , Institute of Psychiatry, University of Sao Paulo , Sao Paulo , Brazil
| | - Michael Gawlik
- i Department of Psychiatry and Psychotherapy , University of Würzburg , Germany
| | - Manfred Gerlach
- j Centre for Mental Health, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University of Würzburg , Germany
| | - Edna Grünblatt
- i Department of Psychiatry and Psychotherapy , University of Würzburg , Germany.,k Department of Child and Adolescent Psychiatry and Psychotherapy , Psychiatric Hospital, University of Zürich , Switzerland.,l Neuroscience Centre Zurich , University of Zurich and the ETH Zurich , Switzerland.,m Zurich Centre for Integrative Human Physiology , University of Zurich , Switzerland
| | - Tobias Halene
- d Division of Psychiatric Epigenomics, Departments of Psychiatry and Neuroscience , Mount Sinai School of Medicine , New York , USA
| | - Alkomiet Hasan
- a Department of Psychiatry and Psychotherapy , LMU Munich , Germany
| | - Kenij Hashimoto
- n Division of Clinical Neuroscience , Chiba University Centre for Forensic Mental Health , Chiba , Japan
| | - Yong-Ku Kim
- o Department of Psychiatry , Korea University, College of Medicine , Republic of Korea
| | | | - Johannes Kornhuber
- p Department of Psychiatry and Psychotherapy , Friedrich-Alexander-University Erlangen-Nuremberg , Erlangen , Germany
| | | | - Berend Malchow
- a Department of Psychiatry and Psychotherapy , LMU Munich , Germany
| | - Juliana M Nascimento
- c Laboratory of Neuroproteomics, Department of Biochemistry , Institute of Biology University of Campinas (UNICAMP), Campinas , SP , Brazil
| | - Moritz Rossner
- r Department of Psychiatry, Molecular and Behavioural Neurobiology , LMU Munich , Germany.,s Research Group Gene Expression , Max Planck Institute of Experimental Medicine , Göttingen , Germany
| | - Markus Schwarz
- t Institute for Laboratory Medicine, LMU Munich , Germany
| | - Johann Steiner
- u Department of Psychiatry , University of Magdeburg , Magdeburg , Germany
| | - Leda Talib
- b Laboratory of Neuroscience (LIM27) , Institute of Psychiatry, University of Sao Paulo , Sao Paulo , Brazil
| | - Florence Thibaut
- v Department of Psychiatry , University Hospital Cochin (site Tarnier), University of Paris-Descartes, INSERM U 894 Centre Psychiatry and Neurosciences , Paris , France
| | - Peter Riederer
- w Center of Psychic Health; Department of Psychiatry, Psychosomatics and Psychotherapy , University Hospital of Würzburg , Germany
| | - Peter Falkai
- a Department of Psychiatry and Psychotherapy , LMU Munich , Germany
| | | |
Collapse
|
14
|
Saia-Cereda VM, Santana AG, Schmitt A, Falkai P, Martins-de-Souza D. The Nuclear Proteome of White and Gray Matter from Schizophrenia Postmortem Brains. MOLECULAR NEUROPSYCHIATRY 2017; 3:37-52. [PMID: 28879200 PMCID: PMC5582429 DOI: 10.1159/000477299] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022]
Abstract
Schizophrenia (SCZ) is a serious neuropsychiatric disorder that manifests through several symptoms from early adulthood. Numerous studies over the last decades have led to significant advances in increasing our understanding of the factors involved in SCZ. For example, mass spectrometry-based proteomic analysis has provided important insights by uncovering protein dysfunctions inherent to SCZ. Here, we present a comprehensive analysis of the nuclear proteome of postmortem brain tissues from corpus callosum (CC) and anterior temporal lobe (ATL). We show an overview of the role of deregulated nuclear proteins in these two main regions of the brain: the first, mostly composed of glial cells and axons of neurons, and the second, represented mainly by neuronal cell bodies. These samples were collected from SCZ patients in an attempt to characterize the role of the nucleus in the disease process. With the ATL nucleus enrichment, we found 224 proteins present at different levels, and 76 of these were nuclear proteins. In the CC analysis, we identified 119 present at different levels, and 24 of these were nuclear proteins. The differentially expressed nuclear proteins of ATL are mainly associated with the spliceosome, whereas those of the CC region are associated with calcium/calmodulin signaling.
Collapse
Affiliation(s)
- Verônica M. Saia-Cereda
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Aline G. Santana
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany
- Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- UNICAMP's Neurobiology Center, Campinas, Brazil
| |
Collapse
|
15
|
Lanfredi G, Reis-de-Oliveira G, Saia-Cereda VM, Guest PC, Martins-de-Souza D, Faça VM. Selective Reaction Monitoring Mass Spectrometry for Quantitation of Glycolytic Enzymes in Postmortem Brain Samples. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 974:205-212. [PMID: 28353237 DOI: 10.1007/978-3-319-52479-5_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Patients with psychiatric disorders exhibit dysfunctions in peripheral and central metabolism. This may be a root cause of impaired neuronal function, manifested as changes in mood, behavior, and cognitive capabilities in patients suffering with these conditions. Here we describe a selective reaction monitoring mass spectrometry (SRM-MS)-based targeted proteomic protocol for precise simultaneous quantitation of three glycolytic enzymes in postmortem brain tissue extracts. The SRM-MS approach has several advantages in terms of sensitivity, reproducibility, and reduced sample consumption, compared to traditional MS methods.
Collapse
Affiliation(s)
- Guilherme Lanfredi
- Department of Biochemistry and Immunology, RibeirãoPreto Medical School, University of São Paulo, RibeirãoPreto, SP, Brazil
| | - Guiherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Veronica M Saia-Cereda
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.,UNICAMP's Neurobiology Center, Campinas, Brazil
| | - Vitor M Faça
- Department of Biochemistry and Immunology, RibeirãoPreto Medical School, University of São Paulo, RibeirãoPreto, SP, Brazil. .,Center for Cell Based Therapy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
16
|
Pinacho R, Villalmanzo N, Meana JJ, Ferrer I, Berengueras A, Haro JM, Villén J, Ramos B. Altered CSNK1E, FABP4 and NEFH protein levels in the dorsolateral prefrontal cortex in schizophrenia. Schizophr Res 2016; 177:88-97. [PMID: 27236410 DOI: 10.1016/j.schres.2016.04.050] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 03/15/2016] [Accepted: 04/27/2016] [Indexed: 11/28/2022]
Abstract
Schizophrenia constitutes a complex disease. Negative and cognitive symptoms are enduring and debilitating components of the disorder, highly associated to disability and burden. Disrupted neurotransmission circuits in dorsolateral prefrontal cortex (DLPFC) have been related to these symptoms. To identify candidates altered in schizophrenia, we performed a pilot proteomic analysis on postmortem human DLPFC tissue from patients with schizophrenia (n=4) and control (n=4) subjects in a pool design using differential isotope peptide labelling followed by liquid chromatography tandem mass spectrometry (LC-MS/MS). We quantified 1315 proteins with two or more unique peptides, 116 of which showed altered changes. Of these altered proteins, we selected four with potential roles on cell signaling, neuronal development and synapse functioning for further validation: casein kinase I isoform epsilon (CSNK1E), fatty acid-binding protein 4 (FABP4), neurofilament triplet H protein (NEFH), and retinal dehydrogenase 1 (ALDH1A1). Immunoblot validation confirmed our proteomic findings of these proteins being decreased in abundance in the schizophrenia samples. Additionally, we conducted immunoblot validation of these candidates on an independent sample cohort comprising 23 patients with chronic schizophrenia and 23 matched controls. In this second cohort, CSNK1E, FABP4 and NEFH were reduced in the schizophrenia group while ALDH1A1 did not significantly change. This study provides evidence indicating these proteins are decreased in schizophrenia: CSNK1E, involved in circadian molecular clock signaling, FABP4 with possible implication in synapse functioning, and NEFH, important for cytoarchitecture organization. Hence, these findings suggest the possible implication of these proteins in the cognitive and/or negative symptoms in schizophrenia.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain
| | - Núria Villalmanzo
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain
| | - J Javier Meana
- Departamento de Farmacología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Instituto BioCruces, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Bº Sarriena s/n, 48940 Leioa, Bizkaia, Spain
| | - Isidre Ferrer
- Instituto de Neuropatología, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Centro de Investigación Biomédica en Red para enfermedades neurodegenerativas, CIBERNED, Feixa Llarga s/n, Hospitalet de LLobregat, 08907 Barcelona, Spain
| | - Adriana Berengueras
- Banc de Teixits Neurologics, Parc Sanitari Sant Joan de Déu, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain
| | - Josep M Haro
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain
| | - Judit Villén
- Genome Sciences Department, School of Medicine, University of Washington, 3720 15th Ave NE, Seattle 98195, WA, USA
| | - Belén Ramos
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain.
| |
Collapse
|
17
|
Pinacho R, Vila E, Prades R, Tarragó T, Castro E, Ferrer I, Ramos B. The glial phosphorylase of glycogen isoform is reduced in the dorsolateral prefrontal cortex in chronic schizophrenia. Schizophr Res 2016; 177:37-43. [PMID: 27156240 DOI: 10.1016/j.schres.2016.04.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 04/10/2016] [Accepted: 04/13/2016] [Indexed: 12/13/2022]
Abstract
Reduced glutamatergic activity and energy metabolism in the dorsolateral prefrontal cortex (DLPFC) have been described in schizophrenia. Glycogenolysis in astrocytes is responsible for providing neurons with lactate as a transient energy supply helping to couple glutamatergic neurotransmission and glucose utilization in the brain. This mechanism could be disrupted in schizophrenia. The aim of this study was to explore whether the protein levels of the astrocyte isoform of glycogen phosphorylase (PYGM), key enzyme of glycogenolysis, and the isoform A of Ras-related C3 botulinum toxin substrate 1 (RAC1), a kinase that regulates PYGM activity, are altered in the postmortem DLPFC of chronic schizophrenia patients (n=23) and matched controls (n=23). We also aimed to test NMDAR blockade effect on these proteins in the mouse cortex and cortical astrocytes and antipsychotic treatments in rats. Here we report a reduction in PYGM and RAC1 protein levels in the DLPFC in schizophrenia. We found that treatment with the NMDAR antagonist dizocilpine in mice as a model of psychosis increased PYGM and reduced RAC1 protein levels. The same result was observed in rat cortical astroglial-enriched cultures. 21-day haloperidol treatment increased PYGM levels in rats. These results show that PYGM and RAC1 are altered in the DLPFC in chronic schizophrenia and are controlled by NMDA signalling in the rodent cortex and cortical astrocytes suggesting an altered NMDA-dependent glycogenolysis in astrocytes in schizophrenia. Together, this study provides evidence of a NMDA-dependent transient local energy deficit in neuron-glia crosstalk in schizophrenia, contributing to energy deficits of the disorder.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830, Sant Boi de Llobregat, Barcelona, Spain
| | - Elia Vila
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830, Sant Boi de Llobregat, Barcelona, Spain
| | - Roger Prades
- Iproteos S.L., Baldiri I Reixac, 10, 08028 Barcelona, Spain
| | - Teresa Tarragó
- Iproteos S.L., Baldiri I Reixac, 10, 08028 Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), Baldiri I Reixac, 10, 08028 Barcelona, Spain
| | - Elena Castro
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), Departamento de Fisiología y Farmacología, Universidad de Cantabria, 39011, Santander, Spain, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain
| | - Isidre Ferrer
- Instituto de Neuropatología, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Centro de Investigación Biomédica en Red para enfermedades neurodegenerativas, CIBERNED, Feixa Llarga s/n, 08907 Hospitalet de LLobregat, Barcelona, Spain
| | - Belén Ramos
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830, Sant Boi de Llobregat, Barcelona, Spain.
| |
Collapse
|
18
|
Nakamura H, Yamashita N, Kimura A, Kimura Y, Hirano H, Makihara H, Kawamoto Y, Jitsuki-Takahashi A, Yonezaki K, Takase K, Miyazaki T, Nakamura F, Tanaka F, Goshima Y. Comprehensive behavioral study and proteomic analyses of CRMP2-deficient mice. Genes Cells 2016; 21:1059-1079. [DOI: 10.1111/gtc.12403] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 07/29/2016] [Indexed: 01/02/2023]
Affiliation(s)
- Haruko Nakamura
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- Department of Neurology and Stroke Medicine; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- JSPS Postdoctoral Fellowship for Research Abroad; Tokyo 102-0083 Japan
| | - Ayuko Kimura
- Advanced Medical Research Center; Yokohama City University; Yokohama 236-0004 Japan
| | - Yayoi Kimura
- Advanced Medical Research Center; Yokohama City University; Yokohama 236-0004 Japan
| | - Hisashi Hirano
- Advanced Medical Research Center; Yokohama City University; Yokohama 236-0004 Japan
| | - Hiroko Makihara
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Yuko Kawamoto
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- Department of Neurology and Stroke Medicine; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Aoi Jitsuki-Takahashi
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Kumiko Yonezaki
- Department of Anesthesiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Kenkichi Takase
- Department of Anesthesiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- Laboratory of Psychology; Jichi Medical University; Shimotsuke 329-0498 Japan
| | - Tomoyuki Miyazaki
- Department of Anesthesiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- Department of Physiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Fumio Nakamura
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| |
Collapse
|
19
|
Makihara H, Nakai S, Ohkubo W, Yamashita N, Nakamura F, Kiyonari H, Shioi G, Jitsuki-Takahashi A, Nakamura H, Tanaka F, Akase T, Kolattukudy P, Goshima Y. CRMP1 and CRMP2 have synergistic but distinct roles in dendritic development. Genes Cells 2016; 21:994-1005. [DOI: 10.1111/gtc.12399] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/02/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Hiroko Makihara
- Department of Molecular Pharmacology and Neurobiology; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
- Biological Science and Nursing; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
| | - Shiori Nakai
- Department of Molecular Pharmacology and Neurobiology; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
| | - Wataru Ohkubo
- Department of Molecular Pharmacology and Neurobiology; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
| | - Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
- JSPS Postdoctoral Fellowship for Research Abroad; Chiyoda-ku 102-0083 Japan
- Department of Biology; Johns Hopkins University; Baltimore MD 21218 USA
| | - Fumio Nakamura
- Department of Molecular Pharmacology and Neurobiology; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
| | - Hiroshi Kiyonari
- Animal Resource Development Unit; RIKEN Center for Life Science Technologies; 2-2-3 Minatojima Minami-machi Chuou-ku Kobe 650-0047 Japan
- Genetic Engineering Team; RIKEN Center for Life Science Technologies; 2-2-3 Minatojima Minami-machi Chuou-ku Kobe 650-0047 Japan
| | - Go Shioi
- Genetic Engineering Team; RIKEN Center for Life Science Technologies; 2-2-3 Minatojima Minami-machi Chuou-ku Kobe 650-0047 Japan
| | - Aoi Jitsuki-Takahashi
- Department of Molecular Pharmacology and Neurobiology; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
| | - Haruko Nakamura
- Department of Molecular Pharmacology and Neurobiology; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
- Department of Neurology and Stroke Medicine; Graduate School of Medicine; Yokohama City University; Yokohama 236-0004 Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine; Graduate School of Medicine; Yokohama City University; Yokohama 236-0004 Japan
| | - Tomoko Akase
- Biological Science and Nursing; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
| | - Pappachan Kolattukudy
- Burnett School of Biomedical Sciences; College of Medicine; University of Central Florida; Orlando FL USA
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology; Graduate School of Medicine; Yokohama City University; 3-9 Fuku-ura Kanazawa-ku Yokohama 236-0004 Japan
| |
Collapse
|
20
|
Sethi S, Chourasia D, Parhar IS. Approaches for targeted proteomics and its potential applications in neuroscience. J Biosci 2016; 40:607-27. [PMID: 26333406 DOI: 10.1007/s12038-015-9537-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
An extensive guide on practicable and significant quantitative proteomic approaches in neuroscience research is important not only because of the existing overwhelming limitations but also for gaining valuable understanding into brain function and deciphering proteomics from the workbench to the bedside. Early methodologies to understand the functioning of biological systems are now improving with high-throughput technologies, which allow analysis of various samples concurrently, or of thousand of analytes in a particular sample. Quantitative proteomic approaches include both gel-based and non-gel-based methods that can be further divided into different labelling approaches. This review will emphasize the role of existing technologies, their advantages and disadvantages, as well as their applications in neuroscience. This review will also discuss advanced approaches for targeted proteomics using isotope-coded affinity tag (ICAT) coupled with laser capture microdissection (LCM) followed by liquid chromatography tandem mass spectrometric (LC-MS/MS) analysis. This technology can further be extended to single cell proteomics in other areas of biological sciences and can be combined with other 'omics' approaches to reveal the mechanism of a cellular alterations. This approach may lead to further investigation in basic biology, disease analysis and surveillance, as well as drug discovery. Although numerous challenges still exist, we are confident that this approach will increase the understanding of pathological mechanisms involved in neuroendocrinology, neuropsychiatric and neurodegenerative disorders by delivering protein biomarker signatures for brain dysfunction.
Collapse
Affiliation(s)
- Sumit Sethi
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, MONASH University, Selangor Darul Ehsan, Malaysia,
| | | | | |
Collapse
|
21
|
Coumans JVF, Palanisamy SKA, McFarlane J, Moens PDJ. Proteomic and Microscopic Strategies towards the Analysis of the Cytoskeletal Networks in Major Neuropsychiatric Disorders. Int J Mol Sci 2016; 17:E581. [PMID: 27104521 PMCID: PMC4849037 DOI: 10.3390/ijms17040581] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 11/17/2022] Open
Abstract
Mental health disorders have become worldwide health priorities. It is estimated that in the next 20 years they will account for a 16 trillion United State dollars (US$) loss. Up to now, the underlying pathophysiology of psychiatric disorders remains elusive. Altered cytoskeleton proteins expression that may influence the assembly, organization and maintenance of cytoskeletal integrity has been reported in major depressive disorders, schizophrenia and to some extent bipolar disorders. The use of quantitative proteomics, dynamic microscopy and super-resolution microscopy to investigate disease-specific protein signatures holds great promise to improve our understanding of these disorders. In this review, we present the currently available quantitative proteomic approaches use in neurology, gel-based, stable isotope-labelling and label-free methodologies and evaluate their strengths and limitations. We also reported on enrichment/subfractionation methods that target the cytoskeleton associated proteins and discuss the need of alternative methods for further characterization of the neurocytoskeletal proteome. Finally, we present live cell imaging approaches and emerging dynamic microscopy technology that will provide the tools necessary to investigate protein interactions and their dynamics in the whole cells. While these areas of research are still in their infancy, they offer huge potential towards the understanding of the neuronal network stability and its modification across neuropsychiatric disorders.
Collapse
Affiliation(s)
- Joëlle V F Coumans
- School of Rural Medicine, University of New England, Armidale, NSW 2351, Australia.
| | - Suresh K A Palanisamy
- Center for Bioactive Discovery in Health and Aging, School of Science and Technology, University of New England, Armidale, NSW 2351, Australia.
| | - Jim McFarlane
- Center for Bioactive Discovery in Health and Aging, School of Science and Technology, University of New England, Armidale, NSW 2351, Australia.
| | - Pierre D J Moens
- Center for Bioactive Discovery in Health and Aging, School of Science and Technology, University of New England, Armidale, NSW 2351, Australia.
| |
Collapse
|
22
|
Cassoli JS, Iwata K, Steiner J, Guest PC, Turck CW, Nascimento JM, Martins-de-Souza D. Effect of MK-801 and Clozapine on the Proteome of Cultured Human Oligodendrocytes. Front Cell Neurosci 2016; 10:52. [PMID: 26973466 PMCID: PMC4776125 DOI: 10.3389/fncel.2016.00052] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 02/15/2016] [Indexed: 01/06/2023] Open
Abstract
Separate lines of evidence have demonstrated the involvement of N-methyl-D-aspartate (NMDA) receptor and oligodendrocyte dysfunctions in schizophrenia. Here, we have carried out shotgun mass spectrometry proteome analysis of oligodendrocytes treated with the NMDA receptor antagonist MK-801 to gain potential insights into these effects at the molecular level. The MK-801 treatment led to alterations in the levels of 68 proteins, which are associated with seven distinct biological processes. Most of these proteins are involved in energy metabolism and many have been found to be dysregulated in previous proteomic studies of post-mortem brain tissues from schizophrenia patients. Finally, addition of the antipsychotic clozapine to MK-801-treated oligodendrocyte cultures resulted in changes in the levels of 45 proteins and treatment with clozapine alone altered 122 proteins and many of these showed opposite changes to the MK-801 effects. Therefore, these proteins and the associated energy metabolism pathways should be explored as potential biomarkers of antipsychotic efficacy. In conclusion, MK-801 treatment of oligodendrocytes may provide a useful model for testing the efficacy of novel treatment approaches.
Collapse
Affiliation(s)
- Juliana S Cassoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Keiko Iwata
- United Graduate School of Child Development, Department of Development of Functional Brain Activities, Research Center for Child Mental Development, Hamamatsu University School of Medicine, Osaka University and Kanazawa University and Chiba University and University of Fukui Fukui, Japan
| | - Johann Steiner
- Department of Psychiatry, University of Magdeburg Magdeburg, Germany
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Christoph W Turck
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry Munich, Germany
| | - Juliana M Nascimento
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of CampinasCampinas, Brazil; D'Or Institute for Research and Education Rio de Janeiro, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of CampinasCampinas, Brazil; UNICAMP Neurobiology CenterCampinas, Brazil
| |
Collapse
|
23
|
Rong R, Yang H, Rong L, Wei X, Li Q, Liu X, Gao H, Xu Y, Zhang Q. Proteomic analysis of PSD-93 knockout mice following the induction of ischemic cerebral injury. Neurotoxicology 2016; 53:1-11. [DOI: 10.1016/j.neuro.2015.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 12/04/2015] [Accepted: 12/05/2015] [Indexed: 01/09/2023]
|
24
|
Kaur G, Sharma A, Gupta M, Kaur T. Obesity and Neuroinflammation. INFLAMMATION: THE COMMON LINK IN BRAIN PATHOLOGIES 2016:297-323. [DOI: 10.1007/978-981-10-1711-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
25
|
Carboni L, Domenici E. Proteome effects of antipsychotic drugs: Learning from preclinical models. Proteomics Clin Appl 2015; 10:430-41. [PMID: 26548651 DOI: 10.1002/prca.201500087] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/27/2015] [Accepted: 11/03/2015] [Indexed: 02/02/2023]
Abstract
Proteome-wide expression analyses are performed in the brain of schizophrenia patients to understand the biological basis of the disease and discover molecular paths for new clinical interventions. A major issue with postmortem analysis is the lack of tools to discern molecular modulation related to the disease from dysregulation due to medications. We review available proteome-wide analysis of antipsychotic treatment in rodents, highlighting shared dysregulated pathways that may contribute to an extended view of molecular processes underlying their pharmacological activity. Fourteen proteomic studies conducted with typical and atypical antipsychotic treatments were examined; hypothesis-based approaches are also briefly discussed. Treatment with antipsychotics mainly affects proteins belonging to metabolic pathways involved in energy generation, both in glycolytic and oxidative phosphorylation pathways, suggesting antipsychotics-induced impairments in metabolism. Nevertheless, schizophrenic patients show impaired glucose metabolism and mitochondrial dysfunctions independent of therapy. Other antipsychotics-induced changes shared by different studies implicate cytoskeletal and synaptic function proteins. The mechanism can be related to the reorganization of dendritic spines resulting from neural plasticity events induced by treatments affecting neurotransmitter circuitry. However, metabolic and plasticity pathways activated by antipsychotics can also play an authentic role in the etiopathological basis of schizophrenia.
Collapse
Affiliation(s)
- Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Enrico Domenici
- Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases Discovery & Translational Medicine Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
26
|
Sethi S, Brietzke E. Omics-Based Biomarkers: Application of Metabolomics in Neuropsychiatric Disorders. Int J Neuropsychopharmacol 2015; 19:pyv096. [PMID: 26453695 PMCID: PMC4815467 DOI: 10.1093/ijnp/pyv096] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/17/2015] [Indexed: 12/22/2022] Open
Abstract
One of the major concerns of modern society is to identify putative biomarkers that serve as a valuable early diagnostic tool to identify a subset of patients with increased risk to develop neuropsychiatric disorders. Biomarker identification in neuropsychiatric disorders is proposed to offer a number of important benefits to patient well-being, including prediction of forthcoming disease, diagnostic precision, and a level of disease description that would guide treatment choice. Nowadays, the metabolomics approach has unlocked new possibilities in diagnostics of devastating disorders like neuropsychiatric disorders. Metabolomics-based technologies have the potential to map early biochemical changes in disease and hence provide an opportunity to develop predictive biomarkers that can be used as indicators of pathological abnormalities prior to development of clinical symptoms of neuropsychiatric disorders. This review highlights different -omics strategies for biomarker discovery in neuropsychiatric disorders. We also highlight initial outcomes from metabolomics studies in psychiatric disorders such as schizophrenia, bipolar disorder, and addictive disorders. This review will also present issues and challenges regarding the implementation of the metabolomics approach as a routine diagnostic tool in the clinical laboratory in context with neuropsychiatric disorders.
Collapse
Affiliation(s)
| | - Elisa Brietzke
- Interdisciplinary Laboratory for Clinical Neuroscience (LiNC), Department of Psychiatry, Universidade Federal de São Paulo - UNIFESP, São Paulo, Brazil.
| |
Collapse
|
27
|
Martins-de-Souza D, Cassoli JS, Nascimento JM, Hensley K, Guest PC, Pinzon-Velasco AM, Turck CW. The protein interactome of collapsin response mediator protein-2 (CRMP2/DPYSL2) reveals novel partner proteins in brain tissue. Proteomics Clin Appl 2015; 9:817-31. [PMID: 25921334 DOI: 10.1002/prca.201500004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/23/2015] [Accepted: 04/27/2015] [Indexed: 01/18/2023]
Abstract
PURPOSE Collapsin response mediator protein-2 (CRMP2) is a CNS protein involved in neuronal development, axonal and neuronal growth, cell migration, and protein trafficking. Recent studies have linked perturbations in CRMP2 function to neurodegenerative disorders such as Alzheimer's disease, neuropathic pain, and Batten disease, and to psychiatric disorders such as schizophrenia. Like most proteins, CRMP2 functions though interactions with a molecular network of proteins and other molecules. EXPERIMENTAL DESIGN Here, we have attempted to identify additional proteins of the CRMP2 interactome to provide further leads about its roles in neurological functions. We used a combined co-immunoprecipitation and shotgun proteomic approach in order to identify CRMP2 protein partners. RESULTS We identified 78 CRMP2 protein partners not previously reported in public protein interaction databases. These were involved in seven biological processes, which included cell signaling, growth, metabolism, trafficking, and immune function, according to Gene Ontology classifications. Furthermore, 32 different molecular functions were found to be associated with these proteins, such as RNA binding, ribosomal functions, transporter activity, receptor activity, serine/threonine phosphatase activity, cell adhesion, cytoskeletal protein binding and catalytic activity. In silico pathway interactome construction revealed a highly connected network with the most overrepresented functions corresponding to semaphorin interactions, along with axon guidance and WNT5A signaling. CONCLUSIONS AND CLINICAL RELEVANCE Taken together, these findings suggest that the CRMP2 pathway is critical for regulating neuronal and synaptic architecture. Further studies along these lines might uncover novel biomarkers and drug targets for use in drug discovery.
Collapse
Affiliation(s)
- Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.,UNICAMP's Neurobiology Center, Campinas, Brazil
| | - Juliana S Cassoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Juliana M Nascimento
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.,D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Kenneth Hensley
- Department of Pathology, University of Toledo, Toledo, OH, USA.,Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Andres M Pinzon-Velasco
- Bioinformatics and Computational Systems Biology Group, Institute for Genetics, National University of Colombia, Bogotá, Colombia
| | - Christoph W Turck
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
28
|
CRMPs: critical molecules for neurite morphogenesis and neuropsychiatric diseases. Mol Psychiatry 2015; 20:1037-45. [PMID: 26077693 DOI: 10.1038/mp.2015.77] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 04/29/2015] [Accepted: 05/08/2015] [Indexed: 12/11/2022]
Abstract
Neuronal polarity and spatial rearrangement of neuronal processes are central to the development of all mature nervous systems. Recent studies have highlighted the dynamic expression of Collapsin-Response-Mediator Proteins (CRMPs) in neuronal dendritic/axonal compartments, described their interaction with cytoskeleton proteins, identified their ability to activate L- and N-type voltage-gated calcium channels (VGCCs) and delineated their crucial role as signaling molecules essential for neuron differentiation and neural network development and maintenance. In addition, evidence obtained from genome-wide/genetic linkage/proteomic/translational approaches revealed that CRMP expression is altered in human pathologies including mental (schizophrenia and mood disorders) and neurological (Alzheimer's, prion encephalopathy, epilepsy and others) disorders. Changes in CRMPs levels have been observed after psychotropic treatments, and disrupting CRMP2 binding to calcium channels blocked neuropathic pain. These observations, altogether with those obtained from genetically modified mice targeting individual CRMPs and RNA interference approaches, pave the way for considering CRMPs as potential early disease markers and modulation of their activity as therapeutic strategy for disorders associated with neurite abnormalities.
Collapse
|
29
|
Nascimento JM, Martins-de-Souza D. The proteome of schizophrenia. NPJ SCHIZOPHRENIA 2015; 1:14003. [PMID: 27336025 PMCID: PMC4849438 DOI: 10.1038/npjschz.2014.3] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 12/24/2022]
Abstract
On observing schizophrenia from a clinical point of view up to its molecular basis, one may conclude that this is likely to be one of the most complex human disorders to be characterized in all aspects. Such complexity is the reflex of an intricate combination of genetic and environmental components that influence brain functions since pre-natal neurodevelopment, passing by brain maturation, up to the onset of disease and disease establishment. The perfect function of tissues, organs, systems, and finally the organism depends heavily on the proper functioning of cells. Several lines of evidence, including genetics, genomics, transcriptomics, neuropathology, and pharmacology, have supported the idea that dysfunctional cells are causative to schizophrenia. Together with the above-mentioned techniques, proteomics have been contributing to understanding the biochemical basis of schizophrenia at the cellular and tissue level through the identification of differentially expressed proteins and consequently their biochemical pathways, mostly in the brain tissue but also in other cells. In addition, mass spectrometry-based proteomics have identified and precisely quantified proteins that may serve as biomarker candidates to prognosis, diagnosis, and medication monitoring in peripheral tissue. Here, we review all data produced by proteomic investigation in the last 5 years using tissue and/or cells from schizophrenic patients, focusing on postmortem brain tissue and peripheral blood serum and plasma. This information has provided integrated pictures of the biochemical systems involved in the pathobiology, and has suggested potential biomarkers, and warrant potential targets to alternative treatment therapies to schizophrenia.
Collapse
Affiliation(s)
- Juliana M Nascimento
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| |
Collapse
|
30
|
Ge L, Zhu MM, Yang JY, Wang F, Zhang R, Zhang JH, Shen J, Tian HF, Wu CF. Differential proteomic analysis of the anti-depressive effects of oleamide in a rat chronic mild stress model of depression. Pharmacol Biochem Behav 2015; 131:77-86. [PMID: 25641667 DOI: 10.1016/j.pbb.2015.01.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 01/20/2015] [Accepted: 01/23/2015] [Indexed: 01/12/2023]
Abstract
Depression is a complex psychiatric disorder, and its etiology and pathophysiology are not completely understood. Depression involves changes in many biogenic amine, neuropeptide, and oxidative systems, as well as alterations in neuroendocrine function and immune-inflammatory pathways. Oleamide is a fatty amide which exhibits pharmacological effects leading to hypnosis, sedation, and anti-anxiety effects. In the present study, the chronic mild stress (CMS) model was used to investigate the antidepressant-like activity of oleamide. Rats were exposed to 10weeks of CMS or control conditions and were then subsequently treated with 2weeks of daily oleamide (5mg/kg, i.p.), fluoxetine (10mg/kg, i.p.), or vehicle. Protein extracts from the hippocampus were then collected, and hippocampal maps were generated by way of two-dimensional gel electrophoresis (2-DE). Altered proteins induced by CMS and oleamide were identified through mass spectrometry and database searches. Compared to the control group, the CMS rats exhibited significantly less body weight gain and decreased sucrose consumption. Treatment with oleamide caused a reversal of the CMS-induced deficit in sucrose consumption. In the proteomic analysis, 12 protein spots were selected and identified. CMS increased the levels of adenylate kinase isoenzyme 1 (AK1), nucleoside diphosphate kinase B (NDKB), histidine triad nucleotide-binding protein 1 (HINT1), acyl-protein thioesterase 2 (APT-2), and glutathione S-transferase A4 (GSTA4). Compared to the CMS samples, seven spots changed significantly following treatment with oleamide, including GSTA4, glutathione S-transferase A6 (GSTA6), GTP-binding nuclear protein Ran (Ran-GTP), ATP synthase subunit d, transgelin-3, small ubiquitin-related modifier 2 (SUMO2), and eukaryotic translation initiation factor 5A-1 (eIF5A1). Of these seven proteins, the level of eIF5A1 was up-regulated, whereas the remaining proteins were down-regulated. In conclusion, oleamide has antidepressant-like properties in the CMS rat model. The identification of proteins altered by CMS and oleamide treatment provides support for targeting these proteins in the development of novel therapies for depression.
Collapse
Affiliation(s)
- Lin Ge
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Ming-Ming Zhu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jing-Yu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Fang Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Rong Zhang
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jing-Hai Zhang
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jing Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Central Laboratory, Beijing Cancer Hospital & Institute, Beijing 100142, PR China
| | - Hui-Fang Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Central Laboratory, Beijing Cancer Hospital & Institute, Beijing 100142, PR China
| | - Chun-Fu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
31
|
Mitochondrial dysfunction in schizophrenia: an evolutionary perspective. Hum Genet 2014; 134:13-21. [PMID: 25312050 DOI: 10.1007/s00439-014-1491-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 09/26/2014] [Indexed: 10/24/2022]
Abstract
Schizophrenia (SCZ) is a severe psychiatric illness with a lifetime prevalence of 0.4 %. A disturbance of energy metabolism has been suggested as part of the etiopathogenesis of the disorder. Several lines of evidence have proposed a connection between etiopathogenesis of SCZ and human brain evolution, which was characterized by an increase in the energy requirement, demanding a co-evolution of the mitochondrial system. Mitochondria are key players in brain energy homeostasis and multiple lines of evidence suggest that the system is disrupted in SCZ. In this review, we will describe the current knowledge on pathways/system involved in the human brain evolution as well as the main theories regarding the evolutionary origin of SCZ. We will furthermore discuss the role of mitochondria in the context of brain energy metabolism and its role in the etiopathogenesis of SCZ. Understanding SCZ in the context of human brain evolution opens a new perspective to elucidate pathophysiological mechanisms involved in the origin and/or portions of the complex symptomatology of this severe mental disorder.
Collapse
|
32
|
English JA, Wynne K, Cagney G, Cotter DR. Targeted proteomics for validation of biomarkers in early psychosis. Biol Psychiatry 2014; 76:e7-9. [PMID: 24332930 DOI: 10.1016/j.biopsych.2013.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/06/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Jane A English
- Department of Psychiatry, Royal College of Surgeons in Ireland, ERC Beaumont Hospital, Dublin, Ireland.
| | - Kieran Wynne
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Gerard Cagney
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - David R Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, ERC Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
33
|
Föcking M, Opstelten R, Prickaerts J, Steinbusch HWM, Dunn MJ, van den Hove DLA, Cotter DR. Proteomic investigation of the hippocampus in prenatally stressed mice implicates changes in membrane trafficking, cytoskeletal, and metabolic function. Dev Neurosci 2014; 36:432-42. [PMID: 25138076 DOI: 10.1159/000365327] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 06/17/2014] [Indexed: 11/19/2022] Open
Abstract
Prenatal stress influences the development of the fetal brain and so contributes to the risk of the development of psychiatric disorders in later life. The hippocampus is particularly sensitive to prenatal stress, and robust abnormalities have been described in the hippocampus in schizophrenia and depression. The aim of this study was to determine whether prenatal stress is associated with distinct patterns of differential protein expression in the hippocampus using a validated mouse model. We therefore performed a comparative proteomic study assessing female hippocampal samples from 8 prenatally stressed mice and 8 control mice. Differential protein expression was assessed using 2-dimensional difference in gel electrophoresis and subsequent mass spectrometry. The observed changes in a selected group of differentially expressed proteins were confirmed by Western blotting. In comparison to controls, 47 protein spots (38 individual proteins) were found to be differentially expressed in the hippocampus of prenatally stressed mice. Functional grouping of these proteins revealed that prenatal stress influenced the expression of proteins involved in brain development, cytoskeletal composition, stress response, and energy metabolism. Western blotting was utilized to validate the changes in calretinin, hippocalcin, profilin-1 and the signal-transducing adaptor molecule STAM1. Septin-5 could not be validated via Western blotting due to methodological issues. Closer investigation of the validated proteins also pointed to an interesting role for membrane trafficking deficits mediated by prenatal stress. Our findings demonstrate that prenatal stress leads to altered hippocampal protein expression, implicating numerous molecular pathways that may provide new targets for psychotropic drug development.
Collapse
Affiliation(s)
- Melanie Föcking
- Department of Psychiatry, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | | | | | | | | | | | | |
Collapse
|
34
|
Risk genes for schizophrenia: Translational opportunities for drug discovery. Pharmacol Ther 2014; 143:34-50. [DOI: 10.1016/j.pharmthera.2014.02.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 01/31/2014] [Indexed: 12/11/2022]
|
35
|
Giusti L, Mantua V, Da Valle Y, Ciregia F, Ventroni T, Orsolini G, Donadio E, Giannaccini G, Mauri M, Cassano GB, Lucacchini A. Search for peripheral biomarkers in patients affected by acutely psychotic bipolar disorder: a proteomic approach. MOLECULAR BIOSYSTEMS 2014; 10:1246-54. [PMID: 24554194 DOI: 10.1039/c4mb00068d] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Data on neurobiological mechanisms underlying mood disorders are elusive; the aetiology of such states is multifactorial, including genetic predisposition and environmental factors. Diagnosis is currently being made only on an interview-based methodology. Biological markers, which could improve the current classification, and in perspective, stratify patients on a biological basis into more homogeneous clinically distinct subgroups, are highly needed. We describe here a comparative proteomic analysis of peripheral lymphocytes from patients affected by acute psychotic bipolar disorder (PBD) (n = 15), major depressive episode (MDE) with no personal or family history of psychosis (n = 11), and a group of demographically matched healthy controls (HC) (n = 15). All patients were evaluated by means of Structured Clinical Interview for DSM-IV-Patient version (SCID-I-P), Positive and Negative Symptoms Scale (PANSS), Young Mania Rating Scale (YMRS), Hamilton Anxiety Rating Scale (HAM-A) and Hamilton Depression Rating Scale (HAM-D-17) questionnaires. Blood lymphocytes were obtained by gradient separation, and 2-DE was carried out on protein extracts. Significant differences in protein patterns among the three groups were observed. Thirty-six protein spots were found to be differentially expressed in patients compared to controls, which collapsed into 25 different proteins after mass spectrometry identification. Twenty-one of these proteins failed to discriminate between PBD and MDE, suggesting common signatures for these disorders. Nevertheless, after the western blot validation only two of the remaining proteins, namely LIM and SH3 domain protein1, and short-chain specific acyl-CoA dehydrogenase mitochondrial protein, resulted in being significantly upregulated in PBD samples suggesting additional mechanisms that could be associated with the psychotic features of bipolar disorder.
Collapse
Affiliation(s)
- Laura Giusti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Iqbal J, Li W, Hasan M, Juan Li Y, Ullah K, Yun W, Awan U, Qing H, Deng Y. Distortion of homeostatic signaling proteins by simulated microgravity in rat hypothalamus: A16
O/18
O-labeled comparative integrated proteomic approach. Proteomics 2014; 14:262-73. [DOI: 10.1002/pmic.201300337] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/21/2013] [Accepted: 11/11/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Javed Iqbal
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| | - Wang Li
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| | - Murtaza Hasan
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| | - Yu Juan Li
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| | - Kaleem Ullah
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| | - Wang Yun
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| | - Umer Awan
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| | - Hong Qing
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| | - Yulin Deng
- School of Life Sciences; Beijing Institute of Technology; Beijing P. R. China
| |
Collapse
|
37
|
Patel S. Role of Proteomics in Biomarker Discovery. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2014; 94:39-75. [DOI: 10.1016/b978-0-12-800168-4.00003-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
38
|
The potential of biomarkers in psychiatry: focus on proteomics. J Neural Transm (Vienna) 2013; 122 Suppl 1:S9-18. [DOI: 10.1007/s00702-013-1134-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/02/2013] [Indexed: 02/06/2023]
|
39
|
Jha MK, Seo M, Kim JH, Kim BG, Cho JY, Suk K. The secretome signature of reactive glial cells and its pathological implications. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2418-28. [PMID: 23269363 DOI: 10.1016/j.bbapap.2012.12.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 11/23/2012] [Accepted: 12/12/2012] [Indexed: 12/12/2022]
|
40
|
Schmitt A, Turck CW, Pilz PK, Malchow B, von Wilmsdorff M, Falkai P, Martins-de-Souza D. Proteomic similarities between heterozygous reeler mice and schizophrenia. Biol Psychiatry 2013; 74:e5-e10. [PMID: 23684383 DOI: 10.1016/j.biopsych.2013.03.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 03/26/2013] [Indexed: 12/15/2022]
|
41
|
Volle J, Brocard J, Saoud M, Gory-Faure S, Brunelin J, Andrieux A, Suaud-Chagny MF. Reduced expression of STOP/MAP6 in mice leads to cognitive deficits. Schizophr Bull 2013; 39:969-78. [PMID: 23002183 PMCID: PMC3756782 DOI: 10.1093/schbul/sbs113] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND STOP/MAP6 null (KO) mice recapitulate behavioral abnormalities related to positive and negative symptoms and cognitive deficits of schizophrenia. Here, we investigated whether decreased expression of STOP/MAP6 proteins in heterozygous mice (only one allele expressed) would result in abnormal behavior related to those displayed by STOP null mice. METHODS Using a comprehensive test battery, we investigated the behavioral phenotype of STOP heterozygous (Het) mice compared with STOP KO and wild type (WT) mice on animals raised either in standard conditions (controls) or submitted to maternal deprivation. RESULTS Control Het mice displayed prominent deficits in social interaction and learning, resembling KO mice. In contrast, they exhibited short-lasting locomotor hyperreactivity to acute mild stress and no impaired locomotor response to amphetamine, much like WT mice. Additionally, perinatal stress deteriorated Het mouse phenotype by exacerbating alterations related to positive symptoms such as their locomotor reactivity to acute mild stress and psychostimulant challenge. CONCLUSION Results show that the dosage of susceptibility genes modulates their putative phenotypic contribution and that STOP expression has a high penetrance on cognitive abilities. Hence, STOP Het mice might be useful to investigate cognitive defects related to those observed in mental diseases and ultimately might be a valuable experimental model to evaluate preventive treatments.
Collapse
Affiliation(s)
- Julien Volle
- Université de Lyon, Lyon, F-69003, France; Université Lyon 1, Lyon, EA 4615
| | - Jacques Brocard
- Institut National de la Santé et de la Recherche Médicale Unité 836, Institut des Neurosciences de Grenoble, Université Joseph Fourier, 38042 Grenoble Cedex 9, France;,Groupe Physiopathologie du Cytosquelette, Institut de Recherches en Technologies et Sciences pour le Vivant Direction des Sciences du Vivant, Commissariat à l’Énergie Atomique, 38054 Grenoble Cedex 9, France
| | - Mohamed Saoud
- Université de Lyon, Lyon, F-69003, France; Université Lyon 1, Lyon, EA 4615;,Centre Hospitalier le Vinatier, F-69677 Bron Cedex, France
| | - Sylvie Gory-Faure
- Institut National de la Santé et de la Recherche Médicale Unité 836, Institut des Neurosciences de Grenoble, Université Joseph Fourier, 38042 Grenoble Cedex 9, France;,Groupe Physiopathologie du Cytosquelette, Institut de Recherches en Technologies et Sciences pour le Vivant Direction des Sciences du Vivant, Commissariat à l’Énergie Atomique, 38054 Grenoble Cedex 9, France
| | - Jérôme Brunelin
- Université de Lyon, Lyon, F-69003, France; Université Lyon 1, Lyon, EA 4615;,Centre Hospitalier le Vinatier, F-69677 Bron Cedex, France
| | - Annie Andrieux
- Institut National de la Santé et de la Recherche Médicale Unité 836, Institut des Neurosciences de Grenoble, Université Joseph Fourier, 38042 Grenoble Cedex 9, France;,Groupe Physiopathologie du Cytosquelette, Institut de Recherches en Technologies et Sciences pour le Vivant Direction des Sciences du Vivant, Commissariat à l’Énergie Atomique, 38054 Grenoble Cedex 9, France
| | - Marie-Françoise Suaud-Chagny
- Université de Lyon, Lyon, F-69003, France; Université Lyon 1, Lyon, EA 4615;,Centre Hospitalier le Vinatier, F-69677 Bron Cedex, France;,To whom correspondence should be addressed; EA 4615, Pôle Est - Pr d’Amato, CH le vinatier, 95 bd Pinel, 69677 Bron cedex, France; tel: +33 4 37 91 55 65, fax: +33 4 37 91 55 49, e-mail:
| |
Collapse
|
42
|
Cole AR. Glycogen synthase kinase 3 substrates in mood disorders and schizophrenia. FEBS J 2013; 280:5213-27. [DOI: 10.1111/febs.12407] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/12/2013] [Accepted: 06/13/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Adam R. Cole
- Neurosignalling Group; Garvan Institute of Medical Research; Sydney Australia
| |
Collapse
|
43
|
Lefèvre J, Savarin P, Gans P, Hamon L, Clément MJ, David MO, Bosc C, Andrieux A, Curmi PA. Structural basis for the association of MAP6 protein with microtubules and its regulation by calmodulin. J Biol Chem 2013; 288:24910-22. [PMID: 23831686 DOI: 10.1074/jbc.m113.457267] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Microtubules are highly dynamic αβ-tubulin polymers. In vitro and in living cells, microtubules are most often cold- and nocodazole-sensitive. When present, the MAP6/STOP family of proteins protects microtubules from cold- and nocodazole-induced depolymerization but the molecular and structure determinants by which these proteins stabilize microtubules remain under debate. We show here that a short protein fragment from MAP6-N, which encompasses its Mn1 and Mn2 modules (MAP6(90-177)), recapitulates the function of the full-length MAP6-N protein toward microtubules, i.e. its ability to stabilize microtubules in vitro and in cultured cells in ice-cold conditions or in the presence of nocodazole. We further show for the first time, using biochemical assays and NMR spectroscopy, that these effects result from the binding of MAP6(90-177) to microtubules with a 1:1 MAP6(90-177):tubulin heterodimer stoichiometry. NMR data demonstrate that the binding of MAP6(90-177) to microtubules involve its two Mn modules but that a single one is also able to interact with microtubules in a closely similar manner. This suggests that the Mn modules represent each a full microtubule binding domain and that MAP6 proteins may stabilize microtubules by bridging tubulin heterodimers from adjacent protofilaments or within a protofilament. Finally, we demonstrate that Ca(2+)-calmodulin competes with microtubules for MAP6(90-177) binding and that the binding mode of MAP6(90-177) to microtubules and Ca(2+)-calmodulin involves a common stretch of amino acid residues on the MAP6(90-177) side. This result accounts for the regulation of microtubule stability in cold condition by Ca(2+)-calmodulin.
Collapse
Affiliation(s)
- Julien Lefèvre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR829, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université Evry-Val d'Essonne, Evry 91025, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Craft GE, Chen A, Nairn AC. Recent advances in quantitative neuroproteomics. Methods 2013; 61:186-218. [PMID: 23623823 PMCID: PMC3891841 DOI: 10.1016/j.ymeth.2013.04.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 03/29/2013] [Accepted: 04/13/2013] [Indexed: 01/07/2023] Open
Abstract
The field of proteomics is undergoing rapid development in a number of different areas including improvements in mass spectrometric platforms, peptide identification algorithms and bioinformatics. In particular, new and/or improved approaches have established robust methods that not only allow for in-depth and accurate peptide and protein identification and modification, but also allow for sensitive measurement of relative or absolute quantitation. These methods are beginning to be applied to the area of neuroproteomics, but the central nervous system poses many specific challenges in terms of quantitative proteomics, given the large number of different neuronal cell types that are intermixed and that exhibit distinct patterns of gene and protein expression. This review highlights the recent advances that have been made in quantitative neuroproteomics, with a focus on work published over the last five years that applies emerging methods to normal brain function as well as to various neuropsychiatric disorders including schizophrenia and drug addiction as well as of neurodegenerative diseases including Parkinson's disease and Alzheimer's disease. While older methods such as two-dimensional polyacrylamide electrophoresis continued to be used, a variety of more in-depth MS-based approaches including both label (ICAT, iTRAQ, TMT, SILAC, SILAM), label-free (label-free, MRM, SWATH) and absolute quantification methods, are rapidly being applied to neurobiological investigations of normal and diseased brain tissue as well as of cerebrospinal fluid (CSF). While the biological implications of many of these studies remain to be clearly established, that there is a clear need for standardization of experimental design and data analysis, and that the analysis of protein changes in specific neuronal cell types in the central nervous system remains a serious challenge, it appears that the quality and depth of the more recent quantitative proteomics studies is beginning to shed light on a number of aspects of neuroscience that relates to normal brain function as well as of the changes in protein expression and regulation that occurs in neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- George E Craft
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
| | - Anshu Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
- Yale/NIDA Neuroproteomics Center, Yale University School of Medicine, New Haven, CT, 06508
| |
Collapse
|
45
|
Smith KM, Renshaw PF, Bilello J. The diagnosis of depression: current and emerging methods. Compr Psychiatry 2013; 54:1-6. [PMID: 22901834 PMCID: PMC5502713 DOI: 10.1016/j.comppsych.2012.06.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 06/07/2012] [Accepted: 06/11/2012] [Indexed: 10/28/2022] Open
Abstract
Depression is one of the leading causes of disability in adolescents and adults, particularly starting from age 15 years and older. Diagnosis of depression has traditionally been made based on clinical criteria, including patient current symptoms and history. This process is widely used but relies on subjective interpretation. To standardize both the data obtained and data interpretation, various interview-based instruments and noninterview methods exist for screening and testing for depression in various clinical settings. This article evaluates the technical basis for and clinical performance of these various instruments and methods to diagnosis depression in clinical settings. Traditional tools include physician-administered or patient self-administered interview tools that have reasonable clinical accuracy depending on the threshold score and may lead to a full diagnostic evaluation for high-risk patients. In addition, older laboratory methods such as the dexamethasone test have contributed to the diagnosis of depression over a long period. Newer diagnostic methods such as genomics, proteomics, and metabolomics are technically sophisticated and objective and are beginning to emerge in psychiatry. Although promising, further evaluation of these methods is needed to fully demonstrate their clinical value and accuracy.
Collapse
|
46
|
Schmitt A, Reich-Erkelenz D, Gebicke-Härter P, Falkai P. Estudos transcriptômicos no contexto da conectividade perturbada em esquizofrenia. ACTA ACUST UNITED AC 2012. [DOI: 10.1590/s0101-60832012005000001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Esquizofrenia é uma severa doença neurobiológica com fatores genéticos e ambientais desempenhando um papel na fisiopatologia. Diversas regiões cerebrais têm sido implicadas no processo da doença e estão conectadas em complexos circuitos neuronais. Nos níveis molecular e celular, a conectividade afetada entre essas regiões, envolvendo mielinização disfuncional dos axônios neuronais, bem como as alterações no nível sináptico e metabolismo energético levando a distúrbios na plasticidade sináptica, são os maiores achados em estudos post-mortem. Estudos de microarranjos investigando a expressão gênica contribuíram para os achados de alterações em vias complexas em regiões cerebrais relevantes na esquizofrenia. Além disso, estudos utilizando microdissecção e captura a laser permitiram a investigação da expressão gênica em grupos específicos de neurônios. Entretanto, deve ser mantido em mente que em estudos post-mortem, confusos efeitos de medicação, qualidade de RNAm, bem como capacidade de mecanismos regenerativos neuroplásticos do cérebro em indivíduos com história de vida de esquizofrenia, podem influenciar o complexo padrão de alterações no nível molecular. Apesar dessas limitações, estudos transcriptômicos livres de hipóteses em tecido cerebral de pacientes esquizofrênicos oferecem uma possibilidade única para aprender mais sobre os mecanismos subjacentes, levando a novas ópticas da fisiopatologia da doença.
Collapse
Affiliation(s)
- Andrea Schmitt
- Universidade de Göttingen, Alemanha; Universidade Ludwig Maximilians, Alemanha; Universidade de São Paulo, Brasil
| | | | | | - Peter Falkai
- Universidade de Göttingen, Alemanha; Universidade Ludwig Maximilians, Alemanha
| |
Collapse
|
47
|
Oliveira BM, Martins-de-Souza D. Análise proteômica da esquizofrenia. ARCH CLIN PSYCHIAT 2012. [DOI: 10.1590/s0101-60832012005000004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Valioso conhecimento a respeito de esquizofrenia tem sido gerado recentemente para decifrar sua patobiologia e revelar biomarcadores. Entretanto, esforços ainda são necessários, especialmente se levarmos em conta que essa debilitante desordem mental afeta aproximadamente 30 milhões de pessoas ao redor do mundo. Considerando que esquizofrenia é resultado de uma complexa interação entre fatores ambientais, função genética alterada e expressão proteica diferencial sistemática, a proteômica é provavelmente uma ferramenta adequada ao estudo dessa desordem. Aqui sintetizamos os principais achados em estudos proteômicos e posteriores direções a serem tomadas de forma a melhor compreender a bioquímica da esquizofrenia, bem como revelar biomarcadores.
Collapse
|
48
|
Bisgaard CF, Bak S, Christensen T, Jensen ON, Enghild JJ, Wiborg O. Vesicular signalling and immune modulation as hedonic fingerprints: proteomic profiling in the chronic mild stress depression model. J Psychopharmacol 2012; 26:1569-83. [PMID: 23139383 DOI: 10.1177/0269881112460110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Extensive preclinical research has focused at unravelling the underlying molecular mechanisms leading to depression and recovery. In this study, we investigated the quantitative changes in protein abundance in the ventral hippocampal granular cell layer. We compared different phenotypes from the chronic mild stress (CMS) model of depression using chronic administration with two selective serotonin reuptake inhibitors (SSRIs), escitalopram and sertraline. We isolated granular cells using Laser-Capture Microdissection (LCM) and we identified their regulated proteins using two-dimensional (2D) differential gel electrophoresis (DIGE) and tandem mass spectrometry (MS/MS). The majority of the proteins we identified were enzymes involved in different metabolic activities. Additional proteins were functionally classified as vesicular proteins and immune system proteins. Rab GDP dissociation inhibitor alpha (GDIA) and syntaxin-binding protein 1 (STXB1) were potential markers for stress reactivity. Dynamin 1 (DYN1), glutathione S-transferase omega-1 (GSTO1) and peroxiredoxin (PRDX6) were associated with treatment response. In addition, an imbalance between different post-translationally modified versions of DYN1 and GSTO1 potentially accounted for SSRI treatment refraction. In the present study, we searched for new markers of stress reactivity and treatment response as well as any underlying molecular mechanisms correlating to the development of anhedonia and antidepressant therapy refraction. Our results pointed towards an essential role of post-translational modifications in both vesicular and immune protein systems.
Collapse
Affiliation(s)
- Christina F Bisgaard
- Centre for Psychiatric Research, Institute of Clinical Medicine, Aarhus University Hospital, Risskov, Denmark.
| | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- Daniel Martins-de-Souza
- Max Planck Institute of Psychiatry, Munich, Germany; ,Ludwig-Maximilians-UniversitätMunich, Germany; ,Laboratório de Neurociências (LIM-27), Instituto de Psiquiatria, Faculdade de Medicina da Universidade de Sao PauloBrazil,To whom correspondence should be addressed; Max Planck Institute of Psychiatry Kraepelinstr. 2, D-80804 München; tel: +49 89 30622 630, e-mail:
| |
Collapse
|
50
|
Schubert KO, Föcking M, Prehn JHM, Cotter DR. Hypothesis review: are clathrin-mediated endocytosis and clathrin-dependent membrane and protein trafficking core pathophysiological processes in schizophrenia and bipolar disorder? Mol Psychiatry 2012; 17:669-81. [PMID: 21986877 DOI: 10.1038/mp.2011.123] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Clathrin-mediated endocytosis (CME) is the best-characterized mechanism governing cellular membrane and protein trafficking. In this hypothesis review, we integrate recent evidence implicating CME and related cellular trafficking mechanisms in the pathophysiology of psychotic disorders such as schizophrenia and bipolar disorder. The evidence includes proteomic and genomic findings implicating proteins and genes of the clathrin interactome. Additionally, several important candidate genes for schizophrenia, such as dysbindin, are involved in processes closely linked to CME and membrane trafficking. We discuss that key aspects of psychosis neuropathology such as synaptic dysfunction, white matter changes and aberrant neurodevelopment are all influenced by clathrin-dependent processes, and that other cellular trafficking mechanisms previously linked to psychoses interact with the clathrin interactome in important ways. Furthermore, many antipsychotic drugs have been shown to affect clathrin-interacting proteins. We propose that the targeted pharmacological manipulation of the clathrin interactome may offer fruitful opportunities for novel treatments of schizophrenia.
Collapse
Affiliation(s)
- K O Schubert
- Department of Psychiatry, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Republic of Ireland
| | | | | | | |
Collapse
|