1
|
Akhgari A, Michel TM, Vafaee MS. Dendritic spines and their role in the pathogenesis of neurodevelopmental and neurological disorders. Rev Neurosci 2024; 35:489-502. [PMID: 38440811 DOI: 10.1515/revneuro-2023-0151] [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: 12/06/2023] [Accepted: 02/02/2024] [Indexed: 03/06/2024]
Abstract
Since Cajal introduced dendritic spines in the 19th century, they have attained considerable attention, especially in neuropsychiatric and neurologic disorders. Multiple roles of dendritic spine malfunction and pathology in the progression of various diseases have been reported. Thus, it is inevitable to consider these structures as new therapeutic targets for treating neuropsychiatric and neurologic disorders such as autism spectrum disorders, schizophrenia, dementia, Down syndrome, etc. Therefore, we attempted to prepare a narrative review of the literature regarding the role of dendritic spines in the pathogenesis of aforementioned diseases and to shed new light on their pathophysiology.
Collapse
Affiliation(s)
- Aisan Akhgari
- Student Research Committee, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 5166616471, Iran
| | - Tanja Maria Michel
- Research Unit for Psychiatry, Odense University Hospital, J. B. Winsløws Vej 4, Odense 5000, Denmark
- Clinical Institute, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Manouchehr Seyedi Vafaee
- Research Unit for Psychiatry, Odense University Hospital, J. B. Winsløws Vej 4, Odense 5000, Denmark
- Clinical Institute, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| |
Collapse
|
2
|
van Haeringen M, Milaneschi Y, Lamers F, Penninx BW, Jansen R. Dissection of depression heterogeneity using proteomic clusters. Psychol Med 2023; 53:2904-2912. [PMID: 35039097 PMCID: PMC10235664 DOI: 10.1017/s0033291721004888] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 09/23/2021] [Accepted: 11/05/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND The search for relevant biomarkers of major depressive disorder (MDD) is challenged by heterogeneity; biological alterations may vary in patients expressing different symptom profiles. Moreover, most research considers a limited number of biomarkers, which may not be adequate for tagging complex network-level mechanisms. Here we studied clusters of proteins and examined their relation with MDD and individual depressive symptoms. METHODS The sample consisted of 1621 subjects from the Netherlands Study of Depression and Anxiety (NESDA). MDD diagnoses were based on DSM-IV criteria and the Inventory of Depressive Symptomatology questionnaire measured endorsement of 30 symptoms. Serum protein levels were detected using a multi-analyte platform (171 analytes, immunoassay, Myriad RBM DiscoveryMAP 250+). Proteomic clusters were computed using weighted correlation network analysis (WGCNA). RESULTS Six proteomic clusters were identified, of which one was nominally significantly associated with current MDD (p = 9.62E-03, Bonferroni adj. p = 0.057). This cluster contained 21 analytes and was enriched with pathways involved in inflammation and metabolism [including C-reactive protein (CRP), leptin and insulin]. At the individual symptom level, this proteomic cluster was associated with ten symptoms, among which were five atypical, energy-related symptoms. After correcting for several health and lifestyle covariates, hypersomnia, increased appetite, panic and weight gain remained significantly associated with the cluster. CONCLUSIONS Our findings support the idea that alterations in a network of proteins involved in inflammatory and metabolic processes are present in MDD, but these alterations map predominantly to clinical symptoms reflecting an imbalance between energy intake and expenditure.
Collapse
Affiliation(s)
- Marije van Haeringen
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Femke Lamers
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Brenda W.J.H. Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Rick Jansen
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
3
|
de Bartolomeis A, Ciccarelli M, De Simone G, Mazza B, Barone A, Vellucci L. Canonical and Non-Canonical Antipsychotics' Dopamine-Related Mechanisms of Present and Next Generation Molecules: A Systematic Review on Translational Highlights for Treatment Response and Treatment-Resistant Schizophrenia. Int J Mol Sci 2023; 24:ijms24065945. [PMID: 36983018 PMCID: PMC10051989 DOI: 10.3390/ijms24065945] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Schizophrenia is a severe psychiatric illness affecting almost 25 million people worldwide and is conceptualized as a disorder of synaptic plasticity and brain connectivity. Antipsychotics are the primary pharmacological treatment after more than sixty years after their introduction in therapy. Two findings hold true for all presently available antipsychotics. First, all antipsychotics occupy the dopamine D2 receptor (D2R) as an antagonist or partial agonist, even if with different affinity; second, D2R occupancy is the necessary and probably the sufficient mechanism for antipsychotic effect despite the complexity of antipsychotics' receptor profile. D2R occupancy is followed by coincident or divergent intracellular mechanisms, implying the contribution of cAMP regulation, β-arrestin recruitment, and phospholipase A activation, to quote some of the mechanisms considered canonical. However, in recent years, novel mechanisms related to dopamine function beyond or together with D2R occupancy have emerged. Among these potentially non-canonical mechanisms, the role of Na2+ channels at the dopamine at the presynaptic site, dopamine transporter (DAT) involvement as the main regulator of dopamine concentration at synaptic clefts, and the putative role of antipsychotics as chaperones for intracellular D2R sequestration, should be included. These mechanisms expand the fundamental role of dopamine in schizophrenia therapy and may have relevance to considering putatively new strategies for treatment-resistant schizophrenia (TRS), an extremely severe condition epidemiologically relevant and affecting almost 30% of schizophrenia patients. Here, we performed a critical evaluation of the role of antipsychotics in synaptic plasticity, focusing on their canonical and non-canonical mechanisms of action relevant to the treatment of schizophrenia and their subsequent implication for the pathophysiology and potential therapy of TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
4
|
Keshri N, Nandeesha H. Dysregulation of Synaptic Plasticity Markers in Schizophrenia. Indian J Clin Biochem 2023; 38:4-12. [PMID: 36684500 PMCID: PMC9852406 DOI: 10.1007/s12291-022-01068-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/05/2022] [Indexed: 01/25/2023]
Abstract
Schizophrenia is a mental disorder characterized by cognitive impairment resulting in compromised quality of life. Since the regulation of synaptic plasticity has functional implications in various aspects of cognition such as learning, memory, and neural circuit maturation, the dysregulation of synaptic plasticity is considered as a pathobiological feature of schizophrenia. The findings from our recently concluded studies indicate that there is an alteration in levels of synaptic plasticity markers such as neural cell adhesion molecule-1 (NCAM-1), Neurotropin-3 (NT-3) and Matrix-mettaloproteinase-9 (MMP-9) in schizophrenia patients. The objective of the present article is to review the role of markers of synaptic plasticity in schizophrenia. PubMed database (http;//www.ncbi.nlm.nih.gov/pubmed) was used to perform an extensive literature search using the keywords schizophrenia and synaptic plasticity. We conclude that markers of synaptic plasticity are altered in schizophrenia and may lead to complications of schizophrenia including cognitive dysfunction.
Collapse
Affiliation(s)
- Neha Keshri
- Department of Biochemistry, JIPMER, Puducherry, 605006 India
| | | |
Collapse
|
5
|
Paredes R, Ferri F, Seriès P. Influence of E/I balance and pruning in peri-personal space differences in schizophrenia: A computational approach. Schizophr Res 2022; 248:368-377. [PMID: 34509334 DOI: 10.1016/j.schres.2021.06.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/06/2021] [Accepted: 06/22/2021] [Indexed: 11/25/2022]
Abstract
The encoding of the space close to the body, named peri-personal space (PPS), is thought to play a crucial role in the unusual experiences of the self observed in schizophrenia (SCZ). However, it is unclear why SCZ patients and high schizotypal (H-SPQ) individuals present a narrower PPS and why the boundaries of the PPS are more sharply defined in patients. We hypothesise that the unusual PPS representation observed in SCZ is caused by an imbalance of excitation and inhibition (E/I) in recurrent synapses of unisensory neurons or an impairment of bottom-up and top-down connectivity between unisensory and multisensory neurons. These hypotheses were tested computationally by manipulating the effects of E/I imbalance, feedback weights and synaptic density in the network. Using simulations we explored the effects of such impairments in the PPS representation generated by the network and fitted the model to behavioural data. We found that increased excitation of sensory neurons could account for the smaller PPS observed in SCZ and H-SPQ, whereas a decrease of synaptic density caused the sharp definition of the PPS observed in SCZ. We propose a novel conceptual model of PPS representation in the SCZ spectrum that can account for alterations in self-world demarcation, failures in tactile discrimination and symptoms observed in patients.
Collapse
Affiliation(s)
- Renato Paredes
- The University of Edinburgh, School of Informatics, 10 Crichton Street, Edinburgh, United Kingdom; Cognitive Science Group, Instituto de Investigaciones Psicológicas, Facultad de Psicología Universidad Nacional de Córdoba - CONICET, Argentina; Department of Psychology, Pontifical Catholic University of Peru, Lima, Peru
| | - Francesca Ferri
- Department of Neuroscience, Imaging and Clinical Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Peggy Seriès
- The University of Edinburgh, School of Informatics, 10 Crichton Street, Edinburgh, United Kingdom.
| |
Collapse
|
6
|
The glutamate/N-methyl-d-aspartate receptor (NMDAR) model of schizophrenia at 35: On the path from syndrome to disease. Schizophr Res 2022; 242:56-61. [PMID: 35125283 DOI: 10.1016/j.schres.2022.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/07/2023]
|
7
|
Neurogenetics of dynamic connectivity patterns associated with obsessive-compulsive symptoms in healthy children. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 2:411-420. [DOI: 10.1016/j.bpsgos.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/28/2021] [Accepted: 11/14/2021] [Indexed: 01/31/2023] Open
|
8
|
Grent-'t-Jong T, Gajwani R, Gross J, Gumley AI, Krishnadas R, Lawrie SM, Schwannauer M, Schultze-Lutter F, Uhlhaas PJ. 40-Hz Auditory Steady-State Responses Characterize Circuit Dysfunctions and Predict Clinical Outcomes in Clinical High-Risk for Psychosis Participants: A Magnetoencephalography Study. Biol Psychiatry 2021; 90:419-429. [PMID: 34116790 DOI: 10.1016/j.biopsych.2021.03.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/02/2021] [Accepted: 03/17/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND This study aimed to examine whether 40-Hz auditory steady-state responses (ASSRs) are impaired in participants at clinical high-risk for psychosis (CHR-P) and predict clinical outcomes. METHODS Magnetoencephalography data were collected during a 40-Hz ASSR paradigm for a group of 116 CHR-P participants, 33 patients with first-episode psychosis (15 antipsychotic-naïve), a psychosis risk-negative group (n = 38), and 49 healthy control subjects. Analysis of group differences of 40-Hz intertrial phase coherence and 40-Hz amplitude focused on right Heschl's gyrus, superior temporal gyrus, hippocampus, and thalamus after establishing significant activations during 40-Hz ASSR stimulation. Linear regression and linear discriminant analyses were used to predict clinical outcomes in CHR-P participants, including transition to psychosis and persistence of attenuated psychotic symptoms (APSs). RESULTS CHR-P participants and patients with first-episode psychosis were impaired in 40-Hz amplitude in the right thalamus and hippocampus. In addition, patients with first-episode psychosis were impaired in 40-Hz amplitude in the right Heschl's gyrus, and CHR-P participants in 40-Hz intertrial phase coherence in the right Heschl's gyrus. The 40-Hz ASSR deficits were pronounced in CHR-P participants who later transitioned to psychosis (n = 13) or showed persistent APSs (n = 34). Importantly, both APS persistence and transition to psychosis were predicted by 40-Hz ASSR impairments, with ASSR activity in the right hippocampus, superior temporal gyrus, and middle temporal gyrus correctly classifying 69.2% individuals with nonpersistent APSs and 73.5% individuals with persistent APSs (area under the curve = 0.842), and right thalamus 40-Hz activity correctly classifying 76.9% transitioned and 53.6% nontransitioned CHR-P participants (area under the curve = 0.695). CONCLUSIONS Our data indicate that deficits in gamma-band entrainment in the primary auditory cortex and subcortical areas constitute a potential biomarker for predicting clinical outcomes in CHR-P participants.
Collapse
Affiliation(s)
- Tineke Grent-'t-Jong
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom; Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| | - Ruchika Gajwani
- Mental Health and Wellbeing, Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
| | - Joachim Gross
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom; Institute for Biomagnetism and Biosignalanalysis, University of Muenster, Muenster, Germany
| | - Andrew I Gumley
- Mental Health and Wellbeing, Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
| | - Rajeev Krishnadas
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom
| | - Stephen M Lawrie
- Department of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthias Schwannauer
- Department of Clinical Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Frauke Schultze-Lutter
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Department of Psychology and Mental Health, Faculty of Psychology, Airlangga University, Airlangga, Indonesia; University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Peter J Uhlhaas
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom; Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
9
|
Grubisha MJ, Sun X, MacDonald ML, Garver M, Sun Z, Paris KA, Patel DS, DeGiosio RA, Lewis DA, Yates NA, Camacho C, Homanics GE, Ding Y, Sweet RA. MAP2 is differentially phosphorylated in schizophrenia, altering its function. Mol Psychiatry 2021; 26:5371-5388. [PMID: 33526823 PMCID: PMC8325721 DOI: 10.1038/s41380-021-01034-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 01/04/2021] [Accepted: 01/15/2021] [Indexed: 01/30/2023]
Abstract
Schizophrenia (Sz) is a highly polygenic disorder, with common, rare, and structural variants each contributing only a small fraction of overall disease risk. Thus, there is a need to identify downstream points of convergence that can be targeted with therapeutics. Reduction of microtubule-associated protein 2 (MAP2) immunoreactivity (MAP2-IR) is present in individuals with Sz, despite no change in MAP2 protein levels. MAP2 is phosphorylated downstream of multiple receptors and kinases identified as Sz risk genes, altering its immunoreactivity and function. Using an unbiased phosphoproteomics approach, we quantified 18 MAP2 phosphopeptides, 9 of which were significantly altered in Sz subjects. Network analysis grouped MAP2 phosphopeptides into three modules, each with a distinct relationship to dendritic spine loss, synaptic protein levels, and clinical function in Sz subjects. We then investigated the most hyperphosphorylated site in Sz, phosphoserine1782 (pS1782). Computational modeling predicted phosphorylation of S1782 reduces binding of MAP2 to microtubules, which was confirmed experimentally. We generated a transgenic mouse containing a phosphomimetic mutation at S1782 (S1782E) and found reductions in basilar dendritic length and complexity along with reduced spine density. Because only a limited number of MAP2 interacting proteins have been previously identified, we combined co-immunoprecipitation with mass spectrometry to characterize the MAP2 interactome in mouse brain. The MAP2 interactome was enriched for proteins involved in protein translation. These associations were shown to be functional as overexpression of wild type and phosphomimetic MAP2 reduced protein synthesis in vitro. Finally, we found that Sz subjects with low MAP2-IR had reductions in the levels of synaptic proteins relative to nonpsychiatric control (NPC) subjects and to Sz subjects with normal and MAP2-IR, and this same pattern was recapitulated in S1782E mice. These findings suggest a new conceptual framework for Sz-that a large proportion of individuals have a "MAP2opathy"-in which MAP function is altered by phosphorylation, leading to impairments of neuronal structure, synaptic protein synthesis, and function.
Collapse
Affiliation(s)
- M J Grubisha
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - X Sun
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Tsinghua MD Program, School of Medicine, Tsinghua University, Beijing, China
| | - M L MacDonald
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Garver
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Z Sun
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - K A Paris
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - D S Patel
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - R A DeGiosio
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - D A Lewis
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - N A Yates
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - C Camacho
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - G E Homanics
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Y Ding
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - R A Sweet
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Zuo Y, Wei D, Zhu C, Naveed O, Hong W, Yang X. Unveiling the Pathogenesis of Psychiatric Disorders Using Network Models. Genes (Basel) 2021; 12:1101. [PMID: 34356117 PMCID: PMC8304351 DOI: 10.3390/genes12071101] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/13/2023] Open
Abstract
Psychiatric disorders are complex brain disorders with a high degree of genetic heterogeneity, affecting millions of people worldwide. Despite advances in psychiatric genetics, the underlying pathogenic mechanisms of psychiatric disorders are still largely elusive, which impedes the development of novel rational therapies. There has been accumulating evidence suggesting that the genetics of complex disorders can be viewed through an omnigenic lens, which involves contextualizing genes in highly interconnected networks. Thus, applying network-based multi-omics integration methods could cast new light on the pathophysiology of psychiatric disorders. In this review, we first provide an overview of the recent advances in psychiatric genetics and highlight gaps in translating molecular associations into mechanistic insights. We then present an overview of network methodologies and review previous applications of network methods in the study of psychiatric disorders. Lastly, we describe the potential of such methodologies within a multi-tissue, multi-omics approach, and summarize the future directions in adopting diverse network approaches.
Collapse
Affiliation(s)
- Yanning Zuo
- Department of Biological Chemistry, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA; (Y.Z.); (D.W.); (W.H.)
- Department of Neurobiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, CA 90095, USA; (C.Z.); (O.N.)
| | - Don Wei
- Department of Biological Chemistry, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA; (Y.Z.); (D.W.); (W.H.)
- Department of Neurobiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Department of Psychiatry, Semel Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Carissa Zhu
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, CA 90095, USA; (C.Z.); (O.N.)
| | - Ormina Naveed
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, CA 90095, USA; (C.Z.); (O.N.)
| | - Weizhe Hong
- Department of Biological Chemistry, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA; (Y.Z.); (D.W.); (W.H.)
- Department of Neurobiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, CA 90095, USA; (C.Z.); (O.N.)
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Liu S, Wang C, Yang H, Zhu T, Jiang H, Chen J. Weighted gene co-expression network analysis identifies FCER1G as a key gene associated with diabetic kidney disease. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1427. [PMID: 33313172 PMCID: PMC7723642 DOI: 10.21037/atm-20-1087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Diabetic kidney disease (DKD) is the primary cause of end-stage renal disease. However, the pathogenesis of DKD remains unclarified, and there is an urgent need for improved treatments. Recently, many crucial genes closely linked to the molecular mechanism underlying various diseases were discovered using weighted gene co-expression network analysis. Methods We used a gene expression omnibus series dataset GSE104948 with 12 renal glomerular DKD tissue samples and 18 control samples obtained from the gene expression omnibus database and performed weighted gene co-expression network analysis. After obtaining the trait-related modules, gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses of the modules were conducted and the key gene associated with DKD was selected from the top two most significant gene ontology terms using the maximal clique centrality method. Finally, we verified the key gene using protein-protein interaction analysis, additional datasets, and explored the relationship between the key gene and DKD renal function using the Nephroseq v5 online database. Results Among the 10 gene co-expression modules identified, the darkorange2 and red modules were highly related to DKD and the normal biological process, respectively. Majority of the genes in the darkorange2 module were related to immune and inflammatory responses, and potentially related to the progression of DKD due to their abnormal up-regulation. After performing sub-network analysis of the genes extracted from the top two most significant gene ontology terms and calculating the maximal clique centrality values of each gene, FCER1G, located at the center of the protein-protein interaction network, was identified as a key gene related to DKD. Furthermore, gene expression omnibus validation in additional datasets also showed that FCER1G was overexpressed in DKD compared with normal tissues. Finally, Pearson’s correlation analysis between the expression of FCER1G and DKD renal function revealed that the abnormal upregulation of FCER1G was related to diabetic glomerular lesions. Conclusions Our study demonstrated for the first time that FCER1G is a crucial gene associated with the pathogenesis of DKD.
Collapse
Affiliation(s)
- Shanshan Liu
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Cuili Wang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Huiying Yang
- Department of Nephrology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Tingting Zhu
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Hong Jiang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Kidney Disease Immunology Laboratory, the Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou, China.,Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Kukharsky MS, Ninkina NN, An H, Telezhkin V, Wei W, Meritens CRD, Cooper-Knock J, Nakagawa S, Hirose T, Buchman VL, Shelkovnikova TA. Long non-coding RNA Neat1 regulates adaptive behavioural response to stress in mice. Transl Psychiatry 2020; 10:171. [PMID: 32467583 PMCID: PMC7256041 DOI: 10.1038/s41398-020-0854-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/10/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
NEAT1 is a highly and ubiquitously expressed long non-coding RNA (lncRNA) which serves as an important regulator of cellular stress response. However, the physiological role of NEAT1 in the central nervous system (CNS) is still poorly understood. In the current study, we addressed this by characterising the CNS function of the Neat1 knockout mouse model (Neat1-/- mice), using a combination of behavioural phenotyping, electrophysiology and expression analysis. RNAscope® in situ hybridisation revealed that in wild-type mice, Neat1 is expressed across the CNS regions, with high expression in glial cells and low expression in neurons. Loss of Neat1 in mice results in an inadequate reaction to physiological stress manifested as hyperlocomotion and panic escape response. In addition, Neat1-/- mice display deficits in social interaction and rhythmic patterns of activity but retain normal motor function and memory. Neat1-/- mice do not present with neuronal loss, overt neuroinflammation or gross synaptic dysfunction in the brain. However, cultured Neat1-/- neurons are characterised by hyperexcitability and dysregulated calcium homoeostasis, and stress-induced neuronal activity is also augmented in Neat1-/- mice in vivo. Gene expression analysis showed that Neat1 may act as a weak positive regulator of multiple genes in the brain. Furthermore, loss of Neat1 affects alternative splicing of genes important for the CNS function and implicated in neurological diseases. Overall, our data suggest that Neat1 is involved in stress signalling in the brain and fine-tunes the CNS functions to enable adaptive behaviour in response to physiological stress.
Collapse
Affiliation(s)
- Michail S Kukharsky
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
- Institute of Physiologically Active Compounds of Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Natalia N Ninkina
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
- Institute of Physiologically Active Compounds of Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Haiyan An
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
- Medicines Discovery Institute, Cardiff University, Cardiff, CF10 3AT, UK
| | - Vsevolod Telezhkin
- School of Dental Sciences, Newcastle University, Newcastle upon Tyne, NE2 4BW, UK
| | - Wenbin Wei
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK
| | | | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Shinichi Nakagawa
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Tetsuro Hirose
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK
| | - Vladimir L Buchman
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
- Institute of Physiologically Active Compounds of Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Tatyana A Shelkovnikova
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK.
- Institute of Physiologically Active Compounds of Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation.
- Medicines Discovery Institute, Cardiff University, Cardiff, CF10 3AT, UK.
| |
Collapse
|
13
|
Hrovatin K, Kunej T, Dolžan V. Genetic variability of serotonin pathway associated with schizophrenia onset, progression, and treatment. Am J Med Genet B Neuropsychiatr Genet 2020; 183:113-127. [PMID: 31674148 DOI: 10.1002/ajmg.b.32766] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 09/11/2019] [Accepted: 10/07/2019] [Indexed: 12/22/2022]
Abstract
Schizophrenia (SZ) onset and treatment outcome have important genetic components, however individual genes do not have strong effects on SZ phenotype. Therefore, it is important to use the pathway-based approach and study metabolic and signaling pathways, such as dopaminergic and serotonergic. Serotonin pathway has an important role in brain signaling, nevertheless, its role in SZ is not as thoroughly examined as that of dopamine pathway. In this study, we reviewed serotonin pathway genes and genetic variations associated with SZ, including variations at DNA, RNA, and epigenetic level. We obtained 30 serotonin pathway genes from Kyoto encyclopedia of genes and genomes and used these genes for the literature review. We extracted 20 protein coding serotonin pathway genes with genetic variations associated with SZ onset, development, and treatment from 31 research papers. Genes associated with SZ are present on all levels of serotonin pathway: serotonin synthesis, transport, receptor binding, intracellular signaling, and reuptake; however, regulatory genes are poorly researched. We summarized common challenges of genetic association studies and presented some solutions. The analysis of reported serotonin pathway-SZ associations revealed lack of information about certain serotonin pathway genes potentially associated with SZ. Furthermore, it is becoming clear that interactions among serotonin pathway genes and their regulators may bring further knowledge about their involvement in SZ.
Collapse
Affiliation(s)
- Karin Hrovatin
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Ljubljana, Slovenia
| | - Tanja Kunej
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Ljubljana, Slovenia
| | - Vita Dolžan
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Pharmacogenetics Laboratory, Ljubljana, Slovenia
| |
Collapse
|
14
|
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: 2.5] [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
|
15
|
Transcriptome analysis of fibroblasts from schizophrenia patients reveals differential expression of schizophrenia-related genes. Sci Rep 2020; 10:630. [PMID: 31959813 PMCID: PMC6971273 DOI: 10.1038/s41598-020-57467-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/19/2019] [Indexed: 01/05/2023] Open
Abstract
Schizophrenia is a complex neurodevelopmental disorder with high rate of morbidity and mortality. While the heritability rate is high, the precise etiology is still unknown. Although schizophrenia is a central nervous system disorder, studies using peripheral tissues have also been established to search for patient specific biomarkers and to increase understanding of schizophrenia etiology. Among all peripheral tissues, fibroblasts stand out as they are easy to obtain and culture. Furthermore, they keep genetic stability for long period and exhibit molecular similarities to cells from nervous system. Using a unique set of fibroblast samples from a genetically isolated population in northern Sweden, we performed whole transcriptome sequencing to compare differentially expressed genes in seven controls and nine patients. We found differential fibroblast expression between cases and controls for 48 genes, including eight genes previously implicated in schizophrenia or schizophrenia related pathways; HGF, PRRT2, EGR1, EGR3, C11orf87, TLR3, PLEKHH2 and PIK3CD. Weighted gene correlation network analysis identified three differentially co-expressed networks of genes significantly-associated with schizophrenia. All three modules were significantly suppressed in patients compared to control, with one module highly enriched in genes involved in synaptic plasticity, behavior and synaptic transmission. In conclusion, our results support the use of fibroblasts for identification of differentially expressed genes in schizophrenia and highlight dysregulation of synaptic networks as an important mechanism in schizophrenia.
Collapse
|
16
|
Synaptic Plasticity Shapes Brain Connectivity: Implications for Network Topology. Int J Mol Sci 2019; 20:ijms20246193. [PMID: 31817968 PMCID: PMC6940892 DOI: 10.3390/ijms20246193] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
Abstract
Studies of brain network connectivity improved understanding on brain changes and adaptation in response to different pathologies. Synaptic plasticity, the ability of neurons to modify their connections, is involved in brain network remodeling following different types of brain damage (e.g., vascular, neurodegenerative, inflammatory). Although synaptic plasticity mechanisms have been extensively elucidated, how neural plasticity can shape network organization is far from being completely understood. Similarities existing between synaptic plasticity and principles governing brain network organization could be helpful to define brain network properties and reorganization profiles after damage. In this review, we discuss how different forms of synaptic plasticity, including homeostatic and anti-homeostatic mechanisms, could be directly involved in generating specific brain network characteristics. We propose that long-term potentiation could represent the neurophysiological basis for the formation of highly connected nodes (hubs). Conversely, homeostatic plasticity may contribute to stabilize network activity preventing poor and excessive connectivity in the peripheral nodes. In addition, synaptic plasticity dysfunction may drive brain network disruption in neuropsychiatric conditions such as Alzheimer's disease and schizophrenia. Optimal network architecture, characterized by efficient information processing and resilience, and reorganization after damage strictly depend on the balance between these forms of plasticity.
Collapse
|
17
|
Zhang J, Ju S. Identifying genuine protein-protein interactions within communities of gene co-expression networks using a deconvolution method. IET Syst Biol 2019; 13:290-296. [PMID: 31778125 PMCID: PMC8687158 DOI: 10.1049/iet-syb.2019.0060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/24/2019] [Accepted: 07/09/2019] [Indexed: 11/20/2022] Open
Abstract
Direct relationships between biological molecules connected in a gene co-expression network tend to reflect real biological activities such as gene regulation, protein-protein interactions (PPIs), and metabolisation. As correlation-based networks contain numerous indirect connections, those direct relationships are always 'hidden' in them. Compared with the global network, network communities imply more biological significance on predicting protein function, detecting protein complexes and studying network evolution. Therefore, identifying direct relationships in communities is a pervasive and important topic in the biological sciences. Unfortunately, this field has not been well studied. A major thrust of this study is to apply a deconvolution algorithm on communities stemming from different gene co-expression networks, which are constructed by fixing different thresholds for robustness analysis. Using the fifth Dialogue on Reverse Engineering Assessment and Methods challenge (DREAM5) framework, the authors demonstrate that nearly all new communities extracted from a 'deconvolution filter' contain more genuine PPIs than before deconvolution.
Collapse
Affiliation(s)
- Jin Zhang
- School of Information Science and Engineering, University of Jinan, Jinan 250022, People's Republic of China.
| | - Shan Ju
- School of International Trade and Economics, Shandong University of Finance and Economics, Jinan 250014, People's Republic of China
| |
Collapse
|
18
|
Palaniyappan L. Inefficient neural system stabilization: a theory of spontaneous resolutions and recurrent relapses in psychosis. J Psychiatry Neurosci 2019; 44:367-383. [PMID: 31245961 PMCID: PMC6821513 DOI: 10.1503/jpn.180038] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 02/07/2019] [Accepted: 03/05/2019] [Indexed: 12/21/2022] Open
Abstract
A striking feature of psychosis is its heterogeneity. Presentations of psychosis vary from transient symptoms with no functional consequence in the general population to a tenacious illness at the other extreme, with a wide range of variable trajectories in between. Even among patients with schizophrenia, who are diagnosed on the basis of persistent deterioration, marked variation is seen in response to treatment, frequency of relapses and degree of eventual recovery. Existing theoretical accounts of psychosis focus almost exclusively on how symptoms are initially formed, with much less emphasis on explaining their variable course. In this review, I present an account that links several existing notions of the biology of psychosis with the variant clinical trajectories. My aim is to incorporate perspectives of systems neuroscience in a staging framework to explain the individual variations in illness course that follow the onset of psychosis.
Collapse
Affiliation(s)
- Lena Palaniyappan
- From the Department of Psychiatry and Robarts Research Institute, University of Western Ontario and Lawson Health Research Institute, London, Ont., Canada
| |
Collapse
|
19
|
Huang J, Zhuo C, Xu Y, Lin X. Auditory verbal hallucination and the auditory network: From molecules to connectivity. Neuroscience 2019; 410:59-67. [PMID: 31082536 DOI: 10.1016/j.neuroscience.2019.04.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/20/2022]
Abstract
Auditory verbal hallucinations (AVHs) frequently occur across multiple psychiatric diseases especially in schizophrenia (SCZ) patients. Functional imaging studies have revealed the hyperactivity of the auditory cortex and disrupted auditory-verbal network activity underlying AVH etiology. This review will firstly summarize major findings from both human AVH patients and animal models, with focuses on the auditory cortex and associated cortical/sub-cortical areas. Besides mesoscale connectivity or activity data, structure and functions at synaptic level will be discussed, in conjunction with molecular mechanisms. We have summarized major findings for the pathogenesis of AVH in SCZ patients, with focuses in the auditory cortex and prefrontal cortex (PFC). Those discoveries provide explanations for AVH from different perspectives including inter-regional connectivity, local activity in specific areas, structure and functions of synapse, and potentially molecular targets. Due to the uniqueness of AVH in humans, full replica using animals seems impossible. However, we can still extract useful information from animal SCZ models based on the disruption of auditory pathway during AVH episodes. Therefore, we will further interpolate the synaptic structures and molecular targets, whose dysregulation in SCZ models may be highly related with AVH episodes. As the last part, implications for future development of treatment strategies will be discussed.
Collapse
Affiliation(s)
- Jianjie Huang
- Department of Psychiatric-Neuroimging-Genetics Laboratory(PNG-Lab), Wenzhou Seventh People's Hospital, Wenzhou, Zhejiang Province, 325000, China
| | - Chuanjun Zhuo
- Department of Psychiatric-Neuroimging-Genetics Laboratory(PNG-Lab), Wenzhou Seventh People's Hospital, Wenzhou, Zhejiang Province, 325000, China; Department of Psychiatry, Institute of Mental Health, Jining University, Jining Shandong Province, 272191, China; Department of Psychiatric-Neuroimaging-Genetics and Comorbidity Laboratory (PNGC-Lab), Tianjin Mental Health Centre, Mental Health Teaching Hospital of Tianjin Medical University, Tianjin Anding Hospital, China, Tianjin, 300222, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China; MDT Center for Cognitive Impairment and Sleep Disorders, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Yong Xu
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Xiaodong Lin
- Department of Psychiatric-Neuroimging-Genetics Laboratory(PNG-Lab), Wenzhou Seventh People's Hospital, Wenzhou, Zhejiang Province, 325000, China
| |
Collapse
|
20
|
Bimber BN, Yan MY, Peterson SM, Ferguson B. mGAP: the macaque genotype and phenotype resource, a framework for accessing and interpreting macaque variant data, and identifying new models of human disease. BMC Genomics 2019; 20:176. [PMID: 30841849 PMCID: PMC6402181 DOI: 10.1186/s12864-019-5559-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 02/22/2019] [Indexed: 11/17/2022] Open
Abstract
Background Non-human primates (NHPs), particularly macaques, serve as critical and highly relevant pre-clinical models of human disease. The similarity in human and macaque natural disease susceptibility, along with parallel genetic risk alleles, underscores the value of macaques in the development of effective treatment strategies. Nonetheless, there are limited genomic resources available to support the exploration and discovery of macaque models of inherited disease. Notably, there are few public databases tailored to searching NHP sequence variants, and no other database making use of centralized variant calling, or providing genotype-level data and predicted pathogenic effects for each variant. Results The macaque Genotype And Phenotype (mGAP) resource is the first public website providing searchable, annotated macaque variant data. The mGAP resource includes a catalog of high confidence variants, derived from whole genome sequence (WGS). The current mGAP release at time of publication (1.7) contains 17,087,212 variants based on the sequence analysis of 293 rhesus macaques. A custom pipeline was developed to enable annotation of the macaque variants, leveraging human data sources that include regulatory elements (ENCODE, RegulomeDB), known disease- or phenotype-associated variants (GRASP), predicted impact (SIFT, PolyPhen2), and sequence conservation (Phylop, PhastCons). Currently mGAP includes 2767 variants that are identical to alleles listed in the human ClinVar database, of which 276 variants, spanning 258 genes, are identified as pathogenic. An additional 12,472 variants are predicted as high impact (SnpEff) and 13,129 are predicted as damaging (PolyPhen2). In total, these variants are predicted to be associated with more than 2000 human disease or phenotype entries reported in OMIM (Online Mendelian Inheritance in Man). Importantly, mGAP also provides genotype-level data for all subjects, allowing identification of specific individuals harboring alleles of interest. Conclusions The mGAP resource provides variant and genotype data from hundreds of rhesus macaques, processed in a consistent manner across all subjects (https://mgap.ohsu.edu). Together with the extensive variant annotations, mGAP presents unprecedented opportunity to investigate potential genetic associations with currently characterized disease models, and to uncover new macaque models based on parallels with human risk alleles. Electronic supplementary material The online version of this article (10.1186/s12864-019-5559-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Benjamin N Bimber
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA.,Division of Pathobiology, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA
| | - Melissa Y Yan
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA
| | - Samuel M Peterson
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA
| | - Betsy Ferguson
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA. .,Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA. .,Molecular and Medical Genetics Department, Oregon Health and Sciences University, Portland, OR, 97239, USA.
| |
Collapse
|
21
|
MacDonald ML, Favo D, Garver M, Sun Z, Arion D, Ding Y, Yates N, Sweet RA, Lewis DA. Laser capture microdissection-targeted mass spectrometry: a method for multiplexed protein quantification within individual layers of the cerebral cortex. Neuropsychopharmacology 2019; 44:743-748. [PMID: 30390066 PMCID: PMC6372704 DOI: 10.1038/s41386-018-0260-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/17/2018] [Indexed: 11/09/2022]
Abstract
The mammalian neocortex is organized into layers distinguished by the size, packing density, and connectivity of their constituent neurons. Many neuropsychiatric illnesses are complex trait disorders with etiologic factors converging on neuronal protein networks. Cortical pathology of neuropsychiatric diseases, such as schizophrenia, is often restricted to, or more pronounced in, certain cortical layers, suggesting that genetic vulnerabilities manifest with laminar specificity. Thus, the ability to investigate cortical layer-specific protein levels in human postmortem brain is highly desirable. Here, we developed and validated a laser capture microdissection-mass spectrometry (LCM-MS) approach to quantify over 200 proteins in cortical layers 3 and 5 of two cohorts of human subjects as well as a monkey model of postmortem interval. LCM-MS was readily implementable and reliably identified protein patterns that differed between cortical layers 3 and 5. Our findings suggest that LCM-MS facilitates the precise quantification of proteins within individual cortical layers from human postmortem brain tissue, providing a powerful tool in the study of neuropsychiatric disease.
Collapse
Affiliation(s)
- Matthew L MacDonald
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
- Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Daley Favo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Megan Garver
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhe Sun
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dominique Arion
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Ding
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nathan Yates
- Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Venuto S, Castellana S, Monti M, Appolloni I, Fusilli C, Fusco C, Pucci P, Malatesta P, Mazza T, Merla G, Micale L. TRIM8-driven transcriptomic profile of neural stem cells identified glioma-related nodal genes and pathways. Biochim Biophys Acta Gen Subj 2018; 1863:491-501. [PMID: 30528352 DOI: 10.1016/j.bbagen.2018.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/26/2018] [Accepted: 12/03/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND We recently reported TRIM8, encoding an E3 ubiquitin ligase, as a gene aberrantly expressed in glioblastoma whose expression suppresses cell growth and induces a significant reduction of clonogenic potential in glioblastoma cell lines. METHODS we provided novel insights on TRIM8 functions by profiling the transcriptome of TRIM8-expressing primary mouse embryonal neural stem cells by RNA-sequencing and bioinformatic analysis. Functional analysis including luciferase assay, western blot, PCR arrays, Real time quantitative PCR were performed to validate the transcriptomic data. RESULTS Our study identified enriched pathways related to the neurotransmission and to the central nervous system (CNS) functions, including axonal guidance, GABA receptor, Ephrin B, synaptic long-term potentiation/depression, and glutamate receptor signalling pathways. Finally, we provided additional evidence about the existence of a functional interactive crosstalk between TRIM8 and STAT3. CONCLUSIONS Our results substantiate the role of TRIM8 in the brain functions through the dysregulation of genes involved in different CNS-related pathways, including JAK-STAT. GENERAL SIGNIFICANCE This study provides novel insights on the physiological TRIM8 function by profiling for the first time the primary Neural Stem Cell over-expressing TRIM8 by using RNA-Sequencing methodology.
Collapse
Affiliation(s)
- Santina Venuto
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy; Experimental and Regenerative Medicine, University of Foggia, Via A. Gramsci, 89/91, 71122, Foggia, Italy.
| | - Stefano Castellana
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Maria Monti
- CEINGE Advanced Biotechnology, Department of Chemical Sciences, Federico II University, Via Gaetano Salvatore, 486, 80145, Napoli, Italy.
| | - Irene Appolloni
- U.O. Medicina Rigenerativa Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Caterina Fusilli
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Piero Pucci
- CEINGE Advanced Biotechnology, Department of Chemical Sciences, Federico II University, Via Gaetano Salvatore, 486, 80145, Napoli, Italy.
| | - Paolo Malatesta
- U.O. Medicina Rigenerativa Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine (DiMES), University of Genova, Via Leon Battista Alberti, 2, 16132 Genova, Italy.
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Giuseppe Merla
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| |
Collapse
|
23
|
Kondo HM, Pressnitzer D, Shimada Y, Kochiyama T, Kashino M. Inhibition-excitation balance in the parietal cortex modulates volitional control for auditory and visual multistability. Sci Rep 2018; 8:14548. [PMID: 30267021 PMCID: PMC6162284 DOI: 10.1038/s41598-018-32892-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 09/18/2018] [Indexed: 11/25/2022] Open
Abstract
Perceptual organisation must select one interpretation from several alternatives to guide behaviour. Computational models suggest that this could be achieved through an interplay between inhibition and excitation across competing types of neural population coding for each interpretation. Here, to test for such models, we used magnetic resonance spectroscopy to measure non-invasively the concentrations of inhibitory γ-aminobutyric acid (GABA) and excitatory glutamate-glutamine (Glx) in several brain regions. Human participants first performed auditory and visual multistability tasks that produced spontaneous switching between percepts. Then, we observed that longer percept durations during behaviour were associated with higher GABA/Glx ratios in the sensory area coding for each modality. When participants were asked to voluntarily modulate their perception, a common factor across modalities emerged: the GABA/Glx ratio in the posterior parietal cortex tended to be positively correlated with the amount of effective volitional control. Our results provide direct evidence implicating that the balance between neural inhibition and excitation within sensory regions resolves perceptual competition. This powerful computational principle appears to be leveraged by both audition and vision, implemented independently across modalities, but modulated by an integrated control process.
Collapse
Affiliation(s)
- Hirohito M Kondo
- School of Psychology, Chukyo University, Nagoya, Aichi, Japan.
- Human Information Science Laboratory, NTT Communication Science Laboratories, NTT Corporation, Atsugi, Kanagawa, Japan.
| | - Daniel Pressnitzer
- Laboratoire des Systèmes Perceptifs, CNRS UMR 8248, Paris, France
- Département d'Études Cognitive, École Normale Supérieure, Paris, France
| | - Yasuhiro Shimada
- Brain Activity Imaging Center, ATR-Promotions, Seika-cho, Kyoto, Japan
| | - Takanori Kochiyama
- Brain Activity Imaging Center, ATR-Promotions, Seika-cho, Kyoto, Japan
- Department of Cognitive Neuroscience, Advanced Telecommunications Research Institute International, Seika-cho, Kyoto, Japan
| | - Makio Kashino
- Sports Brain Science Project, NTT Communication Science Laboratories, NTT Corporation, Atsugi, Kanagawa, Japan
- School of Engineering, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
24
|
Anodal transcranial direct current stimulation affects auditory cortex plasticity in normal-hearing and noise-exposed rats. Brain Stimul 2018; 11:1008-1023. [DOI: 10.1016/j.brs.2018.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 05/10/2018] [Accepted: 05/28/2018] [Indexed: 12/20/2022] Open
|
25
|
Transcriptome alterations of prefrontal cortical parvalbumin neurons in schizophrenia. Mol Psychiatry 2018; 23:1606-1613. [PMID: 29112193 PMCID: PMC5938166 DOI: 10.1038/mp.2017.216] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/14/2017] [Accepted: 08/30/2017] [Indexed: 12/19/2022]
Abstract
Schizophrenia (SZ) is associated with dysfunction of the dorsolateral prefrontal cortex (DLPFC). This dysfunction is manifest as cognitive deficits that appear to arise from disturbances in gamma frequency oscillations. These oscillations are generated in DLPFC layer 3 (L3) via reciprocal connections between pyramidal cells (PCs) and parvalbumin (PV)-containing interneurons. The density of cortical PV neurons is not altered in SZ, but expression levels of several transcripts involved in PV cell function, including PV, are lower in the disease. However, the transcriptome of PV cells has not been comprehensively assessed in a large cohort of subjects with SZ. In this study, we combined an immunohistochemical approach, laser microdissection, and microarray profiling to analyze the transcriptome of DLPFC L3 PV cells in 36 matched pairs of SZ and unaffected comparison subjects. Over 800 transcripts in PV neurons were identified as differentially expressed in SZ subjects; most of these alterations have not previously been reported. The altered transcripts were enriched for pathways involved in mitochondrial function and tight junction signaling. Comparison with the transcriptome of L3 PCs from the same subjects revealed both shared and distinct disease-related effects on gene expression between cell types. Furthermore, network structures of gene pathways differed across cell types and subject groups. These findings provide new insights into cell type-specific molecular alterations in SZ which may point toward novel strategies for identifying therapeutic targets.
Collapse
|
26
|
Missense variants in ATP1A3 and FXYD gene family are associated with childhood-onset schizophrenia. Mol Psychiatry 2018; 25:821-830. [PMID: 29895895 PMCID: PMC6291354 DOI: 10.1038/s41380-018-0103-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 11/09/2022]
Abstract
Childhood-onset schizophrenia (COS) is a rare and severe form of schizophrenia defined as onset before age of 13. Here we report on two unrelated cases diagnosed with both COS and alternating hemiplegia of childhood (AHC), and for whom two distinct pathogenic de novo variants were identified in the ATP1A3 gene. ATP1A3 encodes the α-subunit of a neuron-specific ATP-dependent transmembrane sodium-potassium pump. Using whole exome sequencing data derived from a cohort of 17 unrelated COS cases, we also examined ATP1A3 and all of its interactors known to be expressed in the brain to establish if variants could be identified. This led to the identification of a third case with a possibly damaging missense mutation in ATP1A3 and three others cases with predicted pathogenic missense variants in the FXYD gene family (FXYD1, FXYD6, and FXYD6-FXYD2 readthrough). FXYD genes encode proteins that modulate the ATP-dependant pump function. This report is the first to identify variants in the same pathway for COS. Our COS study illustrates the interest of stratifying a complex condition according to the age of onset for the identification of deleterious variants. Whereas ATP1A3 is a replicated gene in rare neuropediatric diseases, this gene has previously been linked with COS in only one case report. The association with rare variants in FXYD gene family is novel and highlights the interest of exploring these genes in COS as well as in pediatric neurodevelopmental disorders.
Collapse
|
27
|
Nagels A, Cabanis M, Oppel A, Kirner-Veselinovic A, Schales C, Kircher T. S-Ketamine-Induced NMDA Receptor Blockade during Natural Speech Production and Its Implications for Formal Thought Disorder in Schizophrenia: A Pharmaco-fMRI Study. Neuropsychopharmacology 2018; 43:1324-1333. [PMID: 29105665 PMCID: PMC5916352 DOI: 10.1038/npp.2017.270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/14/2017] [Accepted: 10/16/2017] [Indexed: 02/04/2023]
Abstract
Structural and functional changes in the lateral temporal language areas have been related to formal thought disorder (FTD) in schizophrenia. Continuous, natural speech production activates the right lateral temporal lobe in schizophrenia, as opposed to the left in healthy subjects. Positive and negative FTD can be elicited in healthy subjects by glutamatergic NMDA blockade with ketamine. It is unclear whether the glutamate system is related to the reversed hemispheric lateralization during speaking in patients. In a double-blind, crossover, placebo-controlled study, 15 healthy, male, right-handed volunteers overtly described 7 pictures for 3 min each while BOLD signal changes were acquired with fMRI. As a measure of linguistic demand, the number of words within 20 s epochs was correlated with BOLD responses. Participants developed S-ketamine-induced psychotic symptoms, particularly positive FTD. Ketamine vs placebo was associated with enhanced neural responses in the right middle and inferior temporal gyri. Similar to a previous fMRI study in schizophrenia patients vs healthy controls applying the same design, S-ketamine reversed functional lateralization during speech production in healthy subjects. Results demonstrate an association between glutamatergic imbalance, dysactivations in lateral temporal brain areas, and FTD symptom formation.
Collapse
Affiliation(s)
- Arne Nagels
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
- Department of English and Linguistics, Johannes Gutenberg University, Mainz, Germany
| | - Maurice Cabanis
- Department of Psychiatry and Psychotherapy, Social Neuroscience Lab, University of Lübeck, Lübeck, Germany
- Clinic for Addiction Medicine and Addictive Behaviour, Centre for Mental Health, Stuttgart, Germany
| | - Andrea Oppel
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | | | - Christian Schales
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
28
|
Chelini G, Pantazopoulos H, Durning P, Berretta S. The tetrapartite synapse: a key concept in the pathophysiology of schizophrenia. Eur Psychiatry 2018; 50:60-69. [PMID: 29503098 PMCID: PMC5963512 DOI: 10.1016/j.eurpsy.2018.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 02/01/2018] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
Growing evidence points to synaptic pathology as a core component of the pathophysiology of schizophrenia (SZ). Significant reductions of dendritic spine density and altered expression of their structural and molecular components have been reported in several brain regions, suggesting a deficit of synaptic plasticity. Regulation of synaptic plasticity is a complex process, one that requires not only interactions between pre- and post-synaptic terminals, but also glial cells and the extracellular matrix (ECM). Together, these elements are referred to as the ‘tetrapartite synapse’, an emerging concept supported by accumulating evidence for a role of glial cells and the extracellular matrix in regulating structural and functional aspects of synaptic plasticity. In particular, chondroitin sulfate proteoglycans (CSPGs), one of the main components of the ECM, have been shown to be synthesized predominantly by glial cells, to form organized perisynaptic aggregates known as perineuronal nets (PNNs), and to modulate synaptic signaling and plasticity during postnatal development and adulthood. Notably, recent findings from our group and others have shown marked CSPG abnormalities in several brain regions of people with SZ. These abnormalities were found to affect specialized ECM structures, including PNNs, as well as glial cells expressing the corresponding CSPGs. The purpose of this review is to bring forth the hypothesis that synaptic pathology in SZ arises from a disruption of the interactions between elements of the tetrapartite synapse.
Collapse
Affiliation(s)
- Gabriele Chelini
- Translational Neuroscience Laboratory, Mclean Hospital, 115 Mill Street, Belmont, MA, 02478 USA; Dept. of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115 USA.
| | - Harry Pantazopoulos
- Translational Neuroscience Laboratory, Mclean Hospital, 115 Mill Street, Belmont, MA, 02478 USA; Dept. of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115 USA.
| | - Peter Durning
- Translational Neuroscience Laboratory, Mclean Hospital, 115 Mill Street, Belmont, MA, 02478 USA.
| | - Sabina Berretta
- Translational Neuroscience Laboratory, Mclean Hospital, 115 Mill Street, Belmont, MA, 02478 USA; Dept. of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115 USA; Program in Neuroscience, Harvard Medical School, 220 Longwood Ave., Boston, MA, 02115 USA.
| |
Collapse
|
29
|
Manes NP, Nita-Lazar A. Application of targeted mass spectrometry in bottom-up proteomics for systems biology research. J Proteomics 2018; 189:75-90. [PMID: 29452276 DOI: 10.1016/j.jprot.2018.02.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/25/2018] [Accepted: 02/07/2018] [Indexed: 02/08/2023]
Abstract
The enormous diversity of proteoforms produces tremendous complexity within cellular proteomes, facilitates intricate networks of molecular interactions, and constitutes a formidable analytical challenge for biomedical researchers. Currently, quantitative whole-proteome profiling often relies on non-targeted liquid chromatography-mass spectrometry (LC-MS), which samples proteoforms broadly, but can suffer from lower accuracy, sensitivity, and reproducibility compared with targeted LC-MS. Recent advances in bottom-up proteomics using targeted LC-MS have enabled previously unachievable identification and quantification of target proteins and posttranslational modifications within complex samples. Consequently, targeted LC-MS is rapidly advancing biomedical research, especially systems biology research in diverse areas that include proteogenomics, interactomics, kinomics, and biological pathway modeling. With the recent development of targeted LC-MS assays for nearly the entire human proteome, targeted LC-MS is positioned to enable quantitative proteomic profiling of unprecedented quality and accessibility to support fundamental and clinical research. Here we review recent applications of bottom-up proteomics using targeted LC-MS for systems biology research. SIGNIFICANCE: Advances in targeted proteomics are rapidly advancing systems biology research. Recent applications include systems-level investigations focused on posttranslational modifications (such as phosphoproteomics), protein conformation, protein-protein interaction, kinomics, proteogenomics, and metabolic and signaling pathways. Notably, absolute quantification of metabolic and signaling pathway proteins has enabled accurate pathway modeling and engineering. Integration of targeted proteomics with other technologies, such as RNA-seq, has facilitated diverse research such as the identification of hundreds of "missing" human proteins (genes and transcripts that appear to encode proteins but direct experimental evidence was lacking).
Collapse
Affiliation(s)
- Nathan P Manes
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aleksandra Nita-Lazar
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Li S, Ma L, Wang Y, Wang X, Li Y, Qin L. Auditory steady-state responses in primary and non-primary regions of the auditory cortex in neonatal ventral hippocampal lesion rats. PLoS One 2018; 13:e0192103. [PMID: 29415081 PMCID: PMC5802918 DOI: 10.1371/journal.pone.0192103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 01/18/2018] [Indexed: 12/18/2022] Open
Abstract
Auditory steady-state responses (ASSRs) represent the electrophysiological activity of the auditory nervous system in response to a periodic acoustic stimulus. Spectrogram analysis can reveal the frequency and phase information entrained in ASSRs. Clinically, the ASSR is used to detect abnormalities in electroencephalographs obtained from schizophrenia patients, who show reduced power and phase locking of ASSRs. The neonatal ventral hippocampal lesion (NVHL) rat is a widely used model to investigate the neurodevelopmental mechanisms of schizophrenia. It has been established that NVHL rats exhibit several schizophrenia-like behavioral and molecular abnormalities. However, no clear abnormalities in ASSRs have been reported to date. The present study compared ASSRs of adult NVHL and sham-operated rats. We inserted microelectrodes into the primary auditory cortex (A1) or posterior auditory field (PAF) and recorded the local field potential (LFP) in response to 40- and 80-Hz click train stimuli. Spectrogram analysis was performed to obtain the mean trial power (MTP) and phase-locking factor (PLF) of the click train-evoked LFPs. We found that in the control animals, A1 showed a stronger MTP and PLF of ASSR than PAF, and NVHL operation mainly impaired the ASSR in PAF. Analysis of spike activity also indicated that NVHL operation extended the duration of tone-evoked responses in PAF neurons. Our results reveal, for the first time, that NVHL may distinctly influence the neural activities of primary and non-primary fields of the auditory cortex.
Collapse
Affiliation(s)
- Sibin Li
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Lanlan Ma
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Yuchen Wang
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Xuejiao Wang
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Yingzhuo Li
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Ling Qin
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning Province, P. R. China
- * E-mail:
| |
Collapse
|
31
|
Lozupone M, Seripa D, Stella E, La Montagna M, Solfrizzi V, Quaranta N, Veneziani F, Cester A, Sardone R, Bonfiglio C, Giannelli G, Bisceglia P, Bringiotti R, Daniele A, Greco A, Bellomo A, Logroscino G, Panza F. Innovative biomarkers in psychiatric disorders: a major clinical challenge in psychiatry. Expert Rev Proteomics 2017; 14:809-824. [DOI: 10.1080/14789450.2017.1375857] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Davide Seripa
- Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
| | - Eleonora Stella
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maddalena La Montagna
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Italy
| | | | - Federica Veneziani
- Psychiatric Unit, Department of Basic Medicine, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Alberto Cester
- Department of Medicine Organization Geriatric Unit, CDCD, Dolo Hospital, Venezia, Italy
| | - Rodolfo Sardone
- Department of Epidemiology and Biostatistics, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, Bari, Italy
| | - Caterina Bonfiglio
- Department of Epidemiology and Biostatistics, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, Bari, Italy
| | - Gianluigi Giannelli
- Department of Epidemiology and Biostatistics, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, Bari, Italy
| | - Paola Bisceglia
- Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
| | - Roberto Bringiotti
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - Antonio Greco
- Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giancarlo Logroscino
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
- Department of Clinical Research in Neurology, University of Bari Aldo Moro, “Pia Fondazione Cardinale G. Panico”, Lecce, Italy
| | - Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
- Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
- Department of Clinical Research in Neurology, University of Bari Aldo Moro, “Pia Fondazione Cardinale G. Panico”, Lecce, Italy
| |
Collapse
|
32
|
Forsyth JK, Lewis DA. Mapping the Consequences of Impaired Synaptic Plasticity in Schizophrenia through Development: An Integrative Model for Diverse Clinical Features. Trends Cogn Sci 2017; 21:760-778. [PMID: 28754595 DOI: 10.1016/j.tics.2017.06.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/13/2017] [Accepted: 06/09/2017] [Indexed: 01/19/2023]
Abstract
Schizophrenia is associated with alterations in sensory, motor, and cognitive functions that emerge before psychosis onset; identifying pathogenic processes that can account for this multi-faceted phenotype remains a challenge. Accumulating evidence suggests that synaptic plasticity is impaired in schizophrenia. Given the role of synaptic plasticity in learning, memory, and neural circuit maturation, impaired plasticity may underlie many features of the schizophrenia syndrome. Here, we summarize the neurobiology of synaptic plasticity, review evidence that plasticity is impaired in schizophrenia, and explore a framework in which impaired synaptic plasticity interacts with brain maturation to yield the emergence of sensory, motor, cognitive, and psychotic features at different times during development in schizophrenia. Key gaps in the literature and future directions for testing this framework are discussed.
Collapse
Affiliation(s)
- Jennifer K Forsyth
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA.
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Neary JL, Perez SM, Peterson K, Lodge DJ, Carless MA. Comparative analysis of MBD-seq and MeDIP-seq and estimation of gene expression changes in a rodent model of schizophrenia. Genomics 2017; 109:204-213. [PMID: 28365388 PMCID: PMC5526217 DOI: 10.1016/j.ygeno.2017.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/14/2017] [Accepted: 03/26/2017] [Indexed: 12/11/2022]
Abstract
We conducted a comparative study of multiplexed affinity enrichment sequence methodologies (MBD-seq and MeDIP-seq) in a rodent model of schizophrenia, induced by in utero methylazoxymethanol acetate (MAM) exposure. We also examined related gene expression changes using a pooled sample approach. MBD-seq and MeDIP-seq identified 769 and 1771 differentially methylated regions (DMRs) between F2 offspring of MAM-exposed rats and saline control rats, respectively. The assays showed good concordance, with ~56% of MBD-seq-detected DMRs being identified by or proximal to MeDIP-seq DMRs. There was no significant overlap between DMRs and differentially expressed genes, suggesting that DNA methylation regulatory effects may act upon more distal genes, or are too subtle to detect using our approach. Methylation and gene expression gene ontology enrichment analyses identified biological processes important to schizophrenia pathophysiology, including neuron differentiation, prepulse inhibition, amphetamine response, and glutamatergic synaptic transmission regulation, reinforcing the utility of the MAM rodent model for schizophrenia research.
Collapse
Affiliation(s)
- Jennifer L Neary
- Department of Genetics, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX 78227, USA.
| | - Stephanie M Perez
- Department of Pharmacology, Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| | - Kara Peterson
- Department of Genetics, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX 78227, USA.
| | - Daniel J Lodge
- Department of Pharmacology, Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| | - Melanie A Carless
- Department of Genetics, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX 78227, USA.
| |
Collapse
|
34
|
Scifo E, Calza G, Fuhrmann M, Soliymani R, Baumann M, Lalowski M. Recent advances in applying mass spectrometry and systems biology to determine brain dynamics. Expert Rev Proteomics 2017; 14:545-559. [PMID: 28539064 DOI: 10.1080/14789450.2017.1335200] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Neurological disorders encompass various pathologies which disrupt normal brain physiology and function. Poor understanding of their underlying molecular mechanisms and their societal burden argues for the necessity of novel prevention strategies, early diagnostic techniques and alternative treatment options to reduce the scale of their expected increase. Areas covered: This review scrutinizes mass spectrometry based approaches used to investigate brain dynamics in various conditions, including neurodegenerative and neuropsychiatric disorders. Different proteomics workflows for isolation/enrichment of specific cell populations or brain regions, sample processing; mass spectrometry technologies, for differential proteome quantitation, analysis of post-translational modifications and imaging approaches in the brain are critically deliberated. Future directions, including analysis of cellular sub-compartments, targeted MS platforms (selected/parallel reaction monitoring) and use of mass cytometry are also discussed. Expert commentary: Here, we summarize and evaluate current mass spectrometry based approaches for determining brain dynamics in health and diseases states, with a focus on neurological disorders. Furthermore, we provide insight on current trends and new MS technologies with potential to improve this analysis.
Collapse
Affiliation(s)
- Enzo Scifo
- a Department of Psychiatry, and of Pharmacology and Toxicology , University of Toronto, Campbell Family Mental Health Research Institute of CAMH , Toronto , Canada
| | - Giulio Calza
- b Medicum, Meilahti Clinical Proteomics Core Facility, Biochemistry/Developmental Biology, Faculty of Medicine , FI-00014 University of Helsinki , Helsinki , Finland
| | - Martin Fuhrmann
- c Neuroimmunology and Imaging Group , German Center for Neurodegenerative Diseases (DZNE) , Bonn , Germany
| | - Rabah Soliymani
- b Medicum, Meilahti Clinical Proteomics Core Facility, Biochemistry/Developmental Biology, Faculty of Medicine , FI-00014 University of Helsinki , Helsinki , Finland
| | - Marc Baumann
- b Medicum, Meilahti Clinical Proteomics Core Facility, Biochemistry/Developmental Biology, Faculty of Medicine , FI-00014 University of Helsinki , Helsinki , Finland
| | - Maciej Lalowski
- b Medicum, Meilahti Clinical Proteomics Core Facility, Biochemistry/Developmental Biology, Faculty of Medicine , FI-00014 University of Helsinki , Helsinki , Finland
| |
Collapse
|
35
|
MacDonald ML, Alhassan J, Newman JT, Richard M, Gu H, Kelly RM, Sampson AR, Fish KN, Penzes P, Wills ZP, Lewis DA, Sweet RA. Selective Loss of Smaller Spines in Schizophrenia. Am J Psychiatry 2017; 174:586-594. [PMID: 28359200 PMCID: PMC5800878 DOI: 10.1176/appi.ajp.2017.16070814] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Decreased density of dendritic spines in adult schizophrenia subjects has been hypothesized to result from increased pruning of excess synapses in adolescence. In vivo imaging studies have confirmed that synaptic pruning is largely driven by the loss of large or mature synapses. Thus, increased pruning throughout adolescence would likely result in a deficit of large spines in adulthood. Here, the authors examined the density and volume of dendritic spines in deep layer 3 of the auditory cortex of 20 schizophrenia and 20 matched comparison subjects as well as aberrant voltage-gated calcium channel subunit protein expression linked to spine loss. METHOD Primary auditory cortex deep layer 3 spine density and volume was assessed in 20 pairs of schizophrenia and matched comparison subjects in an initial and replication cohort (12 and eight pairs) by immunohistochemistry-confocal microscopy. Targeted mass spectrometry was used to quantify postsynaptic density and voltage-gated calcium channel protein expression. The effect of increased voltage-gated calcium channel subunit protein expression on spine density and volume was assessed in primary rat neuronal culture. RESULTS Only the smallest spines are lost in deep layer 3 of the primary auditory cortex in subjects with schizophrenia, while larger spines are retained. Levels of the tryptic peptide ALFDFLK, found in the schizophrenia risk gene CACNB4, are inversely correlated with the density of smaller, but not larger, spines in schizophrenia subjects. Consistent with this observation, CACNB4 overexpression resulted in a lower density of smaller spines in primary neuronal cultures. CONCLUSIONS These findings require a rethinking of the overpruning hypothesis, demonstrate a link between small spine loss and a schizophrenia risk gene, and should spur more in-depth investigations of the mechanisms that govern new or small spine generation and stabilization under normal conditions as well as how this process is impaired in schizophrenia.
Collapse
Affiliation(s)
- Matthew L. MacDonald
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jamil Alhassan
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jason T. Newman
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Michelle Richard
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Hong Gu
- Department of Statistics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ryan M. Kelly
- Department of Statistics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Alan R. Sampson
- Department of Statistics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kenneth N. Fish
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine
| | - Zachary P. Wills
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - David A. Lewis
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Robert A. Sweet
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA
| |
Collapse
|
36
|
Vella D, Zoppis I, Mauri G, Mauri P, Di Silvestre D. From protein-protein interactions to protein co-expression networks: a new perspective to evaluate large-scale proteomic data. EURASIP JOURNAL ON BIOINFORMATICS & SYSTEMS BIOLOGY 2017; 2017:6. [PMID: 28477207 PMCID: PMC5359264 DOI: 10.1186/s13637-017-0059-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/09/2017] [Indexed: 12/19/2022]
Abstract
The reductionist approach of dissecting biological systems into their constituents has been successful in the first stage of the molecular biology to elucidate the chemical basis of several biological processes. This knowledge helped biologists to understand the complexity of the biological systems evidencing that most biological functions do not arise from individual molecules; thus, realizing that the emergent properties of the biological systems cannot be explained or be predicted by investigating individual molecules without taking into consideration their relations. Thanks to the improvement of the current -omics technologies and the increasing understanding of the molecular relationships, even more studies are evaluating the biological systems through approaches based on graph theory. Genomic and proteomic data are often combined with protein-protein interaction (PPI) networks whose structure is routinely analyzed by algorithms and tools to characterize hubs/bottlenecks and topological, functional, and disease modules. On the other hand, co-expression networks represent a complementary procedure that give the opportunity to evaluate at system level including organisms that lack information on PPIs. Based on these premises, we introduce the reader to the PPI and to the co-expression networks, including aspects of reconstruction and analysis. In particular, the new idea to evaluate large-scale proteomic data by means of co-expression networks will be discussed presenting some examples of application. Their use to infer biological knowledge will be shown, and a special attention will be devoted to the topological and module analysis.
Collapse
Affiliation(s)
- Danila Vella
- Institute for Biomedical Technologies - National Research Council (ITB-CNR), 93 Fratelli Cervi, Segrate, Milan, Italy.,Department of Computer Science, Systems and Communication DiSCo, University of Milano-Bicocca, 336 Viale Sarca, Milan, Italy
| | - Italo Zoppis
- Department of Computer Science, Systems and Communication DiSCo, University of Milano-Bicocca, 336 Viale Sarca, Milan, Italy
| | - Giancarlo Mauri
- Department of Computer Science, Systems and Communication DiSCo, University of Milano-Bicocca, 336 Viale Sarca, Milan, Italy
| | - Pierluigi Mauri
- Institute for Biomedical Technologies - National Research Council (ITB-CNR), 93 Fratelli Cervi, Segrate, Milan, Italy
| | - Dario Di Silvestre
- Institute for Biomedical Technologies - National Research Council (ITB-CNR), 93 Fratelli Cervi, Segrate, Milan, Italy.
| |
Collapse
|
37
|
McKinney B, Ding Y, Lewis DA, Sweet RA. DNA methylation as a putative mechanism for reduced dendritic spine density in the superior temporal gyrus of subjects with schizophrenia. Transl Psychiatry 2017; 7:e1032. [PMID: 28195572 PMCID: PMC5438028 DOI: 10.1038/tp.2016.297] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/13/2016] [Indexed: 01/11/2023] Open
Abstract
Reduced dendritic spine density (DSD) in cortical layer 3 of the superior temporal gyrus (STG), and multiple other brain regions, is consistently observed in postmortem studies of schizophrenia (SZ). Elucidating the molecular mechanisms of this intermediate phenotype holds promise for understanding SZ pathophysiology, identifying SZ treatment targets and developing animal models. DNA methylation (DNAm), the addition of a methyl group to a cytosine nucleotide, regulates gene transcription and is a strong candidate for such a mechanism. We tested the hypothesis that DNAm correlates with DSD in the human STG and that this relationship is disrupted in SZ. We used the Illumina Infinium HumanMethylation450 Beadchip Array to quantify DNAm on a genome-wide scale in the postmortem STG from 22 SZ subjects and matched non-psychiatric control (NPC) subjects; DSD measures were available for 17 of the 22 subject pairs. We found DNAm to correlate with DSD at more sites than expected by chance in NPC, but not SZ, subjects. In addition, we show that the slopes of the linear DNAm-DSD correlations differed between SZ and NPC subjects at more sites than expected by chance. From these data, we identified 2 candidate genes for mediating DSD abnormalities in SZ: brain-specific angiogenesis inhibitor 1-associated protein 2 (BAIAP2) and discs large, Drosophila, homolog of, 1 (DLG1). Together, these data suggest that altered DNAm in SZ may be a mechanism for SZ-related DSD reductions.
Collapse
Affiliation(s)
- B McKinney
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA,Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Y Ding
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - D A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA,Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - R A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA,Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA,Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA,Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA,Department of Psychiatry, Neurology and VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC), Biomedical Science Tower, Room W-1645, 3811 O'Hara Street, Pittsburgh, PA 15213, USA. E-mail:
| |
Collapse
|
38
|
Lopachev AV, Lopacheva OM, Osipova EA, Vladychenskaya EA, Smolyaninova LV, Fedorova TN, Koroleva OV, Akkuratov EE. Ouabain-induced changes in MAP kinase phosphorylation in primary culture of rat cerebellar cells. Cell Biochem Funct 2017; 34:367-77. [PMID: 27338714 DOI: 10.1002/cbf.3199] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 02/06/2023]
Abstract
Cardiotonic steroid (CTS) ouabain is a well-established inhibitor of Na,K-ATPase capable of inducing signalling processes including changes in the activity of the mitogen activated protein kinases (MAPK) in various cell types. With increasing evidence of endogenous CTS in the blood and cerebrospinal fluid, it is of particular interest to study ouabain-induced signalling in neurons, especially the activation of MAPK, because they are the key kinases activated in response to extracellular signals and regulating cell survival, proliferation and apoptosis. In this study we investigated the effect of ouabain on the level of phosphorylation of three MAPK (ERK1/2, JNK and p38) and on cell survival in the primary culture of rat cerebellar cells. Using Western blotting we described the time course and concentration dependence of phosphorylation for ERK1/2, JNK and p38 in response to ouabain. We discovered that ouabain at a concentration of 1 μM does not cause cell death in cultured neurons while it changes the phosphorylation level of the three MAPK: ERK1/2 is phosphorylated transiently, p38 shows sustained phosphorylation, and JNK is dephosphorylated after a long-term incubation. We showed that ERK1/2 phosphorylation increase does not depend on ouabain-induced calcium increase and p38 activation. Changes in p38 phosphorylation, which is independent from ERK1/2 activation, are calcium dependent. Changes in JNK phosphorylation are calcium dependent and also depend on ERK1/2 and p38 activation. Ten-micromolar ouabain leads to cell death, and we conclude that different effects of 1-μM and 10-μM ouabain depend on different ERK1/2 and p38 phosphorylation profiles. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Alexander V Lopachev
- Research Center of Neurology, Moscow, Russia.,Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Olga M Lopacheva
- Research Center of Neurology, Moscow, Russia.,Lomonosov Moscow State University, International Biotechnological Center, Moscow, Russia
| | - Ekaterina A Osipova
- Lomonosov Moscow State University, International Biotechnological Center, Moscow, Russia.,Lomonosov Moscow State University, Faculty of Chemistry, Department of Chemical Enzymology, Moscow, Russia
| | | | - Larisa V Smolyaninova
- Lomonosov Moscow State University, International Biotechnological Center, Moscow, Russia.,Lomonosov Moscow State University, Faculty of Biology, Department of Biochemistry, Moscow, Russia
| | | | - Olga V Koroleva
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Evgeny E Akkuratov
- St. Petersburg State University, Institute of Translational Biomedicine, St. Petersburg, Russia
| |
Collapse
|
39
|
Smedemark-Margulies N, Brownstein CA, Vargas S, Tembulkar SK, Towne MC, Shi J, Gonzalez-Cuevas E, Liu KX, Bilguvar K, Kleiman RJ, Han MJ, Torres A, Berry GT, Yu TW, Beggs AH, Agrawal PB, Gonzalez-Heydrich J. A novel de novo mutation in ATP1A3 and childhood-onset schizophrenia. Cold Spring Harb Mol Case Stud 2016; 2:a001008. [PMID: 27626066 PMCID: PMC5002930 DOI: 10.1101/mcs.a001008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
We describe a child with onset of command auditory hallucinations and behavioral regression at 6 yr of age in the context of longer standing selective mutism, aggression, and mild motor delays. His genetic evaluation included chromosomal microarray analysis and whole-exome sequencing. Sequencing revealed a previously unreported heterozygous de novo mutation c.385G>A in ATP1A3, predicted to result in a p.V129M amino acid change. This gene codes for a neuron-specific isoform of the catalytic α-subunit of the ATP-dependent transmembrane sodium–potassium pump. Heterozygous mutations in this gene have been reported as causing both sporadic and inherited forms of alternating hemiplegia of childhood and rapid-onset dystonia parkinsonism. We discuss the literature on phenotypes associated with known variants in ATP1A3, examine past functional studies of the role of ATP1A3 in neuronal function, and describe a novel clinical presentation associated with mutation of this gene.
Collapse
Affiliation(s)
- Niklas Smedemark-Margulies
- Division of Immunology, Harvard Medical School, Boston, Massachusetts 02115, USA;; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Catherine A Brownstein
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sigella Vargas
- Developmental Neuropsychiatry Research Program, Department of Psychiatry, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Sahil K Tembulkar
- Developmental Neuropsychiatry Research Program, Department of Psychiatry, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Meghan C Towne
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Jiahai Shi
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Elisa Gonzalez-Cuevas
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Kevin X Liu
- Developmental Neuropsychiatry Research Program, Department of Psychiatry, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Kaya Bilguvar
- Department of Genetics, Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Connecticut 06511, USA
| | - Robin J Kleiman
- Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA;; Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Min-Joon Han
- Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA;; Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Alcy Torres
- Division of Pediatric Neurology, Boston Medical Center and Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Gerard T Berry
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Timothy W Yu
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Alan H Beggs
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Pankaj B Agrawal
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA;; Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Joseph Gonzalez-Heydrich
- Developmental Neuropsychiatry Research Program, Department of Psychiatry, Boston Children's Hospital, Boston, Massachusetts 02115, USA;; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
40
|
Giusti L, Ciregia F, Mazzoni MR, Lucacchini A. Proteomics insight into psychiatric disorders: an update on biological fluid biomarkers. Expert Rev Proteomics 2016; 13:941-950. [DOI: 10.1080/14789450.2016.1230499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Laura Giusti
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Federica Ciregia
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
41
|
Nascimento JM, Garcia S, Saia-Cereda VM, Santana AG, Brandao-Teles C, Zuccoli GS, Junqueira DG, Reis-de-Oliveira G, Baldasso PA, Cassoli JS, Martins-de-Souza D. Proteomics and molecular tools for unveiling missing links in the biochemical understanding of schizophrenia. Proteomics Clin Appl 2016; 10:1148-1158. [DOI: 10.1002/prca.201600021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/21/2016] [Accepted: 07/14/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Juliana M. Nascimento
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Sheila Garcia
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Verônica M. Saia-Cereda
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Aline G. Santana
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Caroline Brandao-Teles
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Giuliana S. Zuccoli
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Danielle G. Junqueira
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Guilherme Reis-de-Oliveira
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Paulo A. Baldasso
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Juliana S. Cassoli
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Daniel Martins-de-Souza
- Department of Biochemistry and Tissue Biology; Laboratory of Neuroproteomics; Institute of Biology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| |
Collapse
|
42
|
Zhang C, Lee S, Mardinoglu A, Hua Q. Investigating the Combinatory Effects of Biological Networks on Gene Co-expression. Front Physiol 2016; 7:160. [PMID: 27445830 PMCID: PMC4916787 DOI: 10.3389/fphys.2016.00160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/15/2016] [Indexed: 11/14/2022] Open
Abstract
Co-expressed genes often share similar functions, and gene co-expression networks have been widely used in studying the functionality of gene modules. Previous analysis indicated that genes are more likely to be co-expressed if they are either regulated by the same transcription factors, forming protein complexes or sharing similar topological properties in protein-protein interaction networks. Here, we reconstructed transcriptional regulatory and protein-protein networks for Saccharomyces cerevisiae using well-established databases, and we evaluated their co-expression activities using publically available gene expression data. Based on our network-dependent analysis, we found that genes that were co-regulated in the transcription regulatory networks and shared similar neighbors in the protein-protein networks were more likely to be co-expressed. Moreover, their biological functions were closely related.
Collapse
Affiliation(s)
- Cheng Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology Shanghai, China
| | - Sunjae Lee
- Science for Life Laboratory, KTH-Royal Institute of Technology Stockholm, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of TechnologyStockholm, Sweden; Department of Biology and Biological Engineering, Chalmers University of TechnologyGöteborg, Sweden
| | - Qiang Hua
- State Key Laboratory of Bioreactor Engineering, East China University of Science and TechnologyShanghai, China; Shanghai Collaborative Innovation Center for Biomanufacturing TechnologyShanghai, China
| |
Collapse
|
43
|
Schizophrenia interactome with 504 novel protein-protein interactions. NPJ SCHIZOPHRENIA 2016; 2:16012. [PMID: 27336055 PMCID: PMC4898894 DOI: 10.1038/npjschz.2016.12] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/17/2016] [Accepted: 02/23/2016] [Indexed: 11/29/2022]
Abstract
Genome-wide association studies of schizophrenia (GWAS) have revealed the role of rare and common genetic variants, but the functional effects of the risk variants remain to be understood. Protein interactome-based studies can facilitate the study of molecular mechanisms by which the risk genes relate to schizophrenia (SZ) genesis, but protein–protein interactions (PPIs) are unknown for many of the liability genes. We developed a computational model to discover PPIs, which is found to be highly accurate according to computational evaluations and experimental validations of selected PPIs. We present here, 365 novel PPIs of liability genes identified by the SZ Working Group of the Psychiatric Genomics Consortium (PGC). Seventeen genes that had no previously known interactions have 57 novel interactions by our method. Among the new interactors are 19 drug targets that are targeted by 130 drugs. In addition, we computed 147 novel PPIs of 25 candidate genes investigated in the pre-GWAS era. While there is little overlap between the GWAS genes and the pre-GWAS genes, the interactomes reveal that they largely belong to the same pathways, thus reconciling the apparent disparities between the GWAS and prior gene association studies. The interactome including 504 novel PPIs overall, could motivate other systems biology studies and trials with repurposed drugs. The PPIs are made available on a webserver, called Schizo-Pi at http://severus.dbmi.pitt.edu/schizo-pi with advanced search capabilities.
Collapse
|
44
|
Sweet RA, MacDonald ML, Kirkwood CM, Ding Y, Schempf T, Jones-Laughner J, Kofler J, Ikonomovic MD, Lopez OL, Garver ME, Fitz NF, Koldamova R, Yates NA. Apolipoprotein E*4 (APOE*4) Genotype Is Associated with Altered Levels of Glutamate Signaling Proteins and Synaptic Coexpression Networks in the Prefrontal Cortex in Mild to Moderate Alzheimer Disease. Mol Cell Proteomics 2016; 15:2252-62. [PMID: 27103636 DOI: 10.1074/mcp.m115.056580] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Indexed: 01/26/2023] Open
Abstract
It has been hypothesized that Alzheimer disease (AD) is primarily a disorder of the synapse. However, assessment of the synaptic proteome in AD subjects has been limited to a small number of proteins and often included subjects with end-stage pathology. Protein from prefrontal cortex gray matter of 59 AD subjects with mild to moderate dementia and 12 normal elderly subjects was assayed using targeted mass spectrometry to quantify 191 synaptically expressed proteins. The profile of synaptic protein expression clustered AD subjects into two groups. One of these was characterized by reduced expression of glutamate receptor proteins, significantly increased synaptic protein network coexpression, and associated withApolipoprotein E*4 (APOE*4) carrier status. The second group, by contrast, showed few differences from control subjects. A subset of AD subjects had altered prefrontal cortex synaptic proteostasis for glutamate receptors and their signaling partners. Efforts to therapeutically target glutamate receptors in AD may have outcomes dependent on APOE*4 genotype.
Collapse
Affiliation(s)
- Robert A Sweet
- From the Departments of ‡Psychiatry, **Neurology, and ‡‡VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC) and
| | | | | | | | | | | | | | - Milos D Ikonomovic
- **Neurology, and §§Geriatric Research, Education and Clinical Center (GRECC), VA Pittsburgh Healthcare System, Pittsburgh, PA
| | - Oscar L Lopez
- From the Departments of ‡Psychiatry, **Neurology, and
| | | | - Nicholas F Fitz
- ¶¶Environmental & Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - Radosveta Koldamova
- ¶¶Environmental & Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | | |
Collapse
|
45
|
Wang T, Ren Z, Ding Y, Fang Z, Sun Z, MacDonald ML, Sweet RA, Wang J, Chen W. FastGGM: An Efficient Algorithm for the Inference of Gaussian Graphical Model in Biological Networks. PLoS Comput Biol 2016; 12:e1004755. [PMID: 26872036 PMCID: PMC4752261 DOI: 10.1371/journal.pcbi.1004755] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/14/2016] [Indexed: 11/19/2022] Open
Abstract
Biological networks provide additional information for the analysis of human diseases, beyond the traditional analysis that focuses on single variables. Gaussian graphical model (GGM), a probability model that characterizes the conditional dependence structure of a set of random variables by a graph, has wide applications in the analysis of biological networks, such as inferring interaction or comparing differential networks. However, existing approaches are either not statistically rigorous or are inefficient for high-dimensional data that include tens of thousands of variables for making inference. In this study, we propose an efficient algorithm to implement the estimation of GGM and obtain p-value and confidence interval for each edge in the graph, based on a recent proposal by Ren et al., 2015. Through simulation studies, we demonstrate that the algorithm is faster by several orders of magnitude than the current implemented algorithm for Ren et al. without losing any accuracy. Then, we apply our algorithm to two real data sets: transcriptomic data from a study of childhood asthma and proteomic data from a study of Alzheimer’s disease. We estimate the global gene or protein interaction networks for the disease and healthy samples. The resulting networks reveal interesting interactions and the differential networks between cases and controls show functional relevance to the diseases. In conclusion, we provide a computationally fast algorithm to implement a statistically sound procedure for constructing Gaussian graphical model and making inference with high-dimensional biological data. The algorithm has been implemented in an R package named “FastGGM”. Gaussian graphical model (GGM), a probability model for characterizing conditional dependence among a set of random variables, has been widely used in studying biological networks. It is important and practical to make inference with rigorous statistical properties and high efficiency under a high-dimensional setting, which is common in biological systems that usually contain tens of thousands of molecular elements, such as genes and proteins. This work proposes a novel efficient algorithm, FastGGM, to implement asymptotically normal estimation of large GGM established by Ren et al [1]. It quickly estimates the precision matrix, partial correlations, as well as p-values and confidence intervals for the graph. Simulation studies demonstrate our algorithm outperforms the current algorithm for Ren et al. and algorithms for some other estimation methods, and real data analyses further prove its efficiency in studying biological networks. In conclusion, FastGGM is a statistically sound and computationally fast algorithm for constructing GGM with high-dimensional data. An R package for implementation can be downloaded from http://www.pitt.edu/~wec47/FastGGM.html.
Collapse
Affiliation(s)
- Ting Wang
- Division of Pulmonary Medicine, Allergy and Immunology; Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Zhao Ren
- Department of Statistics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ying Ding
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Zhou Fang
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Zhe Sun
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Matthew L. MacDonald
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
| | - Jieru Wang
- Division of Pulmonary Medicine, Allergy and Immunology; Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Wei Chen
- Division of Pulmonary Medicine, Allergy and Immunology; Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
46
|
In Sickness and in Health: Perineuronal Nets and Synaptic Plasticity in Psychiatric Disorders. Neural Plast 2015; 2016:9847696. [PMID: 26839720 PMCID: PMC4709762 DOI: 10.1155/2016/9847696] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/27/2015] [Indexed: 12/25/2022] Open
Abstract
Rapidly emerging evidence implicates perineuronal nets (PNNs) and extracellular matrix (ECM) molecules that compose or interact with PNNs, in the pathophysiology of several psychiatric disorders. Studies on schizophrenia, autism spectrum disorders, mood disorders, Alzheimer's disease, and epilepsy point to the involvement of ECM molecules such as chondroitin sulfate proteoglycans, Reelin, and matrix metalloproteases, as well as their cell surface receptors. In many of these disorders, PNN abnormalities have also been reported. In the context of the “quadripartite” synapse concept, that is, the functional unit composed of the pre- and postsynaptic terminals, glial processes, and ECM, and of the role that PNNs and ECM molecules play in regulating synaptic functions and plasticity, these findings resonate with one of the most well-replicated aspects of the pathology of psychiatric disorders, that is, synaptic abnormalities. Here we review the evidence for PNN/ECM-related pathology in these disorders, with particular emphasis on schizophrenia, and discuss the hypothesis that such pathology may significantly contribute to synaptic dysfunction.
Collapse
|
47
|
Rodenas-Cuadrado P, Chen XS, Wiegrebe L, Firzlaff U, Vernes SC. A novel approach identifies the first transcriptome networks in bats: a new genetic model for vocal communication. BMC Genomics 2015; 16:836. [PMID: 26490347 PMCID: PMC4618519 DOI: 10.1186/s12864-015-2068-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 10/13/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Bats are able to employ an astonishingly complex vocal repertoire for navigating their environment and conveying social information. A handful of species also show evidence for vocal learning, an extremely rare ability shared only with humans and few other animals. However, despite their potential for the study of vocal communication, bats remain severely understudied at a molecular level. To address this fundamental gap we performed the first transcriptome profiling and genetic interrogation of molecular networks in the brain of a highly vocal bat species, Phyllostomus discolor. RESULTS Gene network analysis typically needs large sample sizes for correct clustering, this can be prohibitive where samples are limited, such as in this study. To overcome this, we developed a novel bioinformatics methodology for identifying robust co-expression gene networks using few samples (N=6). Using this approach, we identified tissue-specific functional gene networks from the bat PAG, a brain region fundamental for mammalian vocalisation. The most highly connected network identified represented a cluster of genes involved in glutamatergic synaptic transmission. Glutamatergic receptors play a significant role in vocalisation from the PAG, suggesting that this gene network may be mechanistically important for vocal-motor control in mammals. CONCLUSION We have developed an innovative approach to cluster co-expressing gene networks and show that it is highly effective in detecting robust functional gene networks with limited sample sizes. Moreover, this work represents the first gene network analysis performed in a bat brain and establishes bats as a novel, tractable model system for understanding the genetics of vocal mammalian communication.
Collapse
Affiliation(s)
- Pedro Rodenas-Cuadrado
- Max Planck Institute for Psycholinguistics, Wundtlaan 1, Nijmegen, 6525 XD, The Netherlands.
| | - Xiaowei Sylvia Chen
- Max Planck Institute for Psycholinguistics, Wundtlaan 1, Nijmegen, 6525 XD, The Netherlands.
| | - Lutz Wiegrebe
- Ludwig-Maximilians-Universität, Division of Neurobiology, Department Biology II, Großhaderner Straße 2, Planegg-Martinsried, Munich, D-82152, Germany.
| | - Uwe Firzlaff
- Lehrstuhl für Zoologie, TU München, Liesel-Beckmann-Str. 4, Freising-Weihenstephan, Munich, 85350, Germany.
| | - Sonja C Vernes
- Max Planck Institute for Psycholinguistics, Wundtlaan 1, Nijmegen, 6525 XD, The Netherlands. .,Donders Centre for Cognitive Neuroimaging, Kapittelweg 29, Nijmegen, 6525 EN, The Netherlands.
| |
Collapse
|
48
|
Effects of glutamate positive modulators on cognitive deficits in schizophrenia: a systematic review and meta-analysis of double-blind randomized controlled trials. Mol Psychiatry 2015; 20:1151-60. [PMID: 26077694 PMCID: PMC5323255 DOI: 10.1038/mp.2015.68] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/16/2015] [Accepted: 04/27/2015] [Indexed: 12/23/2022]
Abstract
Hypofunction of N-methyl-d-aspartate (NMDA) receptors has been proposed to have an important role in the cognitive impairments observed in schizophrenia. Although glutamate modulators may be effective in reversing such difficult-to-treat conditions, the results of individual studies thus far have been inconsistent. We conducted a systematic review and meta-analysis to examine whether glutamate positive modulators have beneficial effects on cognitive functions in patients with schizophrenia. A literature search was conducted to identify double-blind randomized placebo-controlled trials in schizophrenia or related disorders, using Embase, Medline, and PsycINFO (last search: February 2015). The effects of glutamate positive modulators on cognitive deficits were evaluated for overall cognitive function and eight cognitive domains by calculating standardized mean differences (SMDs) between active drugs and placebo added to antipsychotics. Seventeen studies (N=1391) were included. Glutamate positive modulators were not superior to placebo in terms of overall cognitive function (SMD=0.08, 95% confidence interval=-0.06 to 0.23) (11 studies, n=858) nor each of eight cognitive domains (SMDs=-0.03 to 0.11) (n=367-940) in this population. Subgroup analyses by diagnosis (schizophrenia only studies), concomitant antipsychotics, or pathway of drugs to enhance the glutamatergic neurotransmission (glycine allosteric site of NMDA receptors or α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors) suggested no procognitive effect of glutamate positive modulators. Further, no effect was found in individual compounds on cognition. In conclusion, glutamate positive modulators may not be effective in reversing overall cognitive impairments in patients with schizophrenia as adjunctive therapies.
Collapse
|
49
|
Javitt DC, Sweet RA. Auditory dysfunction in schizophrenia: integrating clinical and basic features. Nat Rev Neurosci 2015; 16:535-50. [PMID: 26289573 PMCID: PMC4692466 DOI: 10.1038/nrn4002] [Citation(s) in RCA: 252] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Schizophrenia is a complex neuropsychiatric disorder that is associated with persistent psychosocial disability in affected individuals. Although studies of schizophrenia have traditionally focused on deficits in higher-order processes such as working memory and executive function, there is an increasing realization that, in this disorder, deficits can be found throughout the cortex and are manifest even at the level of early sensory processing. These deficits are highly amenable to translational investigation and represent potential novel targets for clinical intervention. Deficits, moreover, have been linked to specific structural abnormalities in post-mortem auditory cortex tissue from individuals with schizophrenia, providing unique insights into underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Daniel C Javitt
- Division of Experimental Therapeutics, Departments of Psychiatry and Neuroscience, Columbia University College of Physicians and Surgeons, 1051 Riverside Drive, Unit 21, New York, New York 10032, USA
- Program in Cognitive Neuroscience and Schizophrenia, Nathan S. Kline Institute, 140 Old Orangeburg Rd, Orangeburg, New York 10962, USA
| | - Robert A Sweet
- Departments of Psychiatry and Neurology, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, Pennsylvania 15213, USA
- VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System, Research Office Building (151R), University Drive C, Pittsburgh, Pennsylvania 15240, USA
| |
Collapse
|
50
|
Konopaske GT, Coyle JT. Possible compensatory mechanisms for glutamatergic disconnection found in the auditory cortex in schizophrenia. Biol Psychiatry 2015; 77:923-4. [PMID: 25959563 PMCID: PMC4806226 DOI: 10.1016/j.biopsych.2015.03.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 11/25/2022]
Affiliation(s)
- Glenn T. Konopaske
- Mailman Research Center, McLean Hospital, Belmont, MA,
USA,Department of Psychiatry, Harvard Medical School, Boston,
MA, USA
| | - Joseph T. Coyle
- Mailman Research Center, McLean Hospital, Belmont, MA,
USA,Department of Psychiatry, Harvard Medical School, Boston,
MA, USA
| |
Collapse
|