101
|
Cai L, Huang T, Su J, Zhang X, Chen W, Zhang F, He L, Chou KC. Implications of Newly Identified Brain eQTL Genes and Their Interactors in Schizophrenia. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:433-442. [PMID: 30195780 PMCID: PMC6041437 DOI: 10.1016/j.omtn.2018.05.026] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/19/2018] [Accepted: 05/30/2018] [Indexed: 12/21/2022]
Abstract
Schizophrenia (SCZ) is a devastating genetic mental disorder. Identification of the SCZ risk genes in brains is helpful to understand this disease. Thus, we first used the minimum Redundancy-Maximum Relevance (mRMR) approach to integrate the genome-wide sequence analysis results on SCZ and the expression quantitative trait locus (eQTL) data from ten brain tissues to identify the genes related to SCZ. Second, we adopted the variance inflation factor regression algorithm to identify their interacting genes in brains. Third, using multiple analysis methods, we explored and validated their roles. By means of the aforementioned procedures, we have found that (1) the cerebellum may play a crucial role in the pathogenesis of SCZ and (2) ITIH4 may be utilized as a clinical biomarker for the diagnosis of SCZ. These interesting findings may stimulate novel strategy for developing new drugs against SCZ. It has not escaped our notice that the approach reported here is of use for studying many other genome diseases as well.
Collapse
Affiliation(s)
- Lei Cai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai 200240, China; Gordon Life Science Institute, Boston, MA 02478, USA; Shanghai Center for Women and Children's Health, Shanghai 200062, China.
| | - Tao Huang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai 200240, China; Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingjing Su
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Xinxin Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai 200240, China
| | - Wenzhong Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai 200240, China
| | - Fuquan Zhang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi 214015, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai 200240, China; Shanghai Center for Women and Children's Health, Shanghai 200062, China.
| | - Kuo-Chen Chou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai 200240, China; Gordon Life Science Institute, Boston, MA 02478, USA; Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 610054, China; Faculty of Computing and Information Technology in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
102
|
TSPO in diverse CNS pathologies and psychiatric disease: A critical review and a way forward. Pharmacol Ther 2018; 194:44-58. [PMID: 30189290 DOI: 10.1016/j.pharmthera.2018.09.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The use of Translocator Protein 18 kDa (TSPO) as a clinical neuroimaging biomarker of brain injury and neuroinflammation has increased exponentially in the last decade. There has been a furious pace in the development of new radiotracers for TSPO positron emission tomography (PET) imaging and its use has now been extensively described in many neurological and mental disorders. This fast pace of research and the ever-increasing number of new laboratories entering the field often times lack an appreciation of the historical perspective of the field and introduce dogmatic, but unproven facts, related to the underlying neurobiology of the TSPO response to brain injury and neuroinflammation. Paradoxically, while in neurodegenerative disorders and in all types of CNS pathologies brain TSPO levels increase, a new observation in psychiatric disorders such as schizophrenia is decreased brain levels of TSPO measured by PET. The neurobiological bases for this new finding is currently not known, but rigorous experimental design using multiple experimental approaches and careful interpretation of results is critically important to provide the methodological and/or biological underpinnings to this new observation. This review provides a perspective of the early history of validating TSPO as a biomarker of brain injury and neuroinflammation and a critical analysis of controversial topics in the literature related to the cellular sources of the TSPO response. The latter is important in order to provide the correct interpretation of PET studies in neurodegenerative and psychiatric disorders. Furthermore, this review proposes some yet to be explored explanations to new findings in psychiatric disorders and new approaches to quantitatively assess the glial sources of the TSPO response in order to move the field forward.
Collapse
|
103
|
Zhao J, Liu X, Huo C, Zhao T, Ye H. Abnormalities in Prefrontal Cortical Gene Expression Profiles Relevant to Schizophrenia in MK-801-Exposed C57BL/6 Mice. Neuroscience 2018; 390:60-78. [PMID: 30102956 DOI: 10.1016/j.neuroscience.2018.07.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/07/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022]
Abstract
MK-801, a non-competitive NMDA receptor (NMDAR) antagonist, disturbs NMDAR function in rodents and induces psychological and behavioral changes similar to schizophrenia (SCZ). However, the effects of MK-801 treatment on gene expression are largely unknown. Here we performed RNA-sequencing on the prefrontal cortex of MK-801-exposed male mice in order to analyze gene expression and co-expression patterns related to SCZ and to identify mechanisms that underlie the molecular etiology of this disorder. Transcriptome analysis revealed that the differentially expressed genes were more often associated with biological processes that included postsynaptic transmission, immune system process, response to external stimulus and hemostasis. In order to extract comprehensive biological information, we used an approach for biclustering, called FABIA, to simultaneously cluster transcriptomic data across genes and conditions. When combined with analyses using DAVID and STRING databases, we found that co-expression patterns were altered in synapse-related genes and genes central to the mitochondrial network. Abnormal co-expression of genes mediating synaptic vesicle cycling could disturb release, uptake and reuptake of glutamate, and the perturbation in co-expression patterns for mitochondrial respiratory chain complexes was extensive. Our study supports the hypothesis that research using MK-801-exposed male mice as an animal model of SCZ offers important insights into the pathogenesis of SCZ.
Collapse
Affiliation(s)
- Jialu Zhao
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xu Liu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Chunyue Huo
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Yanjing Medical College, Capital Medical University, Beijing 100069, China
| | - Tian Zhao
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Haihong Ye
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Institute for Brain Disorders, Center of Schizophrenia, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
104
|
Chan SHH, Chan JYH. Mitochondria and Reactive Oxygen Species Contribute to Neurogenic Hypertension. Physiology (Bethesda) 2018; 32:308-321. [PMID: 28615314 DOI: 10.1152/physiol.00006.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/05/2017] [Accepted: 04/13/2017] [Indexed: 02/07/2023] Open
Abstract
Beyond its primary role as fuel generators, mitochondria are engaged in a variety of cellular processes, including redox homeostasis. Mitochondrial dysfunction, therefore, may have a profound impact on high-energy-demanding organs such as the brain. Here, we review the roles of mitochondrial biogenesis and bioenergetics, and their associated signaling in cellular redox homeostasis, and illustrate their contributions to the oxidative stress-related neural mechanism of hypertension, focusing on specific brain areas that are involved in the generation or modulation of sympathetic outflows to the cardiovascular system. We also highlight future challenges of research on mitochondrial physiology and pathophysiology.
Collapse
Affiliation(s)
- Samuel H H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Julie Y H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
105
|
De Bastiani MA, Pfaffenseller B, Klamt F. Master Regulators Connectivity Map: A Transcription Factors-Centered Approach to Drug Repositioning. Front Pharmacol 2018; 9:697. [PMID: 30034338 PMCID: PMC6043797 DOI: 10.3389/fphar.2018.00697] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/08/2018] [Indexed: 01/09/2023] Open
Abstract
Drug discovery is a very expensive and time-consuming endeavor. Fortunately, recent omics technologies and Systems Biology approaches introduced interesting new tools to achieve this task, facilitating the repurposing of already known drugs to new therapeutic assignments using gene expression data and bioinformatics. The inherent role of transcription factors in gene expression modulation makes them strong candidates for master regulators of phenotypic transitions. However, transcription factors expression itself usually does not reflect its activity changes due to post-transcriptional modifications and other complications. In this aspect, the use of high-throughput transcriptomic data may be employed to infer transcription factors-targets interactions and assess their activity through co-expression networks, which can be further used to search for drugs capable of reverting the gene expression profile of pathological phenotypes employing the connectivity maps paradigm. Following this idea, we argue that a module-oriented connectivity map approach using transcription factors-centered networks would aid the query for new repositioning candidates. Through a brief case study, we explored this idea in bipolar disorder, retrieving known drugs used in the usual clinical scenario as well as new candidates with potential therapeutic application in this disease. Indeed, the results of the case study indicate just how promising our approach may be to drug repositioning.
Collapse
Affiliation(s)
- Marco A De Bastiani
- Laboratory of Cellular Biochemistry, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,National Institute of Science and Technology for Translational Medicine, Porto Alegre, Brazil
| | - Bianca Pfaffenseller
- Laboratory of Cellular Biochemistry, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Molecular Psychiatry, Clinicas Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Fabio Klamt
- Laboratory of Cellular Biochemistry, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,National Institute of Science and Technology for Translational Medicine, Porto Alegre, Brazil
| |
Collapse
|
106
|
Allen J, Romay-Tallon R, Brymer KJ, Caruncho HJ, Kalynchuk LE. Mitochondria and Mood: Mitochondrial Dysfunction as a Key Player in the Manifestation of Depression. Front Neurosci 2018; 12:386. [PMID: 29928190 PMCID: PMC5997778 DOI: 10.3389/fnins.2018.00386] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022] Open
Abstract
Human and animal studies suggest an intriguing link between mitochondrial diseases and depression. Although depression has historically been linked to alterations in monoaminergic pharmacology and adult hippocampal neurogenesis, new data increasingly implicate broader forms of dampened plasticity, including plasticity within the cell. Mitochondria are the cellular powerhouse of eukaryotic cells, and they also regulate brain function through oxidative stress and apoptosis. In this paper, we make the case that mitochondrial dysfunction could play an important role in the pathophysiology of depression. Alterations in mitochondrial functions such as oxidative phosphorylation (OXPHOS) and membrane polarity, which increase oxidative stress and apoptosis, may precede the development of depressive symptoms. However, the data in relation to antidepressant drug effects are contradictory: some studies reveal they have no effect on mitochondrial function or even potentiate dysfunction, whereas other studies show more beneficial effects. Overall, the data suggest an intriguing link between mitochondrial function and depression that warrants further investigation. Mitochondria could be targeted in the development of novel antidepressant drugs, and specific forms of mitochondrial dysfunction could be identified as biomarkers to personalize treatment and aid in early diagnosis by differentiating between disorders with overlapping symptoms.
Collapse
Affiliation(s)
- Josh Allen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | - Kyle J Brymer
- Department of Psychology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
107
|
Avramopoulos D. Recent Advances in the Genetics of Schizophrenia. MOLECULAR NEUROPSYCHIATRY 2018; 4:35-51. [PMID: 29998117 PMCID: PMC6032037 DOI: 10.1159/000488679] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/21/2018] [Indexed: 12/27/2022]
Abstract
The last decade brought tremendous progress in the field of schizophrenia genetics. As a result of extensive collaborations and multiple technological advances, we now recognize many types of genetic variants that increase the risk. These include large copy number variants, rare coding inherited and de novο variants, and over 100 loci harboring common risk variants. While the type and contribution to the risk vary among genetic variants, there is concordance in the functions of genes they implicate, such as those whose RNA binds the fragile X-related protein FMRP and members of the activity-regulated cytoskeletal complex involved in learning and memory. Gene expression studies add important information on the biology of the disease and recapitulate the same functional gene groups. Studies of alternative phenotypes help us widen our understanding of the genetic architecture of mental function and dysfunction, how diseases overlap not only with each other but also with non-disease phenotypes. The challenge is to apply this new knowledge to prevention and treatment and help patients. The data generated so far and emerging technologies, including new methods in cell engineering, offer significant promise that in the next decade we will unlock the translational potential of these significant discoveries.
Collapse
Affiliation(s)
- Dimitrios Avramopoulos
- Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Psychiatry, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
108
|
Liu J, Su B. Integrated analysis supports ATXN1 as a schizophrenia risk gene. Schizophr Res 2018; 195:298-305. [PMID: 29055568 DOI: 10.1016/j.schres.2017.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/27/2017] [Accepted: 10/08/2017] [Indexed: 12/13/2022]
Abstract
Protein-protein interaction (PPI) is informative in identifying hidden disease risk genes that tend to interact with known risk genes usually working together in the same disease module. With the use of an integrated approach combining PPI information with pathway and expression analysis as well as genome-wide association study (GWAS), we intended to find new risk genes for schizophrenia (SCZ). We showed that ATXN1 was the only direct PPI partner of the know SCZ risk gene ZNF804A, and it also had direct PPIs with other 18 known SCZ risk genes. ATXN1 serves as one of the hub genes in the PPI network containing many known SCZ risk genes, and this network is significantly enriched for the MAPK signaling pathway. Further gene expression analysis indicated that ATXN1 is highly expressed in prefrontal cortex, and SCZ patients had significantly decreased expression compared with healthy controls. Finally, the published GWAS data supports an association of ATXN1 with SCZ as well as other psychiatric disorders though not reaching genome-wide significance. These convergent evidences support ATXN1 as a promising risk gene for SCZ, and the integrated approach serves as a useful tool for dissecting the genetic basis of psychiatric disorders.
Collapse
Affiliation(s)
- Jiewei Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China.
| |
Collapse
|
109
|
Akarsu S, Bolu A, Aydemir E, Zincir SB, Kurt YG, Zincir S, Erdem M, Uzun Ö. The Relationship between the Number of Manic Episodes and Oxidative Stress Indicators in Bipolar Disorder. Psychiatry Investig 2018; 15:514-519. [PMID: 29674601 PMCID: PMC5975995 DOI: 10.30773/pi.2016.12.31] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 12/31/2016] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE Bipolar disorder (BD) is a chronic mood disorder characterized by recurrent episodes that has a lifetime prevalence of 0.4- 5.5%. The neurochemical mechanism of BD is not fully understood. Oxidative stress in neurons causes lipid peroxidation in proteins associated with neuronal membranes and intracellular enzymes and it may lead to dysfunction in neurotransmitter reuptake and enzyme activities. These pathological processes are thought to occur in brain regions associated with affective functions and emotions in BD. The relationship between the number of manic episodes and total oxidant-antioxidant capacity was investigated in this study. METHODS Eighty-two BD patients hospitalized due to manic symptoms and with no episodes of depression were enrolled in the study. Thirty of the 82 patients had had their first episode of mania, and the other 52 patients had had two or more manic episodes. The control group included 45 socio-demographically matched healthy individuals. Serum total antioxidant capacity (TAC) and total oxidant capacity (TOC) measurements of the participants were performed. The oxidative stress index (OSI) was calculated by TOC/TAC. RESULTS There were no significant differences in OSI scores between BD patients with first-episode mania and BD patients with more than one manic episode. However, OSI scores in both groups were significantly higher than in the control group. TOC levels of BD patients with first-episode mania were found to be significantly higher than TOC levels of BD patients with more than one manic episode and healthy controls. There were no significant differences in TAC levels between BD patients with first-episode mania and BD patients with more than one manic episode. TAC levels in both groups were significantly higher than in the control group. CONCLUSION Significant changes in oxidative stress indicators were observed in this study, confirming previous studies. Increased levels of oxidants were shown with increased disease severity rather than with the number of manic episodes. Systematic studies, including of each period of the disorder, are needed for using the findings indicating deterioration of oxidative parameters.
Collapse
Affiliation(s)
- Süleyman Akarsu
- Department of Psychiatry, Freelance Physician, Hatay, Turkey
| | - Abdullah Bolu
- Department of Psychiatry, Gülhane Education and Research Hospital, Ankara, Turkey
| | - Emre Aydemir
- Department of Psychiatry, Beytepe State Hospital, Ankara, Turkey
| | - Selma Bozkurt Zincir
- Department of Psychiatry, Medical Park Göztepe Hospital Complex, Istanbul, Turkey
| | | | - Serkan Zincir
- Department of Psychiatry, Eskişehir State Hospital, Eskişehir, Turkey
| | - Murat Erdem
- Department of Psychiatry, Bilted Psychiatry Treatment Center, Ankara, Turkey
| | - Özcan Uzun
- Department of Psychiatry, Gülhane Education and Research Hospital, Ankara, Turkey
| |
Collapse
|
110
|
Mitochondrial function in individuals at clinical high risk for psychosis. Sci Rep 2018; 8:6216. [PMID: 29670128 PMCID: PMC5906614 DOI: 10.1038/s41598-018-24355-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022] Open
Abstract
Alterations in mitochondrial function have been implicated in the etiology of schizophrenia. Most studies have investigated alterations in mitochondrial function in patients in which the disorder is already established; however, whether mitochondrial dysfunction predates the onset of psychosis remains unknown. We measured peripheral mitochondrial complex (I–V) function and lactate/pyruvate levels in 27 antipsychotic-naïve individuals at clinical high risk for psychosis (CHR) and 16 healthy controls. We also explored the association between mitochondrial function and brain microglial activation and glutathione levels using a translocator protein 18 kDa [18F]FEPPA PET scan and 1H-MRS scan, respectively. There were no significant differences in mitochondrial complex function and lactate/pyruvate levels between CHR and healthy controls. In the CHR group, mitochondrial complex III function (r = −0.51, p = 0.008) and lactate levels (r = 0.61, p = 0.004) were associated with prodromal negative symptoms. As previously reported, there were no significant differences in microglial activation and glutathione levels between groups, however, mitochondrial complex IV function was inversely related to microglial activation in the hippocampus in CHR (r = −0.42, p = 0.04), but not in healthy controls. In conclusion, alterations in mitochondrial function are not yet evident in CHR, but may relate to the severity of prodromal symptoms, particularly negative symptoms.
Collapse
|
111
|
Yang CP, Li X, Wu Y, Shen Q, Zeng Y, Xiong Q, Wei M, Chen C, Liu J, Huo Y, Li K, Xue G, Yao YG, Zhang C, Li M, Chen Y, Luo XJ. Comprehensive integrative analyses identify GLT8D1 and CSNK2B as schizophrenia risk genes. Nat Commun 2018; 9:838. [PMID: 29483533 PMCID: PMC5826945 DOI: 10.1038/s41467-018-03247-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 01/29/2018] [Indexed: 01/01/2023] Open
Abstract
Recent genome-wide association studies (GWAS) have identified multiple risk loci that show strong associations with schizophrenia. However, pinpointing the potential causal genes at the reported loci remains a major challenge. Here we identify candidate causal genes for schizophrenia using an integrative genomic approach. Sherlock integrative analysis shows that ALMS1, GLT8D1, and CSNK2B are schizophrenia risk genes, which are validated using independent brain expression quantitative trait loci (eQTL) data and integrative analysis method (SMR). Consistently, gene expression analysis in schizophrenia cases and controls further supports the potential role of these three genes in the pathogenesis of schizophrenia. Finally, we show that GLT8D1 and CSNK2B knockdown promote the proliferation and inhibit the differentiation abilities of neural stem cells, and alter morphology and synaptic transmission of neurons. These convergent lines of evidence suggest that the ALMS1, CSNK2B, and GLT8D1 genes may be involved in pathophysiology of schizophrenia. More than 100 risk loci for schizophrenia have been identified by genome-wide association studies. Here, the authors apply an integrative genomic approach to prioritize risk genes and validate GLT8D1 and CSNK2B as candidate causal genes by in vitro studies in neural stem cells.
Collapse
Affiliation(s)
- Cui-Ping Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Xiaoyan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Yong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Qiushuo Shen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Yong Zeng
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical College, Kunming, Yunnan, 650031, China
| | - Qiuxia Xiong
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical College, Kunming, Yunnan, 650031, China
| | - Mengping Wei
- State Key Laboratory of Membrane Biology, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Chunhui Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Yongxia Huo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Kaiqin Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Gui Xue
- State Key Laboratory of Cognitive Neuroscience and Learning, and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chen Zhang
- State Key Laboratory of Membrane Biology, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnna, 650223, China.
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnna, 650223, China.
| |
Collapse
|
112
|
Gandal MJ, Haney JR, Parikshak NN, Leppa V, Ramaswami G, Hartl C, Schork AJ, Appadurai V, Buil A, Werge TM, Liu C, White KP, Horvath S, Geschwind DH. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science 2018; 359:693-697. [PMID: 29439242 PMCID: PMC5898828 DOI: 10.1126/science.aad6469] [Citation(s) in RCA: 662] [Impact Index Per Article: 110.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/21/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022]
Abstract
The predisposition to neuropsychiatric disease involves a complex, polygenic, and pleiotropic genetic architecture. However, little is known about how genetic variants impart brain dysfunction or pathology. We used transcriptomic profiling as a quantitative readout of molecular brain-based phenotypes across five major psychiatric disorders-autism, schizophrenia, bipolar disorder, depression, and alcoholism-compared with matched controls. We identified patterns of shared and distinct gene-expression perturbations across these conditions. The degree of sharing of transcriptional dysregulation is related to polygenic (single-nucleotide polymorphism-based) overlap across disorders, suggesting a substantial causal genetic component. This comprehensive systems-level view of the neurobiological architecture of major neuropsychiatric illness demonstrates pathways of molecular convergence and specificity.
Collapse
Affiliation(s)
- Michael J. Gandal
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Psychiatry, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Jillian R. Haney
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Neelroop N. Parikshak
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Virpi Leppa
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Gokul Ramaswami
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Chris Hartl
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Andrew J. Schork
- Institute of Biological Psychiatry, Mental Health Services Copenhagen, Copenhagen, Denmark
| | - Vivek Appadurai
- Institute of Biological Psychiatry, Mental Health Services Copenhagen, Copenhagen, Denmark
| | - Alfonso Buil
- Institute of Biological Psychiatry, Mental Health Services Copenhagen, Copenhagen, Denmark
| | - Thomas M. Werge
- Institute of Biological Psychiatry, Mental Health Services Copenhagen, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Chunyu Liu
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60607, USA
- The State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan, China
| | - Kevin P. White
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
- Tempus Labs, 600 W. Chicago Ave., Chicago IL 60654
| | | | | | | | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Daniel H. Geschwind
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
113
|
Hagihara H, Catts VS, Katayama Y, Shoji H, Takagi T, Huang FL, Nakao A, Mori Y, Huang KP, Ishii S, Graef IA, Nakayama KI, Shannon Weickert C, Miyakawa T. Decreased Brain pH as a Shared Endophenotype of Psychiatric Disorders. Neuropsychopharmacology 2018; 43:459-468. [PMID: 28776581 PMCID: PMC5770757 DOI: 10.1038/npp.2017.167] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/14/2017] [Accepted: 08/01/2017] [Indexed: 01/25/2023]
Abstract
Although the brains of patients with schizophrenia and bipolar disorder exhibit decreased brain pH relative to those of healthy controls upon postmortem examination, it remains controversial whether this finding reflects a primary feature of the diseases or is a result of confounding factors such as medication and agonal state. To date, systematic investigation of brain pH has not been undertaken using animal models that can be studied without confounds inherent in human studies. In the present study, we first reevaluated the pH of the postmortem brains of patients with schizophrenia and bipolar disorder by conducting a meta-analysis of existing data sets from 10 studies. We then measured pH, lactate levels, and related metabolite levels in brain homogenates from five neurodevelopmental mouse models of psychiatric disorders, including schizophrenia, bipolar disorder, and autism spectrum disorder. All mice were drug naive with the same agonal state, postmortem interval, and age within each strain. Our meta-analysis revealed that brain pH was significantly lower in patients with schizophrenia and bipolar disorder than in control participants, even when a few potential confounding factors (postmortem interval, age, and history of antipsychotic use) were considered. In animal experiments, we observed significantly lower pH and higher lactate levels in the brains of model mice relative to controls, as well as a significant negative correlation between pH and lactate levels. Our findings suggest that lower pH associated with increased lactate levels is not a mere artifact, but rather implicated in the underlying pathophysiology of schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Hideo Hagihara
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Vibeke S Catts
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Yuta Katayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Tsuyoshi Takagi
- Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan,RIKEN Tsukuba Institute, Tsukuba, Japan
| | - Freesia L Huang
- Program of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | - Akito Nakao
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kuo-Ping Huang
- Program of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | | | - Isabella A Graef
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan,Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan, Tel: +81 562 93 9376, Fax: +81 562 92 5382, E-mail:
| |
Collapse
|
114
|
Parlak N, Görgülü Y, Köse Çinar R, Sönmez MB, Parlak E. Serum agouti-related protein (AgRP) levels in bipolar disorder: Could AgRP be a state marker for mania? Psychiatry Res 2018; 260:36-40. [PMID: 29172096 DOI: 10.1016/j.psychres.2017.11.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/21/2017] [Accepted: 11/04/2017] [Indexed: 11/28/2022]
Abstract
Orexigenic and anorexigenic peptides, especially agouti-related protein (AgRP) and leptin, play important roles in the regulation of energy homeostasis in bipolar disorder. AgRP regulates energy metabolism by increasing appetite and decreasing energy expenditure. The resting energy expenditures of patients with manic bipolar disorder are higher than those of controls. Due to the effects of AgRP on energy expenditure and the increased physical activity of manic patients, we hypothesised that serum AgRP levels may be lower in manic patients than in euthymic patients and controls. There was a total of 112 participants, including 47 patients in the manic group, 35 patients in the euthymic group and 30 healthy controls. For this study, serum AgRP, leptin, cholesterol, and cortisol levels were measured and compared between the groups. The serum AgRP, leptin, and cholesterol levels were significantly different between the groups. The serum AgRP levels of manic group were significantly lower than those of euthymic and control groups. The lower serum AgRP levels of manic patients could be indicators of impaired energy homeostasis during manic episodes. Since the serum AgRP levels of manic patients are lower than those of euthymic patients and controls, AgRP could be a state marker for manic episodes.
Collapse
Affiliation(s)
- Naci Parlak
- Department of Psychiatry, Izzet Baysal Mental Health and Disease Training and Research Hospital, Bolu 14030, Turkey.
| | - Yasemin Görgülü
- Department of Psychiatry, Trakya University Faculty of Medicine, Edirne 22030, Turkey.
| | - Rugül Köse Çinar
- Department of Psychiatry, Trakya University Faculty of Medicine, Edirne 22030, Turkey.
| | - Mehmet Bülent Sönmez
- Department of Psychiatry, Trakya University Faculty of Medicine, Edirne 22030, Turkey.
| | - Ebru Parlak
- Department of Psychiatry, Izzet Baysal Mental Health and Disease Training and Research Hospital, Bolu 14030, Turkey.
| |
Collapse
|
115
|
Robicsek O, Ene HM, Karry R, Ytzhaki O, Asor E, McPhie D, Cohen BM, Ben-Yehuda R, Weiner I, Ben-Shachar D. Isolated Mitochondria Transfer Improves Neuronal Differentiation of Schizophrenia-Derived Induced Pluripotent Stem Cells and Rescues Deficits in a Rat Model of the Disorder. Schizophr Bull 2018; 44:432-442. [PMID: 28586483 PMCID: PMC5814822 DOI: 10.1093/schbul/sbx077] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dysfunction of mitochondria, key players in various essential cell processes, has been repeatedly reported in schizophrenia (SZ). Recently, several studies have reported functional recovery and cellular viability following mitochondrial transplantation, mostly in ischemia experimental models. Here, we aimed to demonstrate beneficial effects of isolated active normal mitochondria (IAN-MIT) transfer in vitro and in vivo, using SZ-derived induced pluripotent stem cells (iPSCs) differentiating into glutamatergic neuron, as well as a rodent model of SZ. First, we show that IAN-MIT enter various cell types without manipulation. Next, we show that IAN-MIT transfer into SZ-derived lymphoblasts induces long-lasting improvement in various mitochondrial functions including cellular oxygen consumption and mitochondrial membrane potential (Δ ψ m). We also demonstrate improved differentiation of SZ-derived iPSCs into neurons, by increased expression of neuronal and glutamatergic markers β3-tubulin, synapsin1, and Tbr1 and by an activation of the glutamate-glutamine cycle. In the animal model, we show that intra-prefrontal cortex injection of IAN-MIT in adolescent rats exposed prenatally to a viral mimic prevents mitochondrial Δ ψ m and attentional deficit at adulthood. Our results provide evidence for a direct link between mitochondrial function and SZ-related deficits both in vitro and in vivo and suggest a therapeutic potential for IAN-MIT transfer in diseases with bioenergetic and neurodevelopmental abnormalities such as SZ.
Collapse
Affiliation(s)
- Odile Robicsek
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Hila M Ene
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Rachel Karry
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Ofer Ytzhaki
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Eyal Asor
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Donna McPhie
- Department of Psychiatry, Harvard Medical School, Boston, McLean Hospital, Belmont, MA
| | - Bruce M Cohen
- Department of Psychiatry, Harvard Medical School, Boston, McLean Hospital, Belmont, MA
| | - Rotem Ben-Yehuda
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ina Weiner
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel,To whom correspondence should be addressed; Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus and B. Rappaport Faculty of Medicine, Technion ITT, POB 9649, Haifa 31096, Israel; tel: +972-4-8295224, fax: +972-4-8295220, e-mail:
| |
Collapse
|
116
|
Corrêa-Velloso JC, Gonçalves MC, Naaldijk Y, Oliveira-Giacomelli Á, Pillat MM, Ulrich H. Pathophysiology in the comorbidity of Bipolar Disorder and Alzheimer's Disease: pharmacological and stem cell approaches. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:34-53. [PMID: 28476640 DOI: 10.1016/j.pnpbp.2017.04.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/28/2017] [Indexed: 12/22/2022]
Abstract
Neuropsychiatric disorders involve various pathological mechanisms, resulting in neurodegeneration and brain atrophy. Neurodevelopmental processes have shown to be critical for the progression of those disorders, which are based on genetic and epigenetic mechanisms as well as on extrinsic factors. We review here common mechanisms underlying the comorbidity of Bipolar Disorders and Alzheimer's Disease, such as aberrant neurogenesis and neurotoxicity, reporting current therapeutic approaches. The understanding of these mechanisms precedes stem cell-based strategies as a new therapeutic possibility for treatment and prevention of Bipolar and Alzheimer's Disease progression. Taking into account the difficulty of studying the molecular basis of disease progression directly in patients, we also discuss the importance of stem cells for effective drug screening, modeling and treating psychiatric diseases, once in vitro differentiation of patient-induced pluripotent stem cells provides relevant information about embryonic origins, intracellular pathways and molecular mechanisms.
Collapse
Affiliation(s)
- Juliana C Corrêa-Velloso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Maria Cb Gonçalves
- Departamento de Neurologia e Neurociências, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, São Paulo, SP 04039-032, Brazil
| | - Yahaira Naaldijk
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Ágatha Oliveira-Giacomelli
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Micheli M Pillat
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
117
|
Nakao A, Miyazaki N, Ohira K, Hagihara H, Takagi T, Usuda N, Ishii S, Murata K, Miyakawa T. Immature morphological properties in subcellular-scale structures in the dentate gyrus of Schnurri-2 knockout mice: a model for schizophrenia and intellectual disability. Mol Brain 2017; 10:60. [PMID: 29233179 PMCID: PMC5727961 DOI: 10.1186/s13041-017-0339-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/19/2017] [Indexed: 01/18/2023] Open
Abstract
Accumulating evidence suggests that subcellular-scale structures such as dendritic spine and mitochondria may be involved in the pathogenesis/pathophysiology of schizophrenia and intellectual disability. Previously, we proposed mice lacking Schnurri-2 (Shn2; also called major histocompatibility complex [MHC]-binding protein 2 [MBP-2], or human immunodeficiency virus type I enhancer binding protein 2 [HIVEP2]) as a schizophrenia and intellectual disability model with mild chronic inflammation. In the mutants’ brains, there are increases in C4b and C1q genes, which are considered to mediate synapse elimination during postnatal development. However, morphological properties of subcellular-scale structures such as dendritic spine in Shn2 knockout (KO) mice remain unknown. In this study, we conducted three-dimensional morphological analyses in subcellular-scale structures in dentate gyrus granule cells of Shn2 KO mice by serial block-face scanning electron microscopy. Shn2 KO mice showed immature dendritic spine morphology characterized by increases in spine length and decreases in spine diameter. There was a non-significant tendency toward decrease in spine density of Shn2 KO mice over wild-type mice, and spine volume was indistinguishable between genotypes. Shn2 KO mice exhibited a significant reduction in GluR1 expression and a nominally significant decrease in SV2 expression, while PSD95 expression had a non-significant tendency to decrease in Shn2 KO mice. There were significant decreases in dendrite diameter, nuclear volume, and the number of constricted mitochondria in the mutants. Additionally, neuronal density was elevated in Shn2 KO mice. These results suggest that Shn2 KO mice serve as a unique tool for investigating morphological abnormalities of subcellular-scale structures in schizophrenia, intellectual disability, and its related disorders.
Collapse
Affiliation(s)
- Akito Nakao
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Naoyuki Miyazaki
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Koji Ohira
- Department of Food Science and Nutrition, Mukogawa Women's University, Nishinomiya, Japan
| | - Hideo Hagihara
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Tsuyoshi Takagi
- Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan.,RIKEN Tsukuba Institute, Tsukuba, Japan
| | - Nobuteru Usuda
- Department of Anatomy II, Fujita Health University School of Medicine, Toyoake, Japan
| | | | - Kazuyoshi Murata
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
118
|
Wang Q, Dwivedi Y. Transcriptional profiling of mitochondria associated genes in prefrontal cortex of subjects with major depressive disorder. World J Biol Psychiatry 2017; 18:592-603. [PMID: 27269743 PMCID: PMC5389940 DOI: 10.1080/15622975.2016.1197423] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Recent evidences suggest that mitochondrial dysfunction maybe involved in the pathophysiology of major depressive disorder (MDD); however, the role of mitochondrial genes in this disorder has not been studied systematically. In the present study, we profiled expression of mitochondrial genes in dorsolateral prefrontal cortex (dlPFC) of MDD and non-psychiatric control subjects. METHODS Human mitochondrial RT2 profile PCR array plates were used to examine differentially expressed genes in dlPFC of 11 MDD and 11 control subjects. Differentially expressed genes were validated independently by qRT-PCR. Biological relevance of differentially expressed genes was analysed by gene ontology (GO) and ingenuity pathways analysis (IPA). RESULTS We found that 16 genes were differentially expressed in the MDD group compared with control group. Among them, three genes were downregulated and 13 genes upregulated. None of these genes were affected by confounding variables, such as age, post-mortem interval, brain pH, and antidepressant toxicology. Seven differentially expressed genes were successfully validated in MDD subjects. GO and IPA analyses identified several new regulatory networks associated with mitochondrial dysfunctions in MDD. CONCLUSIONS Our findings suggest abnormal mitochondrial systems in the brain of MDD subjects which could be involved in the etiopathogenesis of this disorder.
Collapse
Affiliation(s)
- Qingzhong Wang
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
119
|
Zheng G, Zheng Q, Xu Q. Identification of key signaling pathways in cerebral small vessel disease using differential pathway network analysis. Exp Ther Med 2017; 14:4371-4376. [PMID: 29104648 DOI: 10.3892/etm.2017.5104] [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: 08/05/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
Cerebral small vessel disease (CSVD) primarily affects the perforating cerebral arterioles and capillaries, and results in injury to subcortical grey and white matter. Despite advances in determining the genetic basis of CSVD, the molecular mechanisms underlying the development and progression of CSVD remain unclear. The present study aimed to identify significant signaling pathways associated with CSVD based on differential pathway network analysis. Combining CSVD microarray data with human protein-protein interaction data and data from the Reactome pathway database, pathway interactions were constructed using the Spearman's correlation coefficient strategy. Pathway interactions with weight values of >0.95 were selected to construct the differential pathway network, which contained 715 differential pathway interactions covering 312 nodes and was visualized using Cytoscape software. A total of 15 hub pathways with a top 5% degree distribution in the differential pathway network were identified. The top 5 hub pathways were associated with the synthesis and metabolism of fatty acids. The results of the present study indicate that the synthesis and metabolism of fatty acids is associated with the occurrence and development of CSVD, and may thus provide insights to improve the early diagnosis and treatment of CSVD.
Collapse
Affiliation(s)
- Gang Zheng
- Department of Neurology, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Qianlei Zheng
- Intensive Care Unit, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Qianwei Xu
- Department of Rehabilitation, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
120
|
Manchia M, Piras IS, Huentelman MJ, Pinna F, Zai CC, Kennedy JL, Carpiniello B. Pattern of gene expression in different stages of schizophrenia: Down-regulation of NPTX2 gene revealed by a meta-analysis of microarray datasets. Eur Neuropsychopharmacol 2017; 27:1054-1063. [PMID: 28732597 DOI: 10.1016/j.euroneuro.2017.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 06/13/2017] [Accepted: 07/05/2017] [Indexed: 12/28/2022]
Abstract
Schizophrenia (SCZ) is a severe psychiatric disorder with a genetic susceptibility. Alterations in neurochemical signaling, as well as changes in brain structure and function, manifest during the course of SCZ and are likely causative of the symptoms shown by affected individuals. However, little is known about the timing of these changes, particularly in the pre-morbid and prodromal phases of SCZ. Here, we performed a gene-based and pathway-based meta-analysis of 5 microarray datasets from human induced pluripotent stem cells (hiPSCs)-derived neurons and post-mortem brain tissue from SCZ and healthy controls (HC), with the underlying assumption they might represent the neurobiological make-up of SCZ in the pre-morbid and chronic stages of illness, respectively. Thus, we identified 1 microarray expression profiling dataset of hiPSCs-derived neurons (GSE25673) and performed a systematic search of microarray expression profiling datasets from SCZ post-mortem brain publicly available on the Gene Expression Omnibus (GEO) repository. We selected 4 different SCZ post-mortem brain microarray expression profiling datasets (GSE17612, GSE21935, GSE12649, and GSE21338) according to specific inclusion and exclusion criteria. We downloaded raw data and performed quality controls, differential expression analysis, and gene-based, as well as pathway-based meta-analysis. Neuronal pentraxin 2 (NPTX2) gene was consistently down-regulated across all datasets, with highly significant association in the meta-analysis (FDR<1.0E-04). These results highlight the heuristic value of microarray meta-analysis and suggest a role of NPTX2 as a disease biomarker, provided that it achieves biological validation in future studies examining whether this down-regulation has predictive value with respect to the developmental trajectory of SCZ.
Collapse
Affiliation(s)
- Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Ignazio S Piras
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Matthew J Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Federica Pinna
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Clement C Zai
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - James L Kennedy
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Bernardo Carpiniello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
121
|
Zuccoli GS, Saia-Cereda VM, Nascimento JM, Martins-de-Souza D. The Energy Metabolism Dysfunction in Psychiatric Disorders Postmortem Brains: Focus on Proteomic Evidence. Front Neurosci 2017; 11:493. [PMID: 28936160 PMCID: PMC5594406 DOI: 10.3389/fnins.2017.00493] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/22/2017] [Indexed: 12/27/2022] Open
Abstract
Psychiatric disorders represent a great medical and social challenge and people suffering from these conditions face many impairments regarding personal and professional life. In addition, a mental disorder will manifest itself in approximately one quarter of the world's population at some period of their life. Dysfunction in energy metabolism is one of the most consistent scientific findings associated with these disorders. With this is mind, this review compiled data on disturbances in energy metabolism found by proteomic analyses of postmortem brains collected from patients affected by the most prevalent psychiatric disorders: schizophrenia (SCZ), bipolar disorder (BPD), and major depressive disorder (MDD). We searched in the PubMed database to gather the studies and compiled all the differentially expressed proteins reported in each work. SCZ studies revealed 92 differentially expressed proteins related to energy metabolism, while 95 proteins were discovered in BPD, and 41 proteins in MDD. With the compiled data, it was possible to determine which proteins related to energy metabolism were found to be altered in all the disorders as well as which ones were altered exclusively in one of them. In conclusion, the information gathered in this work could contribute to a better understanding of the impaired metabolic mechanisms and hopefully bring insights into the underlying neuropathology of psychiatric disorders.
Collapse
Affiliation(s)
- Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of CampinasCampinas, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e TecnologicoSão Paulo, Brazil
| | - Verônica M Saia-Cereda
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of CampinasCampinas, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e TecnologicoSão Paulo, Brazil
| | - Juliana M Nascimento
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of CampinasCampinas, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e TecnologicoSão Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of CampinasCampinas, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e TecnologicoSão Paulo, Brazil
| |
Collapse
|
122
|
An emerging role for mitochondrial dynamics in schizophrenia. Schizophr Res 2017; 187:26-32. [PMID: 28526279 PMCID: PMC5646380 DOI: 10.1016/j.schres.2017.05.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 12/27/2022]
Abstract
Abnormal brain development has long been thought to contribute to the pathophysiology of schizophrenia. Impaired dendritic arborization, synaptogenesis, and long term potentiation and memory have been demonstrated in animal models of schizophrenia. In addition to aberrant nervous system development, altered brain metabolism and mitochondrial function has long been observed in schizophrenic patients. Single nucleotide polymorphisms in the mitochondrial genome as well as impaired mitochondrial function have both been associated with increased risk for developing schizophrenia. Mitochondrial function in neurons is highly dependent on fission, fusion, and transport of the organelle, collectively referred to as mitochondrial dynamics. Indeed, there is mounting evidence that mitochondrial dynamics strongly influences neuron development and synaptic transmission. While there are a few studies describing altered mitochondrial shape in schizophrenic patients, as well as in animal and in vitro models of schizophrenia, the precise role of mitochondrial dynamics in the pathophysiology of schizophrenia is all but unexplored. Here we discuss the influence of mitochondrial dynamics and mitochondrial function on nervous system development, and highlight recent work suggesting a link between aberrant mitochondrial dynamics and schizophrenia.
Collapse
|
123
|
Kato T. Neurobiological basis of bipolar disorder: Mitochondrial dysfunction hypothesis and beyond. Schizophr Res 2017; 187:62-66. [PMID: 27839913 DOI: 10.1016/j.schres.2016.10.037] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/24/2016] [Accepted: 10/27/2016] [Indexed: 02/07/2023]
Abstract
Bipolar disorder is one of two major psychotic disorders together with schizophrenia and causes severe psychosocial disturbance. Lack of adequate animal models hampers development of new mood stabilizers. We proposed a mitochondrial dysfunction hypothesis and have been studying the neurobiology of bipolar disorder based on this hypothesis. We showed that deletions of mitochondrial DNA (ΔmtDNA) play a pathophysiological role at least in some patients with bipolar disorder possibly by affecting intracellular calcium regulation. Mutant polymerase γ transgenic mice that accumulate ΔmtDNA in the brain showed recurrent spontaneous depression-like episodes which were prevented by a serotonin-selective reuptake inhibitor and worsened by lithium withdrawal. The animal model would be useful to develop new mood stabilizers.
Collapse
Affiliation(s)
- Tadafumi Kato
- Laboratory for Molecular Dynamic of Mental Disorders, RIKEN Brain Science Institute, Japan.
| |
Collapse
|
124
|
Niculescu AB, Le-Niculescu H, Levey DF, Phalen PL, Dainton HL, Roseberry K, Niculescu EM, Niezer JO, Williams A, Graham DL, Jones TJ, Venugopal V, Ballew A, Yard M, Gelbart T, Kurian SM, Shekhar A, Schork NJ, Sandusky GE, Salomon DR. Precision medicine for suicidality: from universality to subtypes and personalization. Mol Psychiatry 2017; 22:1250-1273. [PMID: 28809398 PMCID: PMC5582166 DOI: 10.1038/mp.2017.128] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 01/15/2023]
Abstract
Suicide remains a clear, present and increasing public health problem, despite being a potentially preventable tragedy. Its incidence is particularly high in people with overt or un(der)diagnosed psychiatric disorders. Objective and precise identification of individuals at risk, ways of monitoring response to treatments and novel preventive therapeutics need to be discovered, employed and widely deployed. We sought to investigate whether blood gene expression biomarkers for suicide (that is, a 'liquid biopsy' approach) can be identified that are more universal in nature, working across psychiatric diagnoses and genders, using larger cohorts than in previous studies. Such markers may reflect and/or be a proxy for the core biology of suicide. We were successful in this endeavor, using a comprehensive stepwise approach, leading to a wealth of findings. Steps 1, 2 and 3 were discovery, prioritization and validation for tracking suicidality, resulting in a Top Dozen list of candidate biomarkers comprising the top biomarkers from each step, as well as a larger list of 148 candidate biomarkers that survived Bonferroni correction in the validation step. Step 4 was testing the Top Dozen list and Bonferroni biomarker list for predictive ability for suicidal ideation (SI) and for future hospitalizations for suicidality in independent cohorts, leading to the identification of completely novel predictive biomarkers (such as CLN5 and AK2), as well as reinforcement of ours and others previous findings in the field (such as SLC4A4 and SKA2). Additionally, we examined whether subtypes of suicidality can be identified based on mental state at the time of high SI and identified four potential subtypes: high anxiety, low mood, combined and non-affective (psychotic). Such subtypes may delineate groups of individuals that are more homogenous in terms of suicidality biology and behavior. We also studied a more personalized approach, by psychiatric diagnosis and gender, with a focus on bipolar males, the highest risk group. Such a personalized approach may be more sensitive to gender differences and to the impact of psychiatric co-morbidities and medications. We compared testing the universal biomarkers in everybody versus testing by subtypes versus personalized by gender and diagnosis, and show that the subtype and personalized approaches permit enhanced precision of predictions for different universal biomarkers. In particular, LHFP appears to be a strong predictor for suicidality in males with depression. We also directly examined whether biomarkers discovered using male bipolars only are better predictors in a male bipolar independent cohort than universal biomarkers and show evidence for a possible advantage of personalization. We identified completely novel biomarkers (such as SPTBN1 and C7orf73), and reinforced previously known biomarkers (such as PTEN and SAT1). For diagnostic ability testing purposes, we also examined as predictors phenotypic measures as apps (for suicide risk (CFI-S, Convergent Functional Information for Suicidality) and for anxiety and mood (SASS, Simplified Affective State Scale)) by themselves, as well as in combination with the top biomarkers (the combination being our a priori primary endpoint), to provide context and enhance precision of predictions. We obtained area under the curves of 90% for SI and 77% for future hospitalizations in independent cohorts. Step 5 was to look for mechanistic understanding, starting with examining evidence for the Top Dozen and Bonferroni biomarkers for involvement in other psychiatric and non-psychiatric disorders, as a mechanism for biological predisposition and vulnerability. The biomarkers we identified also provide a window towards understanding the biology of suicide, implicating biological pathways related to neurogenesis, programmed cell death and insulin signaling from the universal biomarkers, as well as mTOR signaling from the male bipolar biomarkers. In particular, HTR2A increase coupled with ARRB1 and GSK3B decreases in expression in suicidality may provide a synergistic mechanistical corrective target, as do SLC4A4 increase coupled with AHCYL1 and AHCYL2 decrease. Step 6 was to move beyond diagnostics and mechanistical risk assessment, towards providing a foundation for personalized therapeutics. Items scored positive in the CFI-S and subtypes identified by SASS in different individuals provide targets for personalized (psycho)therapy. Some individual biomarkers are targets of existing drugs used to treat mood disorders and suicidality (lithium, clozapine and omega-3 fatty acids), providing a means toward pharmacogenomics stratification of patients and monitoring of response to treatment. Such biomarkers merit evaluation in clinical trials. Bioinformatics drug repurposing analyses with the gene expression biosignatures of the Top Dozen and Bonferroni-validated universal biomarkers identified novel potential therapeutics for suicidality, such as ebselen (a lithium mimetic), piracetam (a nootropic), chlorogenic acid (a polyphenol) and metformin (an antidiabetic and possible longevity promoting drug). Finally, based on the totality of our data and of the evidence in the field to date, a convergent functional evidence score prioritizing biomarkers that have all around evidence (track suicidality, predict it, are reflective of biological predisposition and are potential drug targets) brought to the fore APOE and IL6 from among the universal biomarkers, suggesting an inflammatory/accelerated aging component that may be a targetable common denominator.
Collapse
Affiliation(s)
- A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA,Indianapolis VA Medical Center, Indianapolis, IN, USA,INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Psychiatry, Indiana University School of Medicine, Neuroscience Research Building 200B, 320 West 15th Street, Indianapolis, IN 46202, USA. E-mail:
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D F Levey
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - P L Phalen
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - H L Dainton
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Roseberry
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E M Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J O Niezer
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Williams
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - D L Graham
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - T J Jones
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - V Venugopal
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Ballew
- Marion County Coroner’s Office, Indianapolis, IN, USA
| | - M Yard
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA
| | - T Gelbart
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - S M Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N J Schork
- J. Craig Venter Institute, La Jolla, CA, USA
| | - G E Sandusky
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D R Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
125
|
Lee BJ, Marchionni L, Andrews CE, Norris AL, Nucifora LG, Wu YC, Wright RA, Pevsner J, Ross CA, Margolis RL, Sawa A, Nucifora FC. Analysis of differential gene expression mediated by clozapine in human postmortem brains. Schizophr Res 2017; 185:58-66. [PMID: 28038920 PMCID: PMC6541388 DOI: 10.1016/j.schres.2016.12.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 12/11/2022]
Abstract
Clozapine is the only medication indicated for treating refractory schizophrenia, due to its superior efficacy among all antipsychotic agents, but its mechanism of action is poorly understood. To date, no studies of human postmortem brain have characterized the gene expression response to clozapine. Therefore, we addressed this question by analyzing expression data extracted from published microarray studies involving brains of patients on antipsychotic therapy. We first performed a systematic review and identified four microarray studies of postmortem brains from antipsychotic-treated patients, then extracted the expression data. We then performed generalized linear model analysis on each study separately, and identified the genes differentially expressed in response to clozapine compared to other atypical antipsychotic medications, as well as their associated canonical pathways. We also found a number of genes common to all four studies that we analyzed: GCLM, ZNF652, and GYPC. In addition, pathway analysis highlighted the following processes in all four studies: clathrin-mediated endocytosis, SAPK/JNK signaling, 3-phosphoinositide synthesis, and paxillin signaling. Our analysis yielded the first comprehensive compendium of genes and pathways differentially expressed upon clozapine treatment in the human brain, which may provide insight into the mechanism and unique efficacy of clozapine, as well as the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Brian J Lee
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, 1830 E. Monument St., Baltimore, MD 21205, USA
| | - Luigi Marchionni
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, MD 21287, USA
| | - Carrie E Andrews
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA
| | - Alexis L Norris
- Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA; Department of Neurology, Kennedy Krieger Institute, Johns Hopkins University School of Medicine, 707 N. Broadway, Baltimore, MD 21205, USA
| | - Leslie G Nucifora
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA
| | - Yeewen Candace Wu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA
| | - Robert A Wright
- William H. Welch Medical Library, Johns Hopkins University, 1900 E. Monument St., Baltimore, MD 21205, USA
| | - Jonathan Pevsner
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA; Department of Neurology, Kennedy Krieger Institute, Johns Hopkins University School of Medicine, 707 N. Broadway, Baltimore, MD 21205, USA
| | - Christopher A Ross
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, 1830 E. Monument St., Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA
| | - Russell L Margolis
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, 1830 E. Monument St., Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, 1830 E. Monument St., Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA
| | - Frederick C Nucifora
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA.
| |
Collapse
|
126
|
Abstract
Schizophrenia is a devastating illness that affects up to 1% of the population; it is characterized by a combination of positive symptoms, negative symptoms, and cognitive impairment. Currently, treatment consists of one class of medications known as antipsychotics, which include typical (first-generation) and atypical (second-generation) agents. Unfortunately, antipsychotic medications have limited efficacy, with up to a third of patients lacking a full response. Clozapine, the first atypical antipsychotic developed, is the only medication shown to be superior to all other antipsychotics. However, owing to several life-threatening side effects and required enrollment in a registry with routine blood monitoring, clozapine is greatly underutilized in the US. Developing a medication as efficacious as clozapine with limited side effects would likely become the first-line therapy for schizophrenia and related disorders. In this review, we discuss the history of clozapine, landmark studies, and its clinical advantages and disadvantages. We further discuss the hypotheses for clozapine's superior efficacy based on neuroreceptor binding, and the limitations of a receptor-based approach to antipsychotic development. We highlight some of the advances from pharmacogenetic studies on clozapine and then focus on studies of clozapine using unbiased approaches such as pharmacogenomics and gene expression profiling. Finally, we examine how these approaches could provide insights into clozapine's mechanism of action and side-effect profile, and lead to novel and improved therapeutics.
Collapse
Affiliation(s)
- Frederick C Nucifora
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | - Brian J Lee
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
127
|
Abstract
Schizophrenia (SZ) is a debilitating brain disorder with a complex genetic architecture. Genetic studies, especially recent genome-wide association studies (GWAS), have identified multiple variants (loci) conferring risk to SZ. However, how to efficiently extract meaningful biological information from bulk genetic findings of SZ remains a major challenge. There is a pressing need to integrate multiple layers of data from various sources, eg, genetic findings from GWAS, copy number variations (CNVs), association and linkage studies, gene expression, protein-protein interaction (PPI), co-expression, expression quantitative trait loci (eQTL), and Encyclopedia of DNA Elements (ENCODE) data, to provide a comprehensive resource to facilitate the translation of genetic findings into SZ molecular diagnosis and mechanism study. Here we developed the SZDB database (http://www.szdb.org/), a comprehensive resource for SZ research. SZ genetic data, gene expression data, network-based data, brain eQTL data, and SNP function annotation information were systematically extracted, curated and deposited in SZDB. In-depth analyses and systematic integration were performed to identify top prioritized SZ genes and enriched pathways. Multiple types of data from various layers of SZ research were systematically integrated and deposited in SZDB. In-depth data analyses and integration identified top prioritized SZ genes and enriched pathways. We further showed that genes implicated in SZ are highly co-expressed in human brain and proteins encoded by the prioritized SZ risk genes are significantly interacted. The user-friendly SZDB provides high-confidence candidate variants and genes for further functional characterization. More important, SZDB provides convenient online tools for data search and browse, data integration, and customized data analyses.
Collapse
Affiliation(s)
- Yong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China;,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China;,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China;,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China,YGY and XJL are co-corresponding authors who jointly directed this work
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China;,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China;,YGY and XJL are co-corresponding authors who jointly directed this work
| |
Collapse
|
128
|
Morris G, Walder K, McGee SL, Dean OM, Tye SJ, Maes M, Berk M. A model of the mitochondrial basis of bipolar disorder. Neurosci Biobehav Rev 2017; 74:1-20. [DOI: 10.1016/j.neubiorev.2017.01.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
|
129
|
Kim Y, Santos R, Gage FH, Marchetto MC. Molecular Mechanisms of Bipolar Disorder: Progress Made and Future Challenges. Front Cell Neurosci 2017; 11:30. [PMID: 28261061 PMCID: PMC5306135 DOI: 10.3389/fncel.2017.00030] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/01/2017] [Indexed: 12/15/2022] Open
Abstract
Bipolar disorder (BD) is a chronic and progressive psychiatric illness characterized by mood oscillations, with episodes of mania and depression. The impact of BD on patients can be devastating, with up to 15% of patients committing suicide. This disorder is associated with psychiatric and medical comorbidities and patients with a high risk of drug abuse, metabolic and endocrine disorders and vascular disease. Current knowledge of the pathophysiology and molecular mechanisms causing BD is still modest. With no clear biological markers available, early diagnosis is a great challenge to clinicians without previous knowledge of the longitudinal progress of illness. Moreover, despite recommendations from evidence-based guidelines, polypharmacy is still common in clinical treatment of BD, reflecting the gap between research and clinical practice. A major challenge in BD is the development of effective drugs with low toxicity for the patients. In this review article, we focus on the progress made and future challenges we face in determining the pathophysiology and molecular pathways involved in BD, such as circadian and metabolic perturbations, mitochondrial and endoplasmic reticulum (ER) dysfunction, autophagy and glutamatergic neurotransmission; which may lead to the development of new drugs.
Collapse
Affiliation(s)
- Yeni Kim
- Laboratory of Genetics, The Salk Institute for Biological StudiesLa Jolla, CA, USA; Department of Child and Adolescent Psychiatry, National Center for Mental HealthSeoul, South Korea
| | - Renata Santos
- Laboratory of Genetics, The Salk Institute for Biological StudiesLa Jolla, CA, USA; Ecole Normale Supérieure, PSL Research University, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Biologie de l'Ecole Normale Supérieure (IBENS)Paris, France
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies La Jolla, CA, USA
| | - Maria C Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies La Jolla, CA, USA
| |
Collapse
|
130
|
Bansal Y, Kuhad A. Mitochondrial Dysfunction in Depression. Curr Neuropharmacol 2017; 14:610-8. [PMID: 26923778 PMCID: PMC4981740 DOI: 10.2174/1570159x14666160229114755] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 06/02/2015] [Accepted: 02/27/2016] [Indexed: 02/06/2023] Open
Abstract
Abstract: Background Depression is the most debilitating neuropsychiatric disorder with significant impact on socio-occupational and well being of individual. The exact pathophysiology of depression is still enigmatic though various theories have been put forwarded. There are evidences showing that mitochondrial dysfunction in various brain regions is associated with depression. Recent findings have sparked renewed appreciation for the role of mitochondria in many intracellular processes coupled to synaptic plasticity and cellular resilience. New insights in depression pathophysiology are revolving around the impairment of neuroplasticity. Mitochondria have potential role in ATP production, intracellular Ca2+ signalling to establish membrane stability, reactive oxygen species (ROS) balance and to execute the complex processes of neurotransmission and plasticity. So understanding the various concepts of mitochondrial dysfunction in pathogenesis of depression indubitably helps to generate novel and more targeted therapeutic approaches for depression treatment. Objective The review was aimed to give a comprehensive insight on role of mitochondrial dysfunction in depression. Result Targeting mitochondrial dysfunction and enhancing the mitochondrial functions might act as potential target for the treatment of depression. Conclusion Literature cited in this review highly supports the role of mitochondrial dysfunction in depression. As impairment in the mitochondrial functions lead to the generation of various insults that exaggerate the pathogenesis of depression. So, it is useful to study mitochondrial dysfunction in relation to mood disorders, synaptic plasticity, neurogenesis and enhancing the functions of mitochondria might show promiscuous effects in the treatment of depressed patients.
Collapse
Affiliation(s)
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences UGC-Centre of Advanced Study, Panjab University, Chandigarh - 160 014 India.
| |
Collapse
|
131
|
Selfish brain and selfish immune system interplay: A theoretical framework for metabolic comorbidities of mood disorders. Neurosci Biobehav Rev 2017; 72:43-49. [DOI: 10.1016/j.neubiorev.2016.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/06/2016] [Accepted: 11/16/2016] [Indexed: 12/17/2022]
|
132
|
Mladinov M, Sedmak G, Fuller HR, Babić Leko M, Mayer D, Kirincich J, Štajduhar A, Borovečki F, Hof PR, Šimić G. Gene expression profiling of the dorsolateral and medial orbitofrontal cortex in schizophrenia. Transl Neurosci 2016; 7:139-150. [PMID: 28123834 PMCID: PMC5234522 DOI: 10.1515/tnsci-2016-0021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/05/2016] [Indexed: 12/29/2022] Open
Abstract
Schizophrenia is a complex polygenic disorder of unknown etiology. Over 3,000 candidate genes associated with schizophrenia have been reported, most of which being mentioned only once. Alterations in cognitive processing - working memory, metacognition and mentalization - represent a core feature of schizophrenia, which indicates the involvement of the prefrontal cortex in the pathophysiology of this disorder. Hence we compared the gene expression in postmortem tissue from the left and right dorsolateral prefrontal cortex (DLPFC, Brodmann's area 46), and the medial part of the orbitofrontal cortex (MOFC, Brodmann's area 11/12), in six patients with schizophrenia and six control brains. Although in the past decade several studies performed transcriptome profiling in schizophrenia, this is the first study to investigate both hemispheres, providing new knowledge about possible brain asymmetry at the level of gene expression and its relation to schizophrenia. We found that in the left hemisphere, twelve genes from the DLPFC and eight genes from the MOFC were differentially expressed in patients with schizophrenia compared to controls. In the right hemisphere there was only one gene differentially expressed in the MOFC. We reproduce the involvement of previously reported genes TARDBP and HNRNPC in the pathogenesis of schizophrenia, and report seven novel genes: SART1, KAT7, C1D, NPM1, EVI2A, XGY2, and TTTY15. As the differentially expressed genes only partially overlap with previous studies that analyzed other brain regions, our findings indicate the importance of considering prefrontal cortical regions, especially those in the left hemisphere, for obtaining disease-relevant insights.
Collapse
Affiliation(s)
- Mihovil Mladinov
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia; Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Goran Sedmak
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Heidi R Fuller
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, SY10 7AG, UK and Institute for Science and Technology in Medicine, Keele University, Staffordshire, ST5 5BG, United Kingdom of Great Britain and Northern Ireland
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Davor Mayer
- Department of Forensic Medicine, University of Zagreb Medical School, Zagreb, Croatia
| | - Jason Kirincich
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Andrija Štajduhar
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Fran Borovečki
- Department of Neurology, University Clinical Hospital Zagreb, Zagreb, Croatia
| | - Patrick R Hof
- Fishberg Department of Neuroscience, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
133
|
Föcking M, Dicker P, Lopez LM, Hryniewiecka M, Wynne K, English JA, Cagney G, Cotter DR. Proteomic analysis of the postsynaptic density implicates synaptic function and energy pathways in bipolar disorder. Transl Psychiatry 2016; 6:e959. [PMID: 27898073 PMCID: PMC5290351 DOI: 10.1038/tp.2016.224] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/26/2016] [Accepted: 09/28/2016] [Indexed: 12/17/2022] Open
Abstract
The postsynaptic density (PSD) contains a complex set of proteins of known relevance to neuropsychiatric disorders such as schizophrenia and bipolar disorder. We enriched for this anatomical structure in the anterior cingulate cortex of 16 bipolar disorder samples and 20 controls from the Stanley Medical Research Institute. Unbiased shotgun proteomics incorporating label-free quantitation was used to identify differentially expressed proteins. Quantitative investigation of the PSD identified 2033 proteins, among which 288 were found to be differentially expressed. Validation of expression changes of DNM1, DTNA, NDUFV2, SEPT11 and SSBP was performed by western blotting. Bioinformatics analysis of the differentially expressed proteins implicated metabolic pathways including mitochondrial function, the tricarboxylic acid cycle, oxidative phosphorylation, protein translation and calcium signaling. The data implicate PSD-associated proteins, and specifically mitochondrial function in bipolar disorder. They relate synaptic function in bipolar disorder and the energy pathways that underpin it. Overall, our findings add to a growing literature linking the PSD and mitochondrial function in psychiatric disorders generally, and suggest that mitochondrial function associated with the PSD is particularly important in bipolar disorder.
Collapse
Affiliation(s)
- M Föcking
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland,Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Education and Research Centre, Dublin 9, Ireland. E-mail: or
| | - P Dicker
- Departments of Epidemiology and Public Health, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - L M Lopez
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - M Hryniewiecka
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - K Wynne
- Proteome Research Centre, UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - J A English
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - G Cagney
- Proteome Research Centre, UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - D R Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland,Department of Psychiatry, Beaumont Hospital, Dublin, Ireland,Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Education and Research Centre, Dublin 9, Ireland. E-mail: or
| |
Collapse
|
134
|
Wang J, Qu S, Wang W, Guo L, Zhang K, Chang S, Wang J. A combined analysis of genome-wide expression profiling of bipolar disorder in human prefrontal cortex. J Psychiatr Res 2016; 82:23-9. [PMID: 27459029 DOI: 10.1016/j.jpsychires.2016.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/12/2016] [Accepted: 07/15/2016] [Indexed: 01/29/2023]
Abstract
Numbers of gene expression profiling studies of bipolar disorder have been published. Besides different array chips and tissues, variety of the data processes in different cohorts aggravated the inconsistency of results of these genome-wide gene expression profiling studies. By searching the gene expression databases, we obtained six data sets for prefrontal cortex (PFC) of bipolar disorder with raw data and combinable platforms. We used standardized pre-processing and quality control procedures to analyze each data set separately and then combined them into a large gene expression matrix with 101 bipolar disorder subjects and 106 controls. A standard linear mixed-effects model was used to calculate the differentially expressed genes (DEGs). Multiple levels of sensitivity analyses and cross validation with genetic data were conducted. Functional and network analyses were carried out on basis of the DEGs. In the result, we identified 198 unique differentially expressed genes in the PFC of bipolar disorder and control. Among them, 115 DEGs were robust to at least three leave-one-out tests or different pre-processing methods; 51 DEGs were validated with genetic association signals. Pathway enrichment analysis showed these DEGs were related with regulation of neurological system, cell death and apoptosis, and several basic binding processes. Protein-protein interaction network further identified one key hub gene. We have contributed the most comprehensive integrated analysis of bipolar disorder expression profiling studies in PFC to date. The DEGs, especially those with multiple validations, may denote a common signature of bipolar disorder and contribute to the pathogenesis of disease.
Collapse
Affiliation(s)
- Jinglu Wang
- The Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Susu Qu
- The Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Weixiao Wang
- The Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Liyuan Guo
- The Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Kunlin Zhang
- The Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Suhua Chang
- The Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.
| | - Jing Wang
- The Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
135
|
Yoshimi N, Futamura T, Bergen SE, Iwayama Y, Ishima T, Sellgren C, Ekman CJ, Jakobsson J, Pålsson E, Kakumoto K, Ohgi Y, Yoshikawa T, Landén M, Hashimoto K. Cerebrospinal fluid metabolomics identifies a key role of isocitrate dehydrogenase in bipolar disorder: evidence in support of mitochondrial dysfunction hypothesis. Mol Psychiatry 2016; 21:1504-1510. [PMID: 26782057 PMCID: PMC5078854 DOI: 10.1038/mp.2015.217] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/10/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022]
Abstract
Although evidence for mitochondrial dysfunction in the pathogenesis of bipolar disorder (BD) has been reported, the precise biological basis remains unknown, hampering the search for novel biomarkers. In this study, we performed metabolomics of cerebrospinal fluid (CSF) from male BD patients (n=54) and age-matched male healthy controls (n=40). Subsequently, post-mortem brain analyses, genetic analyses, metabolomics of CSF samples from rats treated with lithium or valproic acid were also performed. After multivariate logistic regression, isocitric acid (isocitrate) levels were significantly higher in the CSF from BD patients than healthy controls. Furthermore, gene expression of two subtypes (IDH3A and IDH3B) of isocitrate dehydrogenase (IDH) in the dorsolateral prefrontal cortex from BD patients was significantly lower than that of controls, although the expression of other genes including, aconitase (ACO1, ACO2), IDH1, IDH2 and IDH3G, were not altered. Moreover, protein expression of IDH3A in the cerebellum from BD patients was higher than that of controls. Genetic analyses showed that IDH genes (IDH1, IDH2, IDH3A, IDH3B) and ACO genes (ACO1, ACO2) were not associated with BD. Chronic (4 weeks) treatment with lithium or valproic acid in rats did not alter CSF levels of isocitrate, and mRNA levels of Idh3a, Idh3b, Aco1 and Aco2 genes in the rat brain. These findings suggest that abnormality in the metabolism of isocitrate by IDH3A in the mitochondria plays a key role in the pathogenesis of BD, supporting the mitochondrial dysfunction hypothesis of BD. Therefore, IDH3 in the citric acid cycle could potentially be a novel therapeutic target for BD.
Collapse
Affiliation(s)
- N Yoshimi
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan,Qs' Research Institute, Otsuka Pharmaceutical, Tokushima, Japan
| | - T Futamura
- Qs' Research Institute, Otsuka Pharmaceutical, Tokushima, Japan
| | - S E Bergen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Y Iwayama
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Wako, Japan
| | - T Ishima
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - C Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - C J Ekman
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - J Jakobsson
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - E Pålsson
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - K Kakumoto
- Tokushima Research Institute, Otsuka Pharmaceutical, Tokushima, Japan
| | - Y Ohgi
- Qs' Research Institute, Otsuka Pharmaceutical, Tokushima, Japan
| | - T Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Wako, Japan
| | - M Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden,Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - K Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan,Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba 260-8670, Japan. E-mail:
| |
Collapse
|
136
|
Hagihara H, Shoji H, Miyakawa T. Immaturity of brain as an endophenotype of neuropsychiatric disorders. Nihon Yakurigaku Zasshi 2016; 148:168-175. [PMID: 27725563 DOI: 10.1254/fpj.148.168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
137
|
Brandt R. Mitochondrial donation and 'the right to know'. JOURNAL OF MEDICAL ETHICS 2016; 42:678-84. [PMID: 27542387 PMCID: PMC5099313 DOI: 10.1136/medethics-2016-103587] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/23/2016] [Indexed: 05/06/2023]
Abstract
In this paper, I examine two key arguments advanced by the Human Fertilization and Embryology Authority (HFEA) and the Nuffield Council justifying anonymous mitochondrial donation, even though the 'right to know' is recognised in standard gamete donation. I argue that the two arguments they offer, what I call the argument from genetic connection and the argument from personal characteristics, are unsuccessful. However, I provide additional reasons for why recognising the right to know in gamete donation but not in mitochondrial donation may be justified. I further argue that the status quo in the UK, which is to not recognise a right to know in mitochondrial donation, is provisionally acceptable.
Collapse
|
138
|
Nagalski A, Kozinski K, Wisniewska MB. Metabolic pathways in the periphery and brain: Contribution to mental disorders? Int J Biochem Cell Biol 2016; 80:19-30. [PMID: 27644152 DOI: 10.1016/j.biocel.2016.09.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 12/20/2022]
Abstract
The association between mental disorders and diabetes has a long history. Recent large-scale, well-controlled epidemiological studies confirmed a link between diabetes and psychiatric illnesses. The scope of this review is to summarize our current understanding of this relationship from a molecular perspective. We first discuss the potential contribution of diabetes-associated metabolic impairments to the etiology of mental conditions. Then, we focus on possible shared molecular risk factors and mechanisms. Simple comorbidity, shared susceptibility loci, and common pathophysiological processes in diabetes and mental illnesses have changed our traditional way of thinking about mental illness. We conclude that schizophrenia and affective disorders are not limited to an imbalance in dopaminergic and serotoninergic neurotransmission in the brain. They are also systemic disorders that can be considered, to some extent, as metabolic disorders.
Collapse
Affiliation(s)
- Andrzej Nagalski
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Kamil Kozinski
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Marta B Wisniewska
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland.
| |
Collapse
|
139
|
Abstract
Mitochondrial diseases are a clinically heterogeneous group of disorders that ultimately result from dysfunction of the mitochondrial respiratory chain. There is some evidence to suggest that mitochondrial dysfunction plays a role in neuropsychiatric illness; however, the data are inconclusive. This article summarizes the available literature published in the area of neuropsychiatric manifestations in both children and adults with primary mitochondrial disease, with a focus on autism spectrum disorder in children and mood disorders and schizophrenia in adults.
Collapse
Affiliation(s)
- Samantha E Marin
- Department of Neurosciences, University of California, San Diego (UCSD), 9500 Gilman Drive #0935, La Jolla, CA 92093-0935, USA
| | - Russell P Saneto
- Department of Neurology, Seattle Children's Hospital, University of Washington, 4800 Sand Point Way Northeast, Seattle, WA 98105, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, 4800 Sand Point Way Northeast, Seattle, WA 98105, USA.
| |
Collapse
|
140
|
Adzic M, Brkic Z, Bulajic S, Mitic M, Radojcic MB. Antidepressant Action on Mitochondrial Dysfunction in Psychiatric Disorders. Drug Dev Res 2016; 77:400-406. [DOI: 10.1002/ddr.21332] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Miroslav Adzic
- Laboratory of Molecular Biology and Endocrinology; VINCA Institute of Nuclear Sciences, University of Belgrade; Serbia
| | - Zeljka Brkic
- Laboratory of Molecular Biology and Endocrinology; VINCA Institute of Nuclear Sciences, University of Belgrade; Serbia
| | - Sonja Bulajic
- School of Medicine; University of Pristina; Kosovska Mitrovica Serbia
| | - Milos Mitic
- Laboratory of Molecular Biology and Endocrinology; VINCA Institute of Nuclear Sciences, University of Belgrade; Serbia
| | - Marija B. Radojcic
- Laboratory of Molecular Biology and Endocrinology; VINCA Institute of Nuclear Sciences, University of Belgrade; Serbia
| |
Collapse
|
141
|
Bergman O, Ben-Shachar D. Mitochondrial Oxidative Phosphorylation System (OXPHOS) Deficits in Schizophrenia: Possible Interactions with Cellular Processes. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2016; 61:457-69. [PMID: 27412728 PMCID: PMC4959648 DOI: 10.1177/0706743716648290] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondria are key players in the generation and regulation of cellular bioenergetics, producing the majority of adenosine triphosphate molecules by the oxidative phosphorylation system (OXPHOS). Linked to numerous signaling pathways and cellular functions, mitochondria, and OXPHOS in particular, are involved in neuronal development, connectivity, plasticity, and differentiation. Impairments in a variety of mitochondrial functions have been described in different general and psychiatric disorders, including schizophrenia (SCZ), a severe, chronic, debilitating illness that heavily affects the lives of patients and their families. This article reviews findings emphasizing the role of OXPHOS in the pathophysiology of SCZ. Evidence accumulated during the past few decades from imaging, transcriptomic, proteomic, and metabolomic studies points at OXPHOS deficit involvement in SCZ. Abnormalities have been reported in high-energy phosphates generated by the OXPHOS, in the activity of its complexes and gene expression, primarily of complex I (CoI). In addition, cellular signaling such as cAMP/protein kinase A (PKA) and Ca(+2), neuronal development, connectivity, and plasticity have been linked to OXPHOS function and are reported to be impaired in SCZ. Finally, CoI has been shown as a site of interaction for both dopamine (DA) and antipsychotic drugs, further substantiating its role in the pathology of SCZ. Understanding the role of mitochondria and the OXPHOS in particular may encourage new insights into the pathophysiology and etiology of this debilitating disorder.
Collapse
Affiliation(s)
- Oded Bergman
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Medical Center, Technion-IIT, Haifa, Israel B. Rappaport Faculty of Medicine, Technion-IIT, Haifa, Israel
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Medical Center, Technion-IIT, Haifa, Israel B. Rappaport Faculty of Medicine, Technion-IIT, Haifa, Israel
| |
Collapse
|
142
|
Scaini G, Rezin GT, Carvalho AF, Streck EL, Berk M, Quevedo J. Mitochondrial dysfunction in bipolar disorder: Evidence, pathophysiology and translational implications. Neurosci Biobehav Rev 2016; 68:694-713. [PMID: 27377693 DOI: 10.1016/j.neubiorev.2016.06.040] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 06/26/2016] [Accepted: 06/30/2016] [Indexed: 01/05/2023]
Abstract
Bipolar disorder (BD) is a chronic psychiatric illness characterized by severe and biphasic changes in mood. Several pathophysiological mechanisms have been hypothesized to underpin the neurobiology of BD, including the presence of mitochondrial dysfunction. A confluence of evidence points to an underlying dysfunction of mitochondria, including decreases in mitochondrial respiration, high-energy phosphates and pH; changes in mitochondrial morphology; increases in mitochondrial DNA polymorphisms; and downregulation of nuclear mRNA molecules and proteins involved in mitochondrial respiration. Mitochondria play a pivotal role in neuronal cell survival or death as regulators of both energy metabolism and cell survival and death pathways. Thus, in this review, we discuss the genetic and physiological components of mitochondria and the evidence for mitochondrial abnormalities in BD. The final part of this review discusses mitochondria as a potential target of therapeutic interventions in BD.
Collapse
Affiliation(s)
- Giselli Scaini
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Laboratory of Bioenergetics, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gislaine T Rezin
- Laboratory of Clinical and Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Sul de Santa Catarina, Tubarão, SC, Brazil
| | - Andre F Carvalho
- Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Emilio L Streck
- Laboratory of Bioenergetics, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, Faculty of Health, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health and The Centre for Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
143
|
Abstract
The etiology and pathophysiology of schizophrenia and related mental disorders such as bipolar disorder and major depression remain largely unclear. Recent advances in mRNA profiling techniques made it possible to perform genome-wide gene expression analysis in a hypothesis-free manner. It was thought that this large-scale data mining approach would reveal unknown molecular cascades involved in mental disorders. Contrary to this initial expectation, however, DNA microarray results in psychiatric fields have been notoriously discordant. Here the authors review the findings of DNA microarray analysis, focusing on systematic gene expression changes in schizophrenia, as well as alterations in the expression of specific genes, that have been reported and replicated. The authors also address the probable causes for the discordance among studies, possible ways to solve the problem, and their preferred approach for data interpretation.
Collapse
Affiliation(s)
- Kazuya Iwamoto
- Laboratory for Molecular Dynamics of Mental Disorders, Brain Science Institute, RIKEN, Saitama, Japan.
| | | |
Collapse
|
144
|
Alshammari TK, Alshammari MA, Nenov MN, Hoxha E, Cambiaghi M, Marcinno A, James TF, Singh P, Labate D, Li J, Meltzer HY, Sacchetti B, Tempia F, Laezza F. Genetic deletion of fibroblast growth factor 14 recapitulates phenotypic alterations underlying cognitive impairment associated with schizophrenia. Transl Psychiatry 2016; 6:e806. [PMID: 27163207 PMCID: PMC5070049 DOI: 10.1038/tp.2016.66] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/25/2016] [Accepted: 03/05/2016] [Indexed: 12/14/2022] Open
Abstract
Cognitive processing is highly dependent on the functional integrity of gamma-amino-butyric acid (GABA) interneurons in the brain. These cells regulate excitability and synaptic plasticity of principal neurons balancing the excitatory/inhibitory tone of cortical networks. Reduced function of parvalbumin (PV) interneurons and disruption of GABAergic synapses in the cortical circuitry result in desynchronized network activity associated with cognitive impairment across many psychiatric disorders, including schizophrenia. However, the mechanisms underlying these complex phenotypes are still poorly understood. Here we show that in animal models, genetic deletion of fibroblast growth factor 14 (Fgf14), a regulator of neuronal excitability and synaptic transmission, leads to loss of PV interneurons in the CA1 hippocampal region, a critical area for cognitive function. Strikingly, this cellular phenotype associates with decreased expression of glutamic acid decarboxylase 67 (GAD67) and vesicular GABA transporter (VGAT) and also coincides with disrupted CA1 inhibitory circuitry, reduced in vivo gamma frequency oscillations and impaired working memory. Bioinformatics analysis of schizophrenia transcriptomics revealed functional co-clustering of FGF14 and genes enriched within the GABAergic pathway along with correlatively decreased expression of FGF14, PVALB, GAD67 and VGAT in the disease context. These results indicate that Fgf14(-/-) mice recapitulate salient molecular, cellular, functional and behavioral features associated with human cognitive impairment, and FGF14 loss of function might be associated with the biology of complex brain disorders such as schizophrenia.
Collapse
Affiliation(s)
- T K Alshammari
- Pharmacology and Toxicology Graduate Program, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- King Saud University Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - M A Alshammari
- Pharmacology and Toxicology Graduate Program, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- King Saud University Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - M N Nenov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - E Hoxha
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience, University of Torino, Turin, Italy
| | - M Cambiaghi
- Department of Neuroscience, University of Torino, Turin, Italy
| | - A Marcinno
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - T F James
- Department of Neuroscience, University of Torino, Turin, Italy
| | - P Singh
- Department of Mathematics, University of Houston, Houston, TX, USA
| | - D Labate
- Department of Mathematics, University of Houston, Houston, TX, USA
| | - J Li
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - H Y Meltzer
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - B Sacchetti
- Department of Neuroscience, University of Torino, Turin, Italy
| | - F Tempia
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience, University of Torino, Turin, Italy
| | - F Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
145
|
Yoshimi N, Futamura T, Kakumoto K, Salehi AM, Sellgren CM, Holmén-Larsson J, Jakobsson J, Pålsson E, Landén M, Hashimoto K. Blood metabolomics analysis identifies abnormalities in the citric acid cycle, urea cycle, and amino acid metabolism in bipolar disorder. BBA CLINICAL 2016; 5:151-8. [PMID: 27114925 PMCID: PMC4832124 DOI: 10.1016/j.bbacli.2016.03.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 11/24/2022]
Abstract
Background Bipolar disorder (BD) is a severe and debilitating psychiatric disorder. However, the precise biological basis remains unknown, hampering the search for novel biomarkers. We performed a metabolomics analysis to discover novel peripheral biomarkers for BD. Methods We quantified serum levels of 116 metabolites in mood-stabilized male BD patients (n = 54) and age-matched male healthy controls (n = 39). Results After multivariate logistic regression, serum levels of pyruvate, N-acetylglutamic acid, α-ketoglutarate, and arginine were significantly higher in BD patients than in healthy controls. Conversely, serum levels of β-alanine, and serine were significantly lower in BD patients than in healthy controls. Chronic (4-weeks) administration of lithium or valproic acid to adult male rats did not alter serum levels of pyruvate, N-acetylglutamic acid, β-alanine, serine, or arginine, but lithium administration significantly increased serum levels of α-ketoglutarate. Conclusions The metabolomics analysis demonstrated altered serum levels of pyruvate, N-acetylglutamic acid, β-alanine, serine, and arginine in BD patients. General significance The present findings suggest that abnormalities in the citric acid cycle, urea cycle, and amino acid metabolism play a role in the pathogenesis of BD. Metabolomics analysis of serum sample from bipolar disorder (BD) was performed. Pyruvate, N-acetylglutamic acid, α-ketoglutarate, and arginine were higher in BD. β-alanine, and serine were lower in BD patients. Abnormalities in citric acid cycle, urea cycle, and amino acid metabolism in BD.
Collapse
Affiliation(s)
- Noriko Yoshimi
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan; Department of CNS Research, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Takashi Futamura
- Department of CNS Research, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Keiji Kakumoto
- Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Alireza M Salehi
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Carl M Sellgren
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jessica Holmén-Larsson
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Joel Jakobsson
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Erik Pålsson
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Mikael Landén
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| |
Collapse
|
146
|
Dudley J, DelBello MP, Weber WA, Adler CM, Strakowski SM, Lee JH. Tissue-dependent cerebral energy metabolism in adolescents with bipolar disorder. J Affect Disord 2016; 191:248-55. [PMID: 26688494 DOI: 10.1016/j.jad.2015.11.045] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 11/18/2015] [Accepted: 11/21/2015] [Indexed: 01/14/2023]
Abstract
OBJECTIVES To investigate tissue-dependent cerebral energy metabolism by measuring high energy phosphate levels in unmedicated adolescents diagnosed with bipolar I disorder. METHODS Phosphorus-31 magnetic resonance spectroscopic imaging data were acquired over the entire brain of 24 adolescents with bipolar I disorder and 19 demographically matched healthy comparison adolescents. Estimates of phosphocreatine (PCr) and adenosine triphosphate (ATP, determined from the γ-resonance) in homogeneous gray and white matter in the right and left hemispheres of the cerebrum of each subject were obtained by extrapolation of linear regression analyses of metabolite concentrations vs. voxel gray matter fractions. RESULTS Multivariate analyses of variance showed a significant effect of group on high energy phosphate concentrations in the right cerebrum (p=0.0002) but not in the left (p=0.17). Post-hoc testing in the right cerebrum revealed significantly reduced concentrations of PCr in gray matter and ATP in white matter in both manic (p=0.002 and 0.0001, respectively) and euthymic (p=0.004 and 0.002, respectively) bipolar I disorder subjects relative to healthy comparisons. LIMITATIONS The small sample sizes yield relatively low statistical power between manic and euthymic groups; cross-sectional observations limit the ability to determine if these findings are truly independent of mood state. CONCLUSIONS Our results suggest bioenergetic impairment - consistent with downregulation of creatine kinase - is an early pathophysiological feature of bipolar I disorder.
Collapse
Affiliation(s)
| | - Melissa P DelBello
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, USA
| | - Wade A Weber
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, USA
| | - Caleb M Adler
- Center for Imaging Research, University of Cincinnati, USA; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, USA
| | - Stephen M Strakowski
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, USA; Department of Biomedical, Chemical, and Environmental Engineering, University of Cincinnati College of Engineering and Applied Science, USA
| | - Jing-Huei Lee
- Center for Imaging Research, University of Cincinnati, USA; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, USA; Department of Biomedical, Chemical, and Environmental Engineering, University of Cincinnati College of Engineering and Applied Science, USA
| |
Collapse
|
147
|
Nod-like receptor pyrin containing 3 (NLRP3) in the post-mortem frontal cortex from patients with bipolar disorder: A potential mediator between mitochondria and immune-activation. J Psychiatr Res 2016; 72:43-50. [PMID: 26540403 DOI: 10.1016/j.jpsychires.2015.10.015] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 10/08/2015] [Accepted: 10/22/2015] [Indexed: 12/22/2022]
Abstract
Mitochondrial complex I dysfunction, oxidative stress and immune-activation are consistently reported in bipolar disorder (BD). Mitochondrial production of reactive oxygen species was recently linked to activation of an inflammatory redox sensor, the nod-like receptor family pyrin domain-containing 3 (NLRP3). Upon its activation, NLRP3 recruits apoptosis-associated speck-like protein (ASC) and caspase-1 to form the NLRP3-inflammasome, activating IL-1β. This study aimed to examine if immune-activation may be a downstream target of complex I dysfunction through the NLRP3-inflammasome in BD. Post-mortem frontal cortex from patients with BD (N = 9), schizophrenia (N = 10), and non-psychiatric controls (N = 9) were donated from the Harvard Brain Tissue Resource Center. Levels of NLRP3, ASC and caspase-1 were measured by western blotting, ELISA and Luminex. While we found no effects of age, sex or post-mortem delay, lower levels of complex I (F2,25 = 3.46, p < 0.05) and NDUFS7, a subunit of complex I (F2,25 = 4.13, p < 0.05), were found in patients with BD. Mitochondrial NLRP3 (F2,25 = 3.86, p < 0.05) and ASC (F2,25 = 4.61, p < 0.05) levels were higher in patients with BD. However, levels of caspase 1 (F2,25 = 4.13, p < 0.05 for both), IL-1β (F2,25 = 7.05, p < 0.01), IL-6 (F2,25 = 5.48, p < 0.05), TNFα (F2,25 = 7.14, p < 0.01) and IL-10 (F2,25 = 5.02, p < 0.05) were increased in both BD and schizophrenia. These findings suggest that immune-activation in the frontal cortex may occur both in patients with BD and schizophrenia, while complex I dysfunction and NLRP3-inflammasome activation may be more specific to BD.
Collapse
|
148
|
Ren Y, Cui Y, Li X, Wang B, Na L, Shi J, Wang L, Qiu L, Zhang K, Liu G, Xu Y. A co-expression network analysis reveals lncRNA abnormalities in peripheral blood in early-onset schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2015; 63:1-5. [PMID: 25967042 DOI: 10.1016/j.pnpbp.2015.05.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 05/05/2015] [Indexed: 01/22/2023]
Abstract
Long non-coding RNAs (lncRNAs) are emerging as important regulators of gene expression and disease processes especially in neuropsychiatric disorders. To explore the potential regulatory roles of lncRNAs in schizophrenia, we performed an integrated co-expression network analysis on lncRNA and mRNA microarray profiles generated from the peripheral blood samples in 19 drug-naïve first-episode early-onset schizophrenia (EOS) patients and 18 demographically matched typically developing controls (TDCs). Using weighted gene co-expression network analysis (WGCNA), we showed that the lncRNAs were organized into co-expressed modules, and two lncRNA modules were associated with EOS. The mRNA networks were constructed and three disease-associated modules were identified. Gene Ontology (GO) analysis indicated that the mRNAs were highly enriched for mitochondrion and related biological processes. Moreover, our results revealed a significant correlation between lncRNAs and mRNAs using the canonical correlation analysis (CCA). Our results suggest that the convergent lncRNA alteration may be involved in the etiologies of EOS, and mitochondrial dysfunction participates in the pathological process of the disease. Our findings may shed light on the pathogenesis of schizophrenia and facilitate future diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
- Yan Ren
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China; Department of Psychiatry, Shanxi Da Yi Hospital/Affiliated Da Yi Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuehua Cui
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China; Department of Statistics and Probability, Michigan State University, East Lansing, MI, USA
| | - Xinrong Li
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Binhong Wang
- Shanxi Province Mental Health Center/Taiyuan Psychiatric Hospital, Taiyuan, China
| | - Long Na
- Shanxi Province Mental Health Center/Taiyuan Psychiatric Hospital, Taiyuan, China
| | - Junyan Shi
- Shanxi Province Mental Health Center/Taiyuan Psychiatric Hospital, Taiyuan, China
| | - Liang Wang
- Shanxi Province Mental Health Center/Taiyuan Psychiatric Hospital, Taiyuan, China
| | - Lixia Qiu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Kerang Zhang
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Guifen Liu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yong Xu
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
149
|
Distinctive transcriptome alterations of prefrontal pyramidal neurons in schizophrenia and schizoaffective disorder. Mol Psychiatry 2015; 20:1397-405. [PMID: 25560755 PMCID: PMC4492919 DOI: 10.1038/mp.2014.171] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 10/14/2014] [Accepted: 11/12/2014] [Indexed: 12/12/2022]
Abstract
Schizophrenia is associated with alterations in working memory that reflect dysfunction of dorsolateral prefrontal cortex (DLPFC) circuitry. Working memory depends on the activity of excitatory pyramidal cells in DLPFC layer 3 and, to a lesser extent, in layer 5. Although many studies have profiled gene expression in DLPFC gray matter in schizophrenia, little is known about cell-type-specific transcript expression in these two populations of pyramidal cells. We hypothesized that interrogating gene expression, specifically in DLPFC layer 3 or 5 pyramidal cells, would reveal new and/or more robust schizophrenia-associated differences that would provide new insights into the nature of pyramidal cell dysfunction in the illness. We also sought to determine the impact of other variables, such as a diagnosis of schizoaffective disorder or medication use at the time of death, on the patterns of gene expression in pyramidal neurons. Individual pyramidal cells in DLPFC layers 3 or 5 were captured by laser microdissection from 36 subjects with schizophrenia or schizoaffective disorder and matched normal comparison subjects. The mRNA from cell collections was subjected to transcriptome profiling by microarray followed by quantitative PCR validation. Expression of genes involved in mitochondrial (MT) or ubiquitin-proteasome system (UPS) functions were markedly downregulated in the patient group (P-values for MT-related and UPS-related pathways were <10(-7) and <10(-5), respectively). MT-related gene alterations were more prominent in layer 3 pyramidal cells, whereas UPS-related gene alterations were more prominent in layer 5 pyramidal cells. Many of these alterations were not present, or found to a lesser degree, in samples of DLPFC gray matter from the same subjects, suggesting that they are pyramidal cell specific. Furthermore, these findings principally reflected alterations in the schizophrenia subjects were not present or present to a lesser degree in the schizoaffective disorder subjects (diagnosis of schizoaffective disorder was the most significant covariate, P<10(-6)) and were not attributable to factors frequently comorbid with schizophrenia. In summary, our findings reveal expression deficits in MT- and UPS-related genes specific to layer 3 and/or layer 5 pyramidal cells in the DLPFC of schizophrenia subjects. These cell type-specific transcriptome signatures are not characteristic of schizoaffective disorder, providing a potential molecular-cellular basis of differences in clinical phenotypes.
Collapse
|
150
|
Vichaya EG, Molkentine JM, Vermeer DW, Walker AK, Feng R, Holder G, Luu K, Mason RM, Saligan L, Heijnen CJ, Kavelaars A, Mason KA, Lee JH, Dantzer R. Sickness behavior induced by cisplatin chemotherapy and radiotherapy in a murine head and neck cancer model is associated with altered mitochondrial gene expression. Behav Brain Res 2015; 297:241-50. [PMID: 26475509 DOI: 10.1016/j.bbr.2015.10.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/05/2015] [Accepted: 10/08/2015] [Indexed: 11/28/2022]
Abstract
The present study was undertaken to explore the possible mechanisms of the behavioral alterations that develop in response to cancer and to cancer therapy. For this purpose we used a syngeneic heterotopic mouse model of human papilloma virus (HPV)-related head and neck cancer in which cancer therapy is curative. Mice implanted or not with HPV+ tumor cells were exposed to sham treatment or a regimen of cisplatin and radiotherapy (chemoradiation). Sickness was measured by body weight loss and reduced food intake. Motivation was measured by burrowing, a highly prevalent species specific behavior. Tumor-bearing mice showed a gradual decrease in burrowing over time and increased brain and liver inflammatory cytokine mRNA expression by 28 days post tumor implantation. Chemoradiation administered to healthy mice resulted in a mild decrease in burrowing, body weight, and food intake. Chemoradiation in tumor-bearing mice decreased tumor growth and abrogated liver and brain inflammation, but failed to attenuate burrowing deficits. PCR array analysis of selected hypoxia and mitochondrial genes revealed that both the tumor and chemoradiation altered the expression of genes involved in mitochondrial energy metabolism within the liver and brain and increased expression of genes related to HIF-1α signaling within the brain. The most prominent changes in brain mitochondrial genes were noted in tumor-bearing mice treated with chemoradiation. These findings indicate that targeting mitochondrial dysfunction following cancer and cancer therapy may be a strategy for prevention of cancer-related symptoms.
Collapse
Affiliation(s)
- Elisabeth G Vichaya
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 384, Houston, TX 77030, United States.
| | - Jessica M Molkentine
- Department of Experimental Radiation Oncology, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 66, Houston, TX 77030, United States
| | - Daniel W Vermeer
- Cancer Biology Research Center, Sanford Research, 2301 E. 60th St. N., Sioux Falls, SD 57104, United States
| | - Adam K Walker
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 384, Houston, TX 77030, United States
| | - Rebekah Feng
- National Institute of Nursing Research, Building 3, Room 5E14, 3 Center Dr., Bethesda, MD 20892, United States
| | - Gerard Holder
- National Institute of Nursing Research, Building 3, Room 5E14, 3 Center Dr., Bethesda, MD 20892, United States
| | - Katherine Luu
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 384, Houston, TX 77030, United States
| | - Ryan M Mason
- Department of Experimental Radiation Oncology, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 66, Houston, TX 77030, United States
| | - Leo Saligan
- National Institute of Nursing Research, Building 3, Room 5E14, 3 Center Dr., Bethesda, MD 20892, United States
| | - Cobi J Heijnen
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 384, Houston, TX 77030, United States
| | - Annemieke Kavelaars
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 384, Houston, TX 77030, United States
| | - Kathy A Mason
- Department of Experimental Radiation Oncology, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 66, Houston, TX 77030, United States
| | - John H Lee
- Cancer Biology Research Center, Sanford Research, 2301 E. 60th St. N., Sioux Falls, SD 57104, United States
| | - Robert Dantzer
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 384, Houston, TX 77030, United States
| |
Collapse
|