1
|
Li Y, Dang X, Chen R, Teng Z, Wang J, Li S, Yue Y, Mitchell BL, Zeng Y, Yao YG, Li M, Liu Z, Yuan Y, Li T, Zhang Z, Luo XJ. Cross-ancestry genome-wide association study and systems-level integrative analyses implicate new risk genes and therapeutic targets for depression. Nat Hum Behav 2025; 9:806-823. [PMID: 39994458 DOI: 10.1038/s41562-024-02073-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 10/23/2024] [Indexed: 02/26/2025]
Abstract
Deciphering the genetic architecture of depression is pivotal for characterizing the associated pathophysiological processes and development of new therapeutics. Here we conducted a cross-ancestry genome-wide meta-analysis on depression (416,437 cases and 1,308,758 controls) and identified 287 risk loci, of which 49 are new. Variant-level fine mapping prioritized potential causal variants and functional genomic analysis identified variants that regulate the binding of transcription factors. We validated that 80% of the identified functional variants are regulatory variants, and expression quantitative trait loci analysis uncovered the potential target genes regulated by the prioritized risk variants. Gene-level analysis, including transcriptome and proteome-wide association studies, colocalization and Mendelian randomization-based analyses, prioritized potential causal genes and drug targets. Gene prioritization analyses highlighted likely causal genes, including TMEM106B, CTNND1, AREL1 and so on. Pathway analysis indicated significant enrichment of depression risk genes in synapse-related pathways. Finally, knockdown of Tmem106b in mice resulted in depression-like behaviours, supporting the involvement of Tmem106b in depression. Our study identified new risk loci, likely causal variants and genes for depression, providing important insights into the genetic architecture of depression and potential therapeutic targets.
Collapse
Affiliation(s)
- Yifan Li
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Xinglun Dang
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Rui 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, China
| | - Zhaowei Teng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shiwu 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, China
| | - Yingying Yue
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Brittany L Mitchell
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Yong Zeng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, 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, Chinese Academy of Sciences, Kunming, 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, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yonggui Yuan
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
| | - Tao Li
- Affiliated Mental Health Center, Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhijun Zhang
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Xiong-Jian Luo
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Liu D, Zhu C, Wei H. Clozapine and rapamycin reverse behavioral abnormalities in an animal model of autoimmune schizophrenia. Neuropharmacology 2025; 266:110286. [PMID: 39733937 DOI: 10.1016/j.neuropharm.2024.110286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
OBJECTIVE Autoantibody-associated psychosis represents a distinct disease subgroup of patients with schizophrenia with a suspected autoimmune origin. Although preliminary studies have suggested adjunctive drug treatment strategies targeting the immune system, further validation of these findings is warranted. Autoantibodies against SFT2D2 have been identified in patients with schizophrenia. ApoE-/- mice immunized with SFT2D2-peptides can be used as a model for testing immunotherapy in this subgroup of patients. We used the atypical antipsychotic drug clozapine and immunosuppressant rapamycin to test their effects in this mouse model. METHODS The mice were evaluated for cognitive and schizophrenia-like behaviors. Following behavioral testing, brain samples were collected for analyzing specific pathological changes and dendritic spine formation. RESULTS Clozapine and rapamycin reversed impaired pre-pulse inhibition, motor impairment, and improved cognitive ability in ApoE -/- mice exposed to anti-SFT2D2 immunoglobulin G. Immunohistochemical assays revealed that both clozapine and rapamycin significantly reduced activated microglial infiltration and restored neuronal dendritic spine density. CONCLUSIONS Our study results suggested that clozapine and rapamycin possess therapeutic benefits for managing autoimmune psychosis and provide mechanistic insights into immunotherapies involving immunosuppressive agents.
Collapse
Affiliation(s)
- Duilin Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Caiyun Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
3
|
Weller AE, Ferraro TN, Doyle GA, Reiner BC, Berrettini WH, Crist RC. Analysis of single-cell transcriptome data from a mouse model implicates protein synthesis dysfunction in schizophrenia. Genes Genomics 2024; 46:1071-1084. [PMID: 39083157 DOI: 10.1007/s13258-024-01545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/08/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Schizophrenia is a mental disorder that causes considerable morbidity, whose risk largely results from genetic factors. Setd1a is a gene implicated in schizophrenia. OBJECTIVE To study the gene expression changes found in heterozygous Setd1a± knockout mice in order to gain useful insight into schizophrenia pathogenesis. METHODS We mined a single-cell RNA sequencing (scRNAseq) dataset from the prefrontal cortex (PFC) and striatum of Setd1a± mice and identified cell type-specific differentially expressed genes (DEGs) and differential transcript usage (DTU). DEGs and genes containing DTU found in each cell type were used to identify affected biological pathways using Ingenuity Pathway Analysis (IPA). RESULTS We identified 273 unique DEGs across all cell types in PFC and 675 unique gene peaks containing DTU. In striatum, we identified 327 unique DEGs across all cell types and 8 unique gene peaks containing DTU. Key IPA findings from the analysis of DEGs found in PFC and striatum implicate processes involved in protein synthesis, mitochondrial function, cell metabolism, and inflammation. IPA analysis of genes containing DTU in PFC points to protein synthesis, as well as cellular activities involving intracellular signaling and neurotransmission. One canonical pathway, 'EIF2 Signaling', which is involved in the regulation of protein synthesis, was detected in PFC DEGs, striatum DEGs, and PFC genes containing DTU, drawing attention to its importance in schizophrenia pathophysiology. CONCLUSION Processes involving protein synthesis in general and the 'EIF2 Signaling' pathway in particular could be targets for the development of new research strategies and biomarkers in schizophrenia.
Collapse
Affiliation(s)
- Andrew E Weller
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US.
| | - Thomas N Ferraro
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, US
| | - Glenn A Doyle
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, 19111, US
| | - Benjamin C Reiner
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US
| | - Wade H Berrettini
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US
| | - Richard C Crist
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US
| |
Collapse
|
4
|
Yeh CH, Lin PC, Tseng RY, Chao YP, Wu CT, Chou TL, Chen RS, Gau SSF, Ni HC, Lin HY. Lack of effects of eight-week left dorsolateral prefrontal theta burst stimulation on white matter macro/microstructure and connection in autism. Brain Imaging Behav 2024; 18:794-807. [PMID: 38492129 DOI: 10.1007/s11682-024-00874-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2024] [Indexed: 03/18/2024]
Abstract
Whether brain stimulation could modulate brain structure in autism remains unknown. This study explored the impact of continuous theta burst stimulation (cTBS) over the left dorsolateral prefrontal cortex (DLPFC) on white matter macro/microstructure in intellectually able children and emerging adults with autism. Sixty autistic participants were randomized (30 active) and received active or sham cTBS for eight weeks twice per week, 16 total sessions using a double-blind (participant-, rater-, analyst-blinded) design. All participants received high-angular resolution diffusion MR imaging at baseline and week 8. Twenty-eight participants in the active group and twenty-seven in the sham group with good imaging quality entered the final analysis. With longitudinal fixel-based analysis and network-based statistics, we found no significant difference between the active and sham groups in changes of white matter macro/microstructure and connections following cTBS. In addition, we found no association between baseline white matter macro/microstructure and autistic symptom changes from baseline to week 8 in the active group. In conclusion, we did not find a significant impact of left DLPFC cTBS on white matter macro/microstructure and connections in children and emerging adults with autism. These findings need to be interpreted in the context that the current intellectually able cohort in a single university hospital site limits the generalizability. Future studies are required to investigate if higher stimulation intensities and/or doses, other personal factors, or rTMS parameters might confer significant brain structural changes visible on MRI in ASD.
Collapse
Affiliation(s)
- Chun-Hung Yeh
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, No.5 Fusing St. Gueishan, Taoyuan, 333, Taiwan
| | - Po-Chun Lin
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, No.5 Fusing St. Gueishan, Taoyuan, 333, Taiwan
| | - Rung-Yu Tseng
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Ping Chao
- Deparment of Computer Science and Information Engineering, Chang Gung University, Taoyuan, Taiwan
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chen-Te Wu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Tai-Li Chou
- Department of Psychology, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| | - Rou-Shayn Chen
- Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Susan Shur-Fen Gau
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Hsing-Chang Ni
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, No.5 Fusing St. Gueishan, Taoyuan, 333, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Hsiang-Yuan Lin
- Azrieli Adult Neurodevelopmental Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Riggs LM, Pereira EFR, Thompson SM, Gould TD. cAMP-dependent protein kinase signaling is required for ( 2R,6R)-hydroxynorketamine to potentiate hippocampal glutamatergic transmission. J Neurophysiol 2024; 131:64-74. [PMID: 38050689 PMCID: PMC11286304 DOI: 10.1152/jn.00326.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023] Open
Abstract
(2R,6R)-Hydroxynorketamine (HNK) is a ketamine metabolite that shows rapid antidepressant-like effects in preclinical studies and lacks the adverse N-methyl-d-aspartate receptor (NMDAR) inhibition-related properties of ketamine. Investigating how (2R,6R)-HNK exerts its antidepressant actions may be informative in the design of novel pharmacotherapies with improved safety and efficacy. We sought to identify the molecular substrates through which (2R,6R)-HNK induces functional changes at excitatory synapses, a prevailing hypothesis for how rapid antidepressant effects are initiated. We recorded excitatory postsynaptic potentials in hippocampal slices from male Wistar Kyoto rats, which have impaired hippocampal plasticity and are resistant to traditional antidepressants. (2R,6R)-HNK (10 µM) led to a rapid potentiation of electrically evoked excitatory postsynaptic potentials at Schaffer collateral CA1 stratum radiatum synapses. This potentiation was associated with a decrease in paired pulse facilitation, suggesting an increase in the probability of glutamate release. The (2R,6R)-HNK-induced potentiation was blocked by inhibiting either cyclic adenosine monophosphate (cAMP) or its downstream target, cAMP-dependent protein kinase (PKA). As cAMP is a potent regulator of brain-derived neurotrophic factor (BDNF) release, we assessed whether (2R,6R)-HNK exerts this acute potentiation through a rapid increase in cAMP-dependent BDNF-TrkB signaling. We found that the cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the acute synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions in vivo. These results suggest that, by potentiating glutamate release via cAMP-PKA signaling, (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission that promote structural plasticity leading to maintained antidepressant action.NEW & NOTEWORTHY Ketamine is a rapid-acting antidepressant and its preclinical effects are mimicked by its (2R,6R)-(HNK) metabolite. We found that (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission by potentiating glutamate release via cAMP-PKA signaling at hippocampal Schaffer collateral synapses. This cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the rapid synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions that are thought to maintain antidepressant action in vivo.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Edna F R Pereira
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Veterans Affairs Maryland Health Care System, Baltimore, Maryland, United States
| |
Collapse
|
6
|
Deng K, Wu M. Leucine-rich repeats containing 4 protein (LRRC4) in memory, psychoneurosis, and glioblastoma. Chin Med J (Engl) 2023; 136:4-12. [PMID: 36780420 PMCID: PMC10106153 DOI: 10.1097/cm9.0000000000002441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Indexed: 02/15/2023] Open
Abstract
ABSTRACT Leucine-rich repeats containing 4 ( LRRC4 , also named netrin-G ligand 2 [NGL-2]) is a member of the NetrinGs ligands (NGLs) family. As a gene with relatively high and specific expression in brain, it is a member of the leucine-rich repeat superfamily and has been proven to be a suppressor gene for gliomas, thus being involved in gliomagenesis. LRRC4 is the core of microRNA-dependent multi-phase regulatory loops that inhibit the proliferation and invasion of glioblastoma (GB) cells, including LRRC4/NGL2-activator protein 2 (AP2)-microRNA (miR) 182-LRRC4 and LRRC4-miR185-DNA methyltransferase 1 (DNMT1)-LRRC4/specific protein 1 (SP1)-DNMT1-LRRC4. In this review, we demonstrated LRRC4 as a new member of the partitioning-defective protein (PAR) polarity complex that promotes axon differentiation, mediates the formation and plasticity of synapses, and assists information input to the hippocampus and storage of memory. As an important synapse regulator, aberrant expression of LRRC4 has been detected in autism, spinal injury and GBs. LRRC4 is a candidate susceptibility gene for autism and a neuro-protective factor in spinal nerve damage. In GBs, LRRC4 is a novel inhibitor of autophagy, and an inhibitor of protein-protein interactions involving in temozolomide resistance, tumor immune microenvironment, and formation of circular RNA.
Collapse
Affiliation(s)
- Kun Deng
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
| | - Minghua Wu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
7
|
Gong W, Wang K, Wang X, Chen Y, Qin X, Lu A, Guan D. Mathematical algorithm–based identification of the functional components and mechanisms in depression treatment: An example of Danggui-Shaoyao-San. Front Cell Dev Biol 2022; 10:937621. [PMID: 36072347 PMCID: PMC9441958 DOI: 10.3389/fcell.2022.937621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/14/2022] [Indexed: 12/02/2022] Open
Abstract
Depression, a complex epidemiological mental disorder, affects around 350 million people worldwide. Despite the availability of antidepressants based on monoamine hypothesis of depression, most patients suffer side effects from these drugs, including psychomotor impairment and dependence liability. Traditional Chinese medicine (TCM) is receiving more and more attention due to the advantages of high therapeutic performance and few side effects in depression treatment. However, complex multicomponents and multi-targets in TCM hinder our ability to identify the functional components and molecular mechanisms of its efficacy. In this study, we designed a novel strategy to capture the functional components and mechanisms of TCM based on a mathematical algorithm. To establish proof of principle, the TCM formula Danggui-Shaoyao-San (DSS), which possesses remarkable antidepressant effect but its functional components and mechanisms are unclear, is used as an example. According to the network motif detection algorithm, key core function motifs (CIM) of DSS in treating depression were captured, followed by a functional analysis and verification. The results demonstrated that 198 pathways were enriched by the target genes of the CIM, and 179 coincided with the enriched pathways of pathogenic genes, accounting for 90.40% of the gene enrichment pathway of the C-T network. Then the functional components group (FCG) comprising 40 components was traced from CIM based on the target coverage accumulation algorithm, after which the pathways enriched by the target genes of FCG were selected to elucidate the potential mechanisms of DSS in treating depression. Finally, the pivotal components in FCG of DSS and the related pathways were selected for experimental validation in vitro and in vivo. Our results indicated good accuracy of the proposed mathematical algorithm in sifting the FCG from the TCM formula, which provided a methodological reference for discovering functional components and interpreting molecular mechanisms of the TCM formula in treating complex diseases.
Collapse
Affiliation(s)
- Wenxia Gong
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, Shanxi, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Taiyuan, Shanxi, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, Shanxi, China
| | - Kexin Wang
- National Key Clinical Specialty/Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Neurosurgery Institute, Guangzhou, China
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xueyuan Wang
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, Shanxi, China
| | - Yupeng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, Shanxi, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Taiyuan, Shanxi, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, Shanxi, China
- *Correspondence: Xuemei Qin, ; Aiping Lu, ; Daogang Guan,
| | - Aiping Lu
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong, China
- *Correspondence: Xuemei Qin, ; Aiping Lu, ; Daogang Guan,
| | - Daogang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
- *Correspondence: Xuemei Qin, ; Aiping Lu, ; Daogang Guan,
| |
Collapse
|
8
|
Camporesi E, Nilsson J, Vrillon A, Cognat E, Hourregue C, Zetterberg H, Blennow K, Becker B, Brinkmalm A, Paquet C, Brinkmalm G. Quantification of the trans-synaptic partners neurexin-neuroligin in CSF of neurodegenerative diseases by parallel reaction monitoring mass spectrometry. EBioMedicine 2022; 75:103793. [PMID: 34990894 PMCID: PMC8743209 DOI: 10.1016/j.ebiom.2021.103793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Synaptic proteins are increasingly studied as biomarkers for synaptic dysfunction and loss, which are early and central events in Alzheimer's disease (AD) and strongly correlate with the degree of cognitive decline. In this study, we specifically investigated the synaptic binding partners neurexin (NRXN) and neuroligin (Nlgn) proteins, to assess their biomarker's potential. METHODS we developed a parallel reaction monitoring mass spectrometric method for the simultaneous quantification of NRXNs and Nlgns in cerebrospinal fluid (CSF) of neurodegenerative diseases, focusing on AD. Specifically, NRXN-1α, NRXN-1β, NRXN-2α, NRXN-3α and Nlgn1, Nlgn2, Nlgn3 and Nlgn4 proteins were targeted. FINDINGS The proteins were investigated in a clinical cohort including CSF from controls (n=22), mild cognitive impairment (MCI) due to AD (n=44), MCI due to other conditions (n=46), AD (n=77) and a group of non-AD dementia (n=28). No difference in levels of NRXNs and Nlgns was found between AD (both at dementia and MCI stages) or controls or the non-AD dementia group for any of the targeted proteins. NRXN and Nlgn proteins correlated strongly with each other, but only a weak correlation with the AD core biomarkers and the synaptic biomarkers neurogranin and growth-associated protein 43, was found, possibly reflecting different pathogenic processing at the synapse. INTERPRETATION we conclude that NRXN and Nlgn proteins do not represent suitable biomarkers for synaptic pathology in AD. The panel developed here could aid in future investigations of the potential involvement of NRXNs and Nlgns in synaptic dysfunction in other disorders of the central nervous system. FUNDING a full list of funding can be found under the acknowledgments section.
Collapse
Affiliation(s)
- Elena Camporesi
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Johanna Nilsson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Agathe Vrillon
- Université de Paris, Cognitive Neurology Center, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France; Université de Paris, Inserm UMR S11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Emmanuel Cognat
- Université de Paris, Cognitive Neurology Center, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France; Université de Paris, Inserm UMR S11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Claire Hourregue
- Université de Paris, Cognitive Neurology Center, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Bruno Becker
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Claire Paquet
- Université de Paris, Cognitive Neurology Center, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France; Université de Paris, Inserm UMR S11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
9
|
Wang QQ, Cheng L, Wu BY, Xu P, Qiu HY, Wang B, Yan XJ, Chen SL. Short-course antidepressant therapy reduces discontinuation syndrome while maintaining treatment efficacy in patients with refractory functional dyspepsia: A randomized controlled trial. Front Psychiatry 2022; 13:1063722. [PMID: 36569610 PMCID: PMC9772443 DOI: 10.3389/fpsyt.2022.1063722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Long-course (LC) antidepressants for the treatment of disorders of gut-brain interaction, such as refractory functional dyspepsia (rFD), pose patients at risk of antidepressant discontinuation syndrome (ADS). Short-course (SC) therapy of rapid-acting antidepressant may reduce discontinuation syndromes while maintaining efficacy for dyspeptic symptoms. However, the evidence-based research is lacking. This study aims to determine whether SC therapy with antidepressants could decrease the risk of ADS with comparable treatment efficacy to LC therapy in rFD. METHODS This randomized clinical trial with rFD patients was conducted at a tertiary hospital in China. Participants (N = 240) were randomly allocated to receive flupentixol-melitracen (FM) plus omeprazole therapy for 2 (SC group) or 4 (LC group) weeks, respectively. Scores for Leeds Dyspepsia Questionnaire (LDQ), Generalized Anxiety Disorder-7 (GAD-7) and Patient Health Questionnaire-9 for Depression (PHQ-9) were assessed at baseline and every 2 weeks, ending at 4 weeks after treatment. ADS was assessed after drug cessation. Medication possession ratio (MPR) for FM was calculated. RESULTS The severity and incidence of ADS of patients in SC group were significantly lower than those in LC group (0.60 ± 0.62 vs. 1.71 ± 1.58 and 3.64 vs. 39.45%; both P < 0.0001). The MPR values for FM were significantly higher in patients of SC group than in LC group (P < 0.0001). Scores for LDQ, GAD-7 and PHQ-9 decreased in patients of both groups, and the symptom improvement in SC group was comparable to that in LC group after treatment. CONCLUSIONS Compared to 4-week FM therapy, the 2-week FM therapy reduces the risk of ADS with non-inferior treatment efficacy in patients with rFD. CLINICAL TRIAL REGISTRATION Clinical trials.gov, identifier NCT05099913.
Collapse
Affiliation(s)
- Qian-Qian Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Cheng
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bi-Yu Wu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Xu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong-Yi Qiu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiu-Juan Yan
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng-Liang Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
10
|
Cui F, Gu S, Gu Y, Yin J, Fang C, Liu L. Alteration in the mRNA expression profile of the autophagy-related mTOR pathway in schizophrenia patients treated with olanzapine. BMC Psychiatry 2021; 21:388. [PMID: 34348681 PMCID: PMC8335969 DOI: 10.1186/s12888-021-03394-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/26/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mammalian target of rapamycin protein (mTOR) signaling pathway is involved in the pathogenesis of schizophrenia and the mechanism of extrapyramidal adverse reactions to antipsychotic drugs, which might be mediated by an mTOR-dependent autophagy impairment. This study aimed to examine the expression of mTOR pathway genes in patients with schizophrenia treated with olanzapine, which is considered an mTOR inhibitor and autophagy inducer. METHODS Thirty-two patients with acute schizophrenia who had been treated with olanzapine for four weeks (average dose 14.24 ± 4.35 mg/d) and 32 healthy volunteers were recruited. Before and after olanzapine treatment, the Positive and Negative Syndrome Scale (PANSS) was used to evaluate the symptoms of patients with schizophrenia, and the mRNA expression levels of mTOR pathway-related genes, including MTOR, RICTOR, RAPTOR, and DEPTOR, were detected in fasting venous blood samples from all subjects using real-time quantitative PCR. RESULTS The MTOR and RICTOR mRNA expression levels in patients with acute schizophrenia were significantly decreased compared with those of healthy controls and further significantly decreased after four weeks of olanzapine treatment. The DEPTOR mRNA expression levels in patients with acute schizophrenia were not significantly different from those of healthy controls but were significantly increased after treatment. The expression levels of the RAPTOR mRNA were not significantly different among the three groups. The pairwise correlations of MTOR, DEPTOR, RAPTOR, and RICTOR mRNA expression levels in patients with acute schizophrenia and healthy controls were significant. After olanzapine treatment, the correlations between the expression levels of the DEPTOR and MTOR mRNAs and between the DEPTOR and RICTOR mRNAs disappeared. CONCLUSIONS Abnormalities in the mTOR pathway, especially DEPTOR and mTORC2, might play important roles in the autophagy mechanism underlying the pathophysiology of schizophrenia and effects of olanzapine treatment.
Collapse
Affiliation(s)
- Fengwei Cui
- grid.89957.3a0000 0000 9255 8984Department of Geriatric Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151 Jiangsu China
| | - Shuguang Gu
- grid.89957.3a0000 0000 9255 8984Department of Geriatric Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151 Jiangsu China
| | - Yue Gu
- grid.89957.3a0000 0000 9255 8984The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166 Jiangsu China
| | - Jiajun Yin
- grid.89957.3a0000 0000 9255 8984Department of Geriatric Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151 Jiangsu China
| | - Chunxia Fang
- Combined TCM & Western Medicine Department, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151, Jiangsu, China.
| | - Liang Liu
- Department of Geriatric Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151, Jiangsu, China.
| |
Collapse
|
11
|
Zhao X, Wilson K, Uteshev V, He JJ. Activation of α7 nicotinic acetylcholine receptor ameliorates HIV-associated neurology and neuropathology. Brain 2021; 144:3355-3370. [PMID: 34196664 PMCID: PMC8677536 DOI: 10.1093/brain/awab251] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/28/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
HIV-associated neurocognitive disorders (HAND) in the era of combination antiretroviral therapy are primarily manifested as impaired behaviours, glial activation/neuroinflammation and compromised neuronal integrity, for which there are no effective treatments currently available. In the current study, we used doxycycline-inducible astrocyte-specific HIV Tat transgenic mice (iTat), a surrogate HAND model, and determined effects of PNU-125096, a positive allosteric modulator of α7 nicotinic acetylcholine receptor (α7 nAChR) on Tat-induced behavioural impairments and neuropathologies. We showed that PNU-125096 treatment significantly improved locomotor, learning and memory deficits of iTat mice while inhibited glial activation and increased PSD-95 expression in the cortex and hippocampus of iTat mice. Using α7 nAChR knockout mice, we showed that α7 nAChR knockout eliminated the protective effects of PNU-125096 on iTat mice. In addition, we showed that inhibition of p38 phosphorylation by SB239063, a p38 MAPK-specific inhibitor exacerbated Tat neurotoxicity in iTat mice. Last, we used primary mouse cortical individual cultures and neuron-astrocytes co-cultures and in vivo staining of iTat mouse brain tissues and showed that glial activation was directly involved in the interplay among Tat neurotoxicity, α7 nAChR activation and the p38 MAPK signalling pathway. Taken together, these findings demonstrated for the first time that α7 nAChR activation led to protection against HAND and suggested that α7 nAChR modulator PNU-125096 holds significant promise for development of therapeutics for HAND.
Collapse
Affiliation(s)
- Xiaojie Zhao
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Kelly Wilson
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Victor Uteshev
- Department of Pharmacology and Neuroscience, Graduate School of Biomedical Sciences of University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Johnny J He
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA
| |
Collapse
|
12
|
Rebiai R, Givogri MI, Gowrishankar S, Cologna SM, Alford ST, Bongarzone ER. Synaptic Function and Dysfunction in Lysosomal Storage Diseases. Front Cell Neurosci 2021; 15:619777. [PMID: 33746713 PMCID: PMC7978225 DOI: 10.3389/fncel.2021.619777] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
Lysosomal storage diseases (LSDs) with neurological involvement are inherited genetic diseases of the metabolism characterized by lysosomal dysfunction and the accumulation of undegraded substrates altering glial and neuronal function. Often, patients with neurological manifestations present with damage to the gray and white matter and irreversible neuronal decline. The use of animal models of LSDs has greatly facilitated studying and identifying potential mechanisms of neuronal dysfunction, including alterations in availability and function of synaptic proteins, modifications of membrane structure, deficits in docking, exocytosis, recycling of synaptic vesicles, and inflammation-mediated remodeling of synapses. Although some extrapolations from findings in adult-onset conditions such as Alzheimer's disease or Parkinson's disease have been reported, the pathogenetic mechanisms underpinning cognitive deficits in LSDs are still largely unclear. Without being fully inclusive, the goal of this mini-review is to present a discussion on possible mechanisms leading to synaptic dysfunction in LSDs.
Collapse
Affiliation(s)
- Rima Rebiai
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Maria I. Givogri
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Swetha Gowrishankar
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Stephania M. Cologna
- Department of Chemistry, College of Liberal Arts and Sciences, The University of Illinois at Chicago, Chicago, IL, United States
| | - Simon T. Alford
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Ernesto R. Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
13
|
Al Shweiki MR, Oeckl P, Steinacker P, Barschke P, Dorner-Ciossek C, Hengerer B, Schönfeldt-Lecuona C, Otto M. Proteomic analysis reveals a biosignature of decreased synaptic protein in cerebrospinal fluid of major depressive disorder. Transl Psychiatry 2020; 10:144. [PMID: 32398672 PMCID: PMC7217933 DOI: 10.1038/s41398-020-0825-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/16/2020] [Accepted: 04/01/2020] [Indexed: 01/06/2023] Open
Abstract
Major depressive disorder (MDD) is a leading cause of morbidity with a lifetime prevalence of 10%. There is increasing evidence suggesting synaptic dysfunction and impaired integrity of certain brain circuits in MDD. Here we investigate the cerebrospinal fluid proteome of psychiatric patients focusing on MDD by deep proteomic profiling approach combined with a further validation step using targeted mass spectrometry. We demonstrate profound CSF proteomic changes during on-going depression episodes in MDD patients (n = 40) in comparison to controls (n = 27), schizophrenia spectrum disorder (n = 13), and bipolar disorder patients (n = 11). The discovery analysis with isobaric tags for relative and absolute quantitation (iTRAQ) reveals changes in proteins associated with synaptic transmission, myelination, and Wnt signaling in CSF of MDD. The multiple reaction monitoring (MRM) validation analysis confirms significantly decreased levels of eight proteins including the membrane synaptic proteins neurexin 3 (NRXN3), contactin-associated protein-like 4 (CNTNAP4), and glutamate ionotropic receptor AMPA type subunit 4 (GRIA4) in the CSF of MDD patients in comparison to the controls. Overall, the study demonstrates proteins that constitute an MDD biosignature for further validation studies and provides insight into the pathophysiology of MDD and other psychiatric disorders.
Collapse
Affiliation(s)
| | - Patrick Oeckl
- Department of Neurology, Ulm University, Ulm, Germany
| | | | | | - Cornelia Dorner-Ciossek
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Bastian Hengerer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany.
| |
Collapse
|
14
|
Oxidation-reduction mechanisms in psychiatric disorders: A novel target for pharmacological intervention. Pharmacol Ther 2020; 210:107520. [PMID: 32165136 DOI: 10.1016/j.pharmthera.2020.107520] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/02/2020] [Indexed: 12/16/2022]
Abstract
While neurotransmitter dysfunction represents a key component in mental illnesses, there is now a wide agreement for a central pathophysiological hub that includes hormones, neuroinflammation, redox mechanisms as well as oxidative stress. With respect to oxidation-reduction (redox) mechanisms, preclinical and clinical evidence suggests that an imbalance in the pro/anti-oxidative homeostasis toward the increased production of substances with oxidizing potential may contribute to the etiology and manifestation of different psychiatric disorders. The substantial and continous demand for energy renders the brain highly susceptible to disturbances in its energy supply, especially following exposure to stressful events, which may lead to overproduction of reactive oxygen and nitrogen species under conditions of perturbed antioxidant defenses. This will eventually induce different molecular alterations, including extensive protein and lipid peroxidation, increased blood-brain barrier permeability and neuroinflammation, which may contribute to the changes in brain function and morphology observed in mental illnesses. This view may also reconcile different key concepts for psychiatric disorders, such as the neurodevelopmental origin of these diseases, as well as the vulnerability of selective cellular populations that are critical for specific functional abnormalities. The possibility to pharmacologically modulate the redox system is receiving increasing interest as a novel therapeutic strategy to counteract the detrimental effects of the unbalance in brain oxidative mechanisms. This review will describe the main mechanisms and mediators of the redox system and will examine the alterations of oxidative stress found in animal models of psychiatric disorders as well as in patients suffering from mental illnesses, such as schizophrenia and major depressive disorder. In addition, it will discuss studies that examined the effects of psychotropic drugs, including antipsychotics and antidepressants, on the oxidative balance as well as studies that investigated the effectiveness of a direct modulation of oxidative mechanisms in counteracting the behavioral and functional alterations associated with psychiatric disorders, which supports the promising role of the redox system as a novel therapeutic target for the improved treatment of brain disorders.
Collapse
|
15
|
Erli F, Palmos AB, Raval P, Mukherjee J, Sellers KJ, Gatford NJF, Moss SJ, Brandon NJ, Penzes P, Srivastava DP. Estradiol reverses excitatory synapse loss in a cellular model of neuropsychiatric disorders. Transl Psychiatry 2020; 10:16. [PMID: 32066698 PMCID: PMC7026123 DOI: 10.1038/s41398-020-0682-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/25/2022] Open
Abstract
Loss of glutamatergic synapses is thought to be a key cellular pathology associated with neuropsychiatric disorders including schizophrenia (SCZ) and major depressive disorder (MDD). Genetic and cellular studies of SCZ and MDD using in vivo and in vitro systems have supported a key role for dysfunction of excitatory synapses in the pathophysiology of these disorders. Recent clinical studies have demonstrated that the estrogen, 17β-estradiol can ameliorate many of the symptoms experienced by patients. Yet, to date, our understanding of how 17β-estradiol exerted these beneficial effects is limited. In this study, we have tested the hypothesis that 17β-estradiol can restore dendritic spine number in a cellular model that recapitulates the loss of synapses associated with SCZ and MDD. Ectopic expression of wildtype, mutant or shRNA-mediated knockdown of Disrupted in Schizophrenia 1 (DISC1) reduced dendritic spine density in primary cortical neurons. Acute or chronic treatment with 17β-estradiol increased spine density to control levels in neurons with altered DISC1 levels. In addition, 17β-estradiol reduced the extent to which ectopic wildtype and mutant DISC1 aggregated. Furthermore, 17β-estradiol also caused the enrichment of synaptic proteins at synapses and increased the number of dendritic spines containing PSD-95 or that overlapped with the pre-synaptic marker bassoon. Taken together, our data indicates that estrogens can restore lost excitatory synapses caused by altered DISC1 expression, potentially through the trafficking of DISC1 and its interacting partners. These data highlight the possibility that estrogens exert their beneficial effects in SCZ and MDD in part by modulating dendritic spine number.
Collapse
Affiliation(s)
- Filippo Erli
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 9RT UK
| | - Alish B. Palmos
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 9RT UK
| | - Pooja Raval
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 9RT UK
| | - Jayanta Mukherjee
- grid.429997.80000 0004 1936 7531AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University, Boston, MA UK
| | - Katherine J. Sellers
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 9RT UK
| | - Nicholas J. F. Gatford
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 9RT UK
| | - Stephen J. Moss
- grid.429997.80000 0004 1936 7531AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University, Boston, MA UK
| | - Nicholas J. Brandon
- grid.429997.80000 0004 1936 7531AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University, Boston, MA UK ,grid.417815.e0000 0004 5929 4381Neuroscience, IMED Biotech Unit, AstraZeneca, Boston, MA UK
| | - Peter Penzes
- grid.16753.360000 0001 2299 3507Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Centre for Autism and Neurodevelopment, Northwestern University, Chicago, IL USA
| | - Deepak P. Srivastava
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 9RT UK ,grid.16753.360000 0001 2299 3507Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL USA ,grid.13097.3c0000 0001 2322 6764MRC Centre for Neurodevelopmental Disorders, King’s College London, London, SE1 1UL UK
| |
Collapse
|
16
|
Brivio P, Paladini MS, Racagni G, Riva MA, Calabrese F, Molteni R. From Healthy Aging to Frailty: In Search of the Underlying Mechanisms. Curr Med Chem 2019; 26:3685-3701. [PMID: 31333079 DOI: 10.2174/0929867326666190717152739] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/14/2018] [Accepted: 03/08/2019] [Indexed: 11/22/2022]
Abstract
Population aging is accelerating rapidly worldwide, from 461 million people older than 65 years in 2004 to an estimated 2 billion people by 2050, leading to critical implications for the planning and delivery of health and social care. The most problematic expression of population aging is the clinical condition of frailty, which is a state of increased vulnerability that develops as a consequence of the accumulation of microscopic damages in many physiological systems that lead to a striking and disproportionate change in health state, even after an apparently small insult. Since little is known about the biology of frailty, an important perspective to understand this phenomenon is to establish how the alterations that physiologically occur during a condition of healthy aging may instead promote cumulative decline with subsequent depletion of homoeostatic reserve and increase the vulnerability also after minor stressor events. In this context, the present review aims to provide a description of the molecular mechanisms that, by having a critical impact on behavior and neuronal function in aging, might be relevant for the development of frailty. Moreover, since these biological systems are also involved in the coping strategies set in motion to respond to environmental challenges, we propose a role for lifestyle stress as an important player to drive frailty in aging.
Collapse
Affiliation(s)
- Paola Brivio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maria Serena Paladini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giorgio Racagni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.,Associazione di Psicofarmacologia, Milan, Italy
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
17
|
Du X, Hill RA. Hypothalamic-pituitary-gonadal axis dysfunction: An innate pathophysiology of schizophrenia? Gen Comp Endocrinol 2019; 275:38-43. [PMID: 30753842 DOI: 10.1016/j.ygcen.2019.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 12/21/2022]
Abstract
The female hormone 17β-estradiol is postulated to be protective against schizophrenia onset and severity. Hypoestrogenism is a common phenomenon in women with schizophrenia that has serious effects that adds to the burden of an already very onerous disease. The cause of hypoestrogenism is largely attributed to antipsychotic-induced hyperprolactinemia. Evidence suggest however that a significant portion of female schizophrenia patients develop hypoestrogenism either before antipsychotic treatment or without regard to the level of prolactin, suggesting that for a sizeable segment of female patients, gonadal abnormality may be an innate and early aspect of the disease. This review aims to summarise the available literature that examines gonadal dysfunction in schizophrenia through this prism as well as to outline some recent developments in treatment strategies that may provide feasible ways to successfully tackle hypoestrogenism in schizophrenia.
Collapse
Affiliation(s)
- X Du
- Behavioural Neuroscience Laboratory, Department of Psychiatry, Monash University, Clayton, Victoria 3168, Australia.
| | - R A Hill
- Behavioural Neuroscience Laboratory, Department of Psychiatry, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
18
|
Yang Y, Liu Y, Wang G, Hei G, Wang X, Li R, Li L, Wu R, Zhao J. Brain-derived neurotrophic factor is associated with cognitive impairments in first-episode and chronic schizophrenia. Psychiatry Res 2019; 273:528-536. [PMID: 30710808 DOI: 10.1016/j.psychres.2019.01.051] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/11/2018] [Accepted: 01/13/2019] [Indexed: 01/16/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) may be related to the pathophysiology of schizophrenia. This study aims to examine the relation between plasma BDNF levels and the cognition of patients with schizophrenia. We recruited 31 patients with chronic schizophrenia, 34 first-episode patients, and 35 healthy control subjects. We examined the MATRICS Consensus Cognitive Battery (MCCB) and the plasma BDNF levels in all groups. The schizophrenic symptoms were assessed using the positive and negative syndrome scale. The BDNF levels of schizophrenic patients were remarkably lower than those of the controls. The cognitive MCCB global composite scores and part index scores of schizophrenic patients were remarkably lower than those of the controls. Moreover, remarkable correlations were observed between BDNF levels and partial cognitive dimensions, such as visual learning, memory, and processing speed. Therefore, BDNF may be involved in the pathophysiology and cognitive impairment of schizophrenia.
Collapse
Affiliation(s)
- Ye Yang
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Yi Liu
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Guodong Wang
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Gangrui Hei
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Xiaoyi Wang
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Ranran Li
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Li Li
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Renrong Wu
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.
| | - Jingping Zhao
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center on Mental Disorders, Changsha, China; National Technology Institute on Mental Disorders, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| |
Collapse
|
19
|
Lleó A, Núñez-Llaves R, Alcolea D, Chiva C, Balateu-Paños D, Colom-Cadena M, Gomez-Giro G, Muñoz L, Querol-Vilaseca M, Pegueroles J, Rami L, Lladó A, Molinuevo JL, Tainta M, Clarimón J, Spires-Jones T, Blesa R, Fortea J, Martínez-Lage P, Sánchez-Valle R, Sabidó E, Bayés À, Belbin O. Changes in Synaptic Proteins Precede Neurodegeneration Markers in Preclinical Alzheimer's Disease Cerebrospinal Fluid. Mol Cell Proteomics 2019; 18:546-560. [PMID: 30606734 PMCID: PMC6398205 DOI: 10.1074/mcp.ra118.001290] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Indexed: 01/08/2023] Open
Abstract
A biomarker of synapse loss, an early event in Alzheimer's disease (AD) pathophysiology that precedes neuronal death and symptom onset, would be a much-needed prognostic biomarker. With direct access to the brain interstitial fluid, the cerebrospinal fluid (CSF) is a potential source of synapse-derived proteins. In this study, we aimed to identify and validate novel CSF biomarkers of synapse loss in AD. Discovery: Combining shotgun proteomics of the CSF with an exhaustive search of the literature and public databases, we identified 251 synaptic proteins, from which we selected 22 for further study. Verification: Twelve proteins were discarded because of poor detection by Selected Reaction Monitoring (SRM). We confirmed the specific expression of 9 of the remaining proteins (Calsynytenin-1, GluR2, GluR4, Neurexin-2A, Neurexin-3A, Neuroligin-2, Syntaxin-1B, Thy-1, Vamp-2) at the human synapse using Array Tomography microscopy and biochemical fractionation methods. Exploration: Using SRM, we monitored these 9 synaptic proteins (20 peptides) in a cohort of CSF from cognitively normal controls and subjects in the pre-clinical and clinical AD stages (n = 80). Compared with controls, peptides from 8 proteins were elevated 1.3 to 1.6-fold (p < 0.04) in prodromal AD patients. Validation: Elevated levels of a GluR4 peptide at the prodromal stage were replicated (1.3-fold, p = 0.04) in an independent cohort (n = 60). Moreover, 7 proteins were reduced at preclinical stage 1 (0.6 to 0.8-fold, p < 0.04), a finding that was replicated (0.7 to 0.8-fold, p < 0.05) for 6 proteins in a third cohort (n = 38). In a cross-cohort meta-analysis, 6 synaptic proteins (Calsyntenin-1, GluR4, Neurexin-2A, Neurexin-3A, Syntaxin-1B and Thy-1) were reduced 0.8-fold (p < 0.05) in preclinical AD, changes that precede clinical symptoms and CSF markers of neurodegeneration. Therefore, these proteins could have clinical value for assessing disease progression, especially in preclinical stages of AD.
Collapse
Affiliation(s)
- Alberto Lleó
- From the ‡Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Raúl Núñez-Llaves
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- ¶Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025Barcelona, Spain
| | - Daniel Alcolea
- From the ‡Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Cristina Chiva
- ‖Proteomics Unit, Center for Genomics Regulation, Barcelona Institute of Science and Technology, 08003 Barcelona
- **University Pompeu Fabra, 08003 Barcelona
| | | | - Martí Colom-Cadena
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- ¶Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025Barcelona, Spain
| | - Gemma Gomez-Giro
- ¶Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025Barcelona, Spain
| | - Laia Muñoz
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- ¶Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025Barcelona, Spain
| | - Marta Querol-Vilaseca
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- ¶Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025Barcelona, Spain
| | - Jordi Pegueroles
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- ¶Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025Barcelona, Spain
| | - Lorena Rami
- ‡‡Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clínic-Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), 08015 Barcelona, Spain
| | - Albert Lladó
- ‡‡Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clínic-Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), 08015 Barcelona, Spain
| | - José L Molinuevo
- ‡‡Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clínic-Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), 08015 Barcelona, Spain
| | - Mikel Tainta
- §§Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, 20009 San Sebastian, Spain
- ¶¶Servicio de Neurologia, Organización Sanitaria Integrada Goierri-Alto Urola, Osakidetza, Zumárraga, España
| | - Jordi Clarimón
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- ¶Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025Barcelona, Spain
| | - Tara Spires-Jones
- ‖‖Centre for Discovery Brain Sciences and UK Dementia Research Institute, University of Edinburgh EH8 9JZ, UK
| | - Rafael Blesa
- From the ‡Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Juan Fortea
- From the ‡Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Pablo Martínez-Lage
- §§Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, 20009 San Sebastian, Spain
| | - Raquel Sánchez-Valle
- ‡‡Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clínic-Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), 08015 Barcelona, Spain
| | - Eduard Sabidó
- ‖Proteomics Unit, Center for Genomics Regulation, Barcelona Institute of Science and Technology, 08003 Barcelona
- **University Pompeu Fabra, 08003 Barcelona
| | - Àlex Bayés
- ***Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025, Barcelona, Spain
- ‡‡‡Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Olivia Belbin
- §Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain;
- ¶Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025Barcelona, Spain
| |
Collapse
|
20
|
Crider A, Nelson T, Davis T, Fagan K, Vaibhav K, Luo M, Kamalasanan S, Terry AV, Pillai A. Estrogen Receptor β Agonist Attenuates Endoplasmic Reticulum Stress-Induced Changes in Social Behavior and Brain Connectivity in Mice. Mol Neurobiol 2018; 55:7606-7618. [PMID: 29430617 PMCID: PMC6070416 DOI: 10.1007/s12035-018-0929-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/24/2018] [Indexed: 11/26/2022]
Abstract
Impaired social interaction is a key feature of several major psychiatric disorders including depression, autism, and schizophrenia. While, anatomically, the prefrontal cortex (PFC) is known as a key regulator of social behavior, little is known about the cellular mechanisms that underlie impairments of social interaction. One etiological mechanism implicated in the pathophysiology of the aforementioned psychiatric disorders is cellular stress and consequent adaptive responses in the endoplasmic reticulum (ER) that can result from a variety of environmental and physical factors. The ER is an organelle that serves essential roles in protein modification, folding, and maturation of proteins; however, the specific role of ER stress in altered social behavior is unknown. In this study, treatment with tunicamycin, an ER stress inducer, enhanced the phosphorylation level of inositol-requiring ER-to-nucleus signal kinase 1 (IRE1) and increased X-box-binding protein 1 (XBP1) mRNA splicing activity in the mouse PFC, whereas inhibition of IRE1/XBP1 pathway in PFC by a viral particle approach attenuated social behavioral deficits caused by tunicamycin treatment. Reduced estrogen receptor beta (ERβ) protein levels were found in the PFC of male mice following tunicamycin treatment. Pretreatment with an ERβ specific agonist, ERB-041 significantly attenuated tunicamycin-induced deficits in social behavior, and activation of IRE1/XBP1 pathway in mouse PFC. Moreover, ERB-041 inhibited tunicamycin-induced increases in functional connectivity between PFC and hippocampus in male mice. Together, these results show that ERβ agonist attenuates ER stress-induced deficits in social behavior through the IRE-1/XBP1 pathway.
Collapse
Affiliation(s)
- Amanda Crider
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, 997 St. Sebastian Way, Augusta, GA, 30912, USA
| | - Tyler Nelson
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, 997 St. Sebastian Way, Augusta, GA, 30912, USA
| | - Talisha Davis
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, 997 St. Sebastian Way, Augusta, GA, 30912, USA
| | - Kiley Fagan
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, 997 St. Sebastian Way, Augusta, GA, 30912, USA
| | - Kumar Vaibhav
- Department of Neurosurgery, and Department of Medical Laboratory Imaging and Radiologic Sciences (MLLIRS-CAHS), Augusta University, Augusta, GA, 30912, USA
| | - Matthew Luo
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, 997 St. Sebastian Way, Augusta, GA, 30912, USA
| | - Sunay Kamalasanan
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, 997 St. Sebastian Way, Augusta, GA, 30912, USA
| | - Alvin V Terry
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, 30912, USA
| | - Anilkumar Pillai
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, 997 St. Sebastian Way, Augusta, GA, 30912, USA.
| |
Collapse
|
21
|
Ryskalin L, Limanaqi F, Frati A, Busceti CL, Fornai F. mTOR-Related Brain Dysfunctions in Neuropsychiatric Disorders. Int J Mol Sci 2018; 19:ijms19082226. [PMID: 30061532 PMCID: PMC6121884 DOI: 10.3390/ijms19082226] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is an ubiquitously expressed serine-threonine kinase, which senses and integrates several intracellular and environmental cues to orchestrate major processes such as cell growth and metabolism. Altered mTOR signalling is associated with brain malformation and neurological disorders. Emerging evidence indicates that even subtle defects in the mTOR pathway may produce severe effects, which are evident as neurological and psychiatric disorders. On the other hand, administration of mTOR inhibitors may be beneficial for a variety of neuropsychiatric alterations encompassing neurodegeneration, brain tumors, brain ischemia, epilepsy, autism, mood disorders, drugs of abuse, and schizophrenia. mTOR has been widely implicated in synaptic plasticity and autophagy activation. This review addresses the role of mTOR-dependent autophagy dysfunction in a variety of neuropsychiatric disorders, to focus mainly on psychiatric syndromes including schizophrenia and drug addiction. For instance, amphetamines-induced addiction fairly overlaps with some neuropsychiatric disorders including neurodegeneration and schizophrenia. For this reason, in the present review, a special emphasis is placed on the role of mTOR on methamphetamine-induced brain alterations.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy.
| |
Collapse
|
22
|
Caraci F, Calabrese F, Molteni R, Bartova L, Dold M, Leggio GM, Fabbri C, Mendlewicz J, Racagni G, Kasper S, Riva MA, Drago F. International Union of Basic and Clinical Pharmacology CIV: The Neurobiology of Treatment-resistant Depression: From Antidepressant Classifications to Novel Pharmacological Targets. Pharmacol Rev 2018; 70:475-504. [PMID: 29884653 DOI: 10.1124/pr.117.014977] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Major depressive disorder is one of the most prevalent and life-threatening forms of mental illnesses and a major cause of morbidity worldwide. Currently available antidepressants are effective for most patients, although around 30% are considered treatment resistant (TRD), a condition that is associated with a significant impairment of cognitive function and poor quality of life. In this respect, the identification of the molecular mechanisms contributing to TRD represents an essential step for the design of novel and more efficacious drugs able to modify the clinical course of this disorder and increase remission rates in clinical practice. New insights into the neurobiology of TRD have shed light on the role of a number of different mechanisms, including the glutamatergic system, immune/inflammatory systems, neurotrophin function, and epigenetics. Advances in drug discovery processes in TRD have also influenced the classification of antidepressant drugs and novel classifications are available, such as the neuroscience-based nomenclature that can incorporate such advances in drug development for TRD. This review aims to provide an up-to-date description of key mechanisms in TRD and describe current therapeutic strategies for TRD before examining novel approaches that may ultimately address important neurobiological mechanisms not targeted by currently available antidepressants. All in all, we suggest that drug targeting different neurobiological systems should be able to restore normal function but must also promote resilience to reduce the long-term vulnerability to recurrent depressive episodes.
Collapse
Affiliation(s)
- F Caraci
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - F Calabrese
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - R Molteni
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - L Bartova
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - M Dold
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - G M Leggio
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - C Fabbri
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - J Mendlewicz
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - G Racagni
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - S Kasper
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - M A Riva
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - F Drago
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| |
Collapse
|
23
|
Rossetti AC, Paladini MS, Colombo M, Gruca P, Lason-Tyburkiewicz M, Tota-Glowczyk K, Papp M, Riva MA, Molteni R. Chronic Stress Exposure Reduces Parvalbumin Expression in the Rat Hippocampus through an Imbalance of Redox Mechanisms: Restorative Effect of the Antipsychotic Lurasidone. Int J Neuropsychopharmacol 2018; 21:883-893. [PMID: 29788232 PMCID: PMC6119300 DOI: 10.1093/ijnp/pyy046] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Psychiatric disorders are associated with altered function of inhibitory neurotransmission within the limbic system, which may be due to the vulnerability of selective neuronal subtypes to challenging environmental conditions, such as stress. In this context, parvalbumin-positive GABAergic interneurons, which are critically involved in processing complex cognitive tasks, are particularly vulnerable to stress exposure, an effect that may be the consequence of dysregulated redox mechanisms. METHODS Adult Male Wistar rats were subjected to the chronic mild stress procedure for 7 weeks. After 2 weeks, both control and stress groups were further divided into matched subgroups to receive chronic administration of vehicle or lurasidone (3 mg/kg/d) for the subsequent 5 weeks. Using real-time RT-PCR and western blot, we investigated the expression of GABAergic interneuron markers and the levels of key mediators of the oxidative balance in the dorsal and ventral hippocampus. RESULTS Chronic mild stress induced a specific decrease of parvalbumin expression in the dorsal hippocampus, an effect normalized by lurasidone treatment. Interestingly, the regulation of parvalbumin levels was correlated to the modulation of the antioxidant master regulator NRF2 and its chaperon protein KEAP1, which were also modulated by pharmacological intervention. CONCLUSIONS Our findings suggest that the susceptibility of parvalbumin neurons to stress may represent a key mechanism contributing to functional and structural impairments in specific brain regions relevant for psychiatric disorders. Moreover, we provide new insights on the mechanism of action of lurasidone, demonstrating that its chronic treatment normalizes chronic mild stress-induced parvalbumin alterations, possibly by potentiating antioxidant mechanisms, which may ameliorate specific functions that are deteriorated in psychiatric patients.
Collapse
Affiliation(s)
- Andrea C Rossetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Maria Serena Paladini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Martina Colombo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Piotr Gruca
- Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | | | | | - Mariusz Papp
- Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy,Correspondence: Marco A. Riva, PhD, Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9 - 20133 Milan, Italy ()
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
24
|
Marballi KK, Gallitano AL. Immediate Early Genes Anchor a Biological Pathway of Proteins Required for Memory Formation, Long-Term Depression and Risk for Schizophrenia. Front Behav Neurosci 2018; 12:23. [PMID: 29520222 PMCID: PMC5827560 DOI: 10.3389/fnbeh.2018.00023] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/29/2018] [Indexed: 01/02/2023] Open
Abstract
While the causes of myriad medical and infectious illnesses have been identified, the etiologies of neuropsychiatric illnesses remain elusive. This is due to two major obstacles. First, the risk for neuropsychiatric disorders, such as schizophrenia, is determined by both genetic and environmental factors. Second, numerous genes influence susceptibility for these illnesses. Genome-wide association studies have identified at least 108 genomic loci for schizophrenia, and more are expected to be published shortly. In addition, numerous biological processes contribute to the neuropathology underlying schizophrenia. These include immune dysfunction, synaptic and myelination deficits, vascular abnormalities, growth factor disruption, and N-methyl-D-aspartate receptor (NMDAR) hypofunction. However, the field of psychiatric genetics lacks a unifying model to explain how environment may interact with numerous genes to influence these various biological processes and cause schizophrenia. Here we describe a biological cascade of proteins that are activated in response to environmental stimuli such as stress, a schizophrenia risk factor. The central proteins in this pathway are critical mediators of memory formation and a particular form of hippocampal synaptic plasticity, long-term depression (LTD). Each of these proteins is also implicated in schizophrenia risk. In fact, the pathway includes four genes that map to the 108 loci associated with schizophrenia: GRIN2A, nuclear factor of activated T-cells (NFATc3), early growth response 1 (EGR1) and NGFI-A Binding Protein 2 (NAB2); each of which contains the "Index single nucleotide polymorphism (SNP)" (most SNP) at its respective locus. Environmental stimuli activate this biological pathway in neurons, resulting in induction of EGR immediate early genes: EGR1, EGR3 and NAB2. We hypothesize that dysfunction in any of the genes in this pathway disrupts the normal activation of Egrs in response to stress. This may result in insufficient electrophysiologic, immunologic, and neuroprotective, processes that these genes normally mediate. Continued adverse environmental experiences, over time, may thereby result in neuropathology that gives rise to the symptoms of schizophrenia. By combining multiple genes associated with schizophrenia susceptibility, in a functional cascade triggered by neuronal activity, the proposed biological pathway provides an explanation for both the polygenic and environmental influences that determine the complex etiology of this mental illness.
Collapse
Affiliation(s)
- Ketan K. Marballi
- Department of Basic Medical Sciences and Psychiatry, University of Arizona College of Medicine—Phoenix, Phoenix, AZ, United States
| | - Amelia L. Gallitano
- Department of Basic Medical Sciences and Psychiatry, University of Arizona College of Medicine—Phoenix, Phoenix, AZ, United States
| |
Collapse
|
25
|
Li Z, Chen J, Yu H, He L, Xu Y, Zhang D, Yi Q, Li C, Li X, Shen J, Song Z, Ji W, Wang M, Zhou J, Chen B, Liu Y, Wang J, Wang P, Yang P, Wang Q, Feng G, Liu B, Sun W, Li B, He G, Li W, Wan C, Xu Q, Li W, Wen Z, Liu K, Huang F, Ji J, Ripke S, Yue W, Sullivan PF, O'Donovan MC, Shi Y. Genome-wide association analysis identifies 30 new susceptibility loci for schizophrenia. Nat Genet 2017; 49:1576-1583. [PMID: 28991256 DOI: 10.1038/ng.3973] [Citation(s) in RCA: 311] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 09/19/2017] [Indexed: 02/06/2023]
Abstract
We conducted a genome-wide association study (GWAS) with replication in 36,180 Chinese individuals and performed further transancestry meta-analyses with data from the Psychiatry Genomics Consortium (PGC2). Approximately 95% of the genome-wide significant (GWS) index alleles (or their proxies) from the PGC2 study were overrepresented in Chinese schizophrenia cases, including ∼50% that achieved nominal significance and ∼75% that continued to be GWS in the transancestry analysis. The Chinese-only analysis identified seven GWS loci; three of these also were GWS in the transancestry analyses, which identified 109 GWS loci, thus yielding a total of 113 GWS loci (30 novel) in at least one of these analyses. We observed improvements in the fine-mapping resolution at many susceptibility loci. Our results provide several lines of evidence supporting candidate genes at many loci and highlight some pathways for further research. Together, our findings provide novel insight into the genetic architecture and biological etiology of schizophrenia.
Collapse
Affiliation(s)
- Zhiqiang Li
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Yu
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Institute of Mental Health, Sixth Hospital, Peking University, Beijing, China
- Department of Psychiatry, Jining Medical University, Jining, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yifeng Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dai Zhang
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Institute of Mental Health, Sixth Hospital, Peking University, Beijing, China
- Peking-Tsinghua Joint Center for Life Sciences/PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Qizhong Yi
- Department of Psychiatry, First Teaching Hospital of Xinjiang Medical University, Urumqi, China
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Disease and Metabolic Disease Institute of Qingdao University, Qingdao, China
| | - Xingwang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Jiawei Shen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijian Song
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Weidong Ji
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
- Changning Mental Health Center, Shanghai, China
| | - Meng Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Boyu Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yahui Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Jiqiang Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Wang
- Wuhu Fourth People's Hospital, Wuhu, China
| | - Ping Yang
- Wuhu Fourth People's Hospital, Wuhu, China
| | - Qingzhong Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Guoyin Feng
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Benxiu Liu
- Longquan Mountain Hospital of Guangxi Province, Liuzhou, China
| | - Wensheng Sun
- Longquan Mountain Hospital of Guangxi Province, Liuzhou, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Xu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjin Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Zujia Wen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Fang Huang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Jue Ji
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin, Berlin, Germany
| | - Weihua Yue
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Institute of Mental Health, Sixth Hospital, Peking University, Beijing, China
| | - Patrick F Sullivan
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Yongyong Shi
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Psychiatry, First Teaching Hospital of Xinjiang Medical University, Urumqi, China
- Changning Mental Health Center, Shanghai, China
| |
Collapse
|
26
|
Sun L, Min L, Zhou H, Li M, Shao F, Wang W. Adolescent social isolation affects schizophrenia-like behavior and astrocyte biomarkers in the PFC of adult rats. Behav Brain Res 2017; 333:258-266. [DOI: 10.1016/j.bbr.2017.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 07/05/2017] [Accepted: 07/09/2017] [Indexed: 10/19/2022]
|
27
|
Xiao W, Ye F, Liu C, Tang X, Li J, Dong H, Sha W, Zhang X. Cognitive impairment in first-episode drug-naïve patients with schizophrenia: Relationships with serum concentrations of brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor. Prog Neuropsychopharmacol Biol Psychiatry 2017; 76:163-168. [PMID: 28342945 DOI: 10.1016/j.pnpbp.2017.03.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/25/2017] [Accepted: 03/19/2017] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Evidence suggests that brain-derived neurotrophic factor (BDNF) and glial cell line -derived neurotrophic factor (GDNF) are important in the regulation of synaptic plasticity, which plays a key role in the cognitive processes in psychiatric disorders. Our work aimed at exploring the associations between serum BDNF and GDNF levels and cognitive functions in first-episode drug-naïve (FEDN) patients with schizophrenia. METHODS The BDNF and GDNF levels of 58 FEDN patients and 55 age- and sex-matched healthy controls were measured and test subjects were examined using several neurocognitive tests including the verbal fluency test (VFT), the trail making test (TMT), the digit span test (DST), and the Stroop test. RESULTS Patients performed significantly worse than controls in nearly all neurocognitive performances except the forward subscale part of the DST. BDNF levels were inversely correlated to TMT-part B scores and positively correlated to VFT-action in the FEDN group. GDNF levels showed a positive correlation with VFT-action scores and a negative correlation with TMT-part B scores of these patients. CONCLUSION Current data suggests that cognitive dysfunction widely exists in the early stages of schizophrenia. BDNF and GDNF may be jointly contributed to the pathological mechanisms involved in cognitive impairment in FEDN patients with schizophrenia.
Collapse
Affiliation(s)
- Wenhuan Xiao
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou 225003, PR China
| | - Fei Ye
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou 225003, PR China
| | - Chunlai Liu
- Department of Psychiatry, Affiliated Kangren Hospital, Ili Kazak Autonomous Prefecture of Xinjiang 835000, PR China
| | - Xiaowei Tang
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou 225003, PR China
| | - Jin Li
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou 225003, PR China
| | - Hui Dong
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou 225003, PR China
| | - Weiwei Sha
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou 225003, PR China
| | - Xiaobin Zhang
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou 225003, PR China.
| |
Collapse
|
28
|
Calabrese F, Brivio P, Gruca P, Lason-Tyburkiewicz M, Papp M, Riva MA. Chronic Mild Stress-Induced Alterations of Local Protein Synthesis: A Role for Cognitive Impairment. ACS Chem Neurosci 2017; 8:817-825. [PMID: 28094500 DOI: 10.1021/acschemneuro.6b00392] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Depression, a major cause of disability worldwide, is characterized by a complex and heterogeneous symptomatology. With this respect, cognitive deterioration represents a major problem that has a strong impact on a patient's function. Thus, within the context of a depressive phenotype, it is important to characterize the mechanisms that sustain cognitive dysfunctions and may represent an important target for pharmacological intervention. Here, using the chronic mild stress (CMS) paradigm of depression, we found that, independently from the anhedonic phenotype, CMS rats showed a deficit in the novel object recognition (NOR) test, which is associated with an inability to phosphorylate GluN2B subunit on Ser1303 and to activate the mTOR pathway. In agreement with the role of these systems in the control of local protein synthesis, we observed an increase phosphorylated eukaryotic elongation factor 2 (eEF2) in the crude synaptosomal fraction after the NOR test specifically in control animals. Since it has been demonstrated that peEF2 leads to the translation of specific mRNAs, we investigated if the gene-specific translational control depends on the presence of upstream open reading frame (uORF). Interestingly, we found a significant increase of oligophrenin-1 (2 uORFs) and of Bmal1 (7 uORFs) protein levels specifically in the control animals exposed to the NOR test. Our results demonstrated that the cognitive decline associated with stress exposure might be due to alterations in local protein translation of specific mRNAs, suggesting that a pharmacological intervention able to correct these defects might be useful in the improvement of deteriorated functions in patients with major depression and stress-related disorders.
Collapse
Affiliation(s)
- Francesca Calabrese
- Department
of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Paola Brivio
- Department
of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Piotr Gruca
- Institute
of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | | | - Mariusz Papp
- Institute
of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Marco A. Riva
- Department
of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| |
Collapse
|
29
|
Abstract
Depression is caused by a change in neural activity resulting from an increase in glutamate that drives excitatory neurons and may be responsible for the decline in the activity and number of the GABAergic inhibitory neurons. This imbalance between the excitatory and inhibitory neurons may contribute to the onset of depression. At the cellular level there is an increase in the concentration of intracellular Ca2+ within the inhibitory neurons that is driven by an increase in entry through the NMDA receptors (NMDARs) and through activation of the phosphoinositide signaling pathway that generates inositol trisphosphate (InsP3) that releases Ca2+ from the internal stores. The importance of these two pathways in driving the elevation of Ca2+ is supported by the fact that depression can be alleviated by ketamine that inhibits the NMDARs and scopolamine that inhibits the M1 receptors that drive InsP3/Ca2+ pathway. This increase in Ca2+ not only contributes to depression but it may also explain why individuals with depression have a strong likelihood of developing Alzheimer's disease. The enhanced levels of Ca2+ may stimulate the formation of Aβ to initiate the onset and progression of Alzheimer's disease. Just how vitamin D acts to reduce depression is unclear. The phenotypic stability hypothesis argues that vitamin D acts by reducing the increased neuronal levels of Ca2+ that are driving depression. This action of vitamin D depends on its function to maintain the expression of the Ca2+ pumps and buffers that reduce Ca2+ levels, which may explain how it acts to reduce the onset of depression.
Collapse
Affiliation(s)
- Michael J Berridge
- Emeritus Babraham Fellow, The Babraham Institute, Cambridge, United Kingdom
| |
Collapse
|
30
|
Emerging Synaptic Molecules as Candidates in the Etiology of Neurological Disorders. Neural Plast 2017; 2017:8081758. [PMID: 28331639 PMCID: PMC5346360 DOI: 10.1155/2017/8081758] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023] Open
Abstract
Synapses are complex structures that allow communication between neurons in the central nervous system. Studies conducted in vertebrate and invertebrate models have contributed to the knowledge of the function of synaptic proteins. The functional synapse requires numerous protein complexes with specialized functions that are regulated in space and time to allow synaptic plasticity. However, their interplay during neuronal development, learning, and memory is poorly understood. Accumulating evidence links synapse proteins to neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. In this review, we describe the way in which several proteins that participate in cell adhesion, scaffolding, exocytosis, and neurotransmitter reception from presynaptic and postsynaptic compartments, mainly from excitatory synapses, have been associated with several synaptopathies, and we relate their functions to the disease phenotype.
Collapse
|