1
|
Zhu K, Fu Y, Zhao Y, Niu B, Lu H. Perineuronal nets: Role in normal brain physiology and aging, and pathology of various diseases. Ageing Res Rev 2025; 108:102756. [PMID: 40254145 DOI: 10.1016/j.arr.2025.102756] [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: 01/23/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Perineuronal nets (PNNs) are a specialized extracellular matrix in the central nervous system. They are widely distributed in the brain, with distribution patterns varying by brain region. Their unique structure and composition allow them to play an important role in a range of physiological and pathological activities. In this article, we review the composition and structure of PNNs across different life stages, and provide a detailed analysis and comparison of the region-specific distribution patterns of PNNs in different brain areas. We also discuss the specific mechanisms by which PNNs are involved in plasticity, memory, and neuroprotection. Furthermore, we describe the abnormal changes in PNNs in aging and various brain diseases, such as Alzheimer's disease, Parkinson's disease, drug addiction, and schizophrenia. Finally, we review emerging and established therapeutic strategies targeting PNNs to modulate brain function and address neurological disorders from three perspectives: gene therapy, nanotechnology, and biomaterials. This review summarizes the physiological roles of PNNs at different stages of life and the mechanisms by which PNNs abnormalities contribute to various brain diseases, providing insights for potential therapeutic approaches.
Collapse
Affiliation(s)
- Kaiqi Zhu
- Shanghai University, School of Life Sciences, Shanghai, China
| | - Yifei Fu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Yinfei Zhao
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Niu
- Shanghai University, School of Life Sciences, Shanghai, China.
| | - Han Lu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Elvira UKA, Rivero O, Postiguillo A, García-Marti G, Escarti MJ, Aguilar EJ, David-Lluesma J, Molto MD, Perez-Rando M, Nacher J. Altered volume of thalamic nuclei and genetic expression in first-episode psychotic patients, and their association with childhood adversity. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111371. [PMID: 40250785 DOI: 10.1016/j.pnpbp.2025.111371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/21/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Childhood maltreatment is a significant risk factor for schizophrenia, and there are correlations between these adversities and thalamic gray matter density. The thalamus, a subcortical structure with various nuclei with specific connections, relays sensory information and participates in higher cognitive processes. Thalamic alterations are evident in psychotic disorders, and early-life adversities may affect its development, potentially contributing to psychosis. However, no evidence exists of volumetric alterations in thalamic nuclei in first-episode psychosis (FEP) patients related to early traumatic events. This study recruited 70 FEP patients and 68 age-matched healthy controls, who underwent 3 T structural MRI and clinical scales, including the Childhood Trauma Questionnaire (CTQ). The thalamus was analyzed for shape and segmented into nuclei to assess volume. Additionally, peripheral blood was analyzed for the expression of VCAN, CSGALNACT1, ST8SIA4, NRGN, SP4, and TOX genes, which are related to neuronal plasticity in the thalamus and psychosis. Results showed volumetric reductions in the whole thalamus and specific nuclei (lateral posterior, lateral geniculate, medial geniculate, ventrolateral, centromedian, anteroventral, mediodorsal, and pulvinar). The thalamus did not show shape alterations. A significant association was observed between physical neglect during childhood and the volume of the left thalamus and its anteroventral nucleus. Reduced expression of ST8SIA4 and SP4 genes was detected in FEP patients compared to healthy controls, with correlations between thalamic nuclei volumes and gene expression differing between groups. In conclusion, this study links thalamic nuclei volume with childhood adversities in FEP and highlights changes in ST8SIA4 and SP4 expression, correlating with thalamic nuclei volumes.
Collapse
Affiliation(s)
- Uriel K A Elvira
- Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Olga Rivero
- CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Biomedical Research Institute of the Clinic Hospital of Valencia (INCLIVA), Valencia, Spain; Department of Genetics. Faculty of Biological Sciences, Universitat de València, Spain
| | - Alba Postiguillo
- CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Biomedical Research Institute of the Clinic Hospital of Valencia (INCLIVA), Valencia, Spain
| | - Gracian García-Marti
- CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Quironsalud Hospital, Valencia, Spain
| | - Maria Jose Escarti
- CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Biomedical Research Institute of the Clinic Hospital of Valencia (INCLIVA), Valencia, Spain; Servicio de Psiquiatría, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Eduardo J Aguilar
- CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Biomedical Research Institute of the Clinic Hospital of Valencia (INCLIVA), Valencia, Spain; Servicio de Psiquiatría, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Javier David-Lluesma
- Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Biomedical Research Institute of the Clinic Hospital of Valencia (INCLIVA), Valencia, Spain; Department of Genetics. Faculty of Biological Sciences, Universitat de València, Spain
| | - Maria Dolores Molto
- CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Biomedical Research Institute of the Clinic Hospital of Valencia (INCLIVA), Valencia, Spain; Department of Genetics. Faculty of Biological Sciences, Universitat de València, Spain
| | - Marta Perez-Rando
- Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Biomedical Research Institute of the Clinic Hospital of Valencia (INCLIVA), Valencia, Spain.
| | - Juan Nacher
- Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; CIBERSAM, ISCIII Spanish National Network for Research in Mental Health, Madrid, Spain; Biomedical Research Institute of the Clinic Hospital of Valencia (INCLIVA), Valencia, Spain.
| |
Collapse
|
3
|
Han L, Liu Z, Jing Z, Liu Y, Peng Y, Chang H, Lei J, Wang K, Xu Y, Liu W, Wu Z, Li Q, Shi X, Zheng M, Wang H, Deng J, Zhong Y, Pan H, Lin J, Zhang R, Chen Y, Wu J, Xu M, Ren B, Cheng M, Yu Q, Song X, Lu Y, Tang Y, Yuan N, Sun S, An Y, Ding W, Sun X, Wei Y, Zhang S, Dou Y, Zhao Y, Han L, Zhu Q, Xu J, Wang S, Wang D, Bai Y, Liang Y, Liu Y, Chen M, Xie C, Bo B, Li M, Zhang X, Ting W, Chen Z, Fang J, Li S, Jiang Y, Tan X, Zuo G, Xie Y, Li H, Tao Q, Li Y, Liu J, Liu Y, Hao M, Wang J, Wen H, Liu J, Yan Y, Zhang H, Sheng Y, Yu S, Liao X, Jiang X, Wang G, Liu H, Wang C, Feng N, Liu X, Ma K, Xu X, Han T, Cao H, Zheng H, Chen Y, Lu H, Yu Z, Zhang J, Wang B, Wang Z, Xie Q, Pan S, Liu C, Xu C, Cui L, Li Y, Liu S, Liao S, Chen A, Wu QF, et alHan L, Liu Z, Jing Z, Liu Y, Peng Y, Chang H, Lei J, Wang K, Xu Y, Liu W, Wu Z, Li Q, Shi X, Zheng M, Wang H, Deng J, Zhong Y, Pan H, Lin J, Zhang R, Chen Y, Wu J, Xu M, Ren B, Cheng M, Yu Q, Song X, Lu Y, Tang Y, Yuan N, Sun S, An Y, Ding W, Sun X, Wei Y, Zhang S, Dou Y, Zhao Y, Han L, Zhu Q, Xu J, Wang S, Wang D, Bai Y, Liang Y, Liu Y, Chen M, Xie C, Bo B, Li M, Zhang X, Ting W, Chen Z, Fang J, Li S, Jiang Y, Tan X, Zuo G, Xie Y, Li H, Tao Q, Li Y, Liu J, Liu Y, Hao M, Wang J, Wen H, Liu J, Yan Y, Zhang H, Sheng Y, Yu S, Liao X, Jiang X, Wang G, Liu H, Wang C, Feng N, Liu X, Ma K, Xu X, Han T, Cao H, Zheng H, Chen Y, Lu H, Yu Z, Zhang J, Wang B, Wang Z, Xie Q, Pan S, Liu C, Xu C, Cui L, Li Y, Liu S, Liao S, Chen A, Wu QF, Wang J, Liu Z, Sun Y, Mulder J, Yang H, Wang X, Li C, Yao J, Xu X, Liu L, Shen Z, Wei W, Sun YG. Single-cell spatial transcriptomic atlas of the whole mouse brain. Neuron 2025:S0896-6273(25)00133-3. [PMID: 40132589 DOI: 10.1016/j.neuron.2025.02.015] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/24/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
A comprehensive atlas of genes, cell types, and their spatial distribution across a whole mammalian brain is fundamental for understanding the function of the brain. Here, using single-nucleus RNA sequencing (snRNA-seq) and Stereo-seq techniques, we generated a mouse brain atlas with spatial information for 308 cell clusters at single-cell resolution, involving over 4 million cells, as well as for 29,655 genes. We have identified cell clusters exhibiting preference for cortical subregions and explored their associations with brain-related diseases. Additionally, we pinpointed 155 genes with distinct regional expression patterns within the brainstem and unveiled 513 long non-coding RNAs showing region-enriched expression in the adult brain. Parcellation of brain regions based on spatial transcriptomic information revealed fine structure for several brain areas. Furthermore, we have uncovered 411 transcription factor regulons showing distinct spatiotemporal dynamics during neurodevelopment. Thus, we have constructed a single-cell-resolution spatial transcriptomic atlas of the mouse brain with genome-wide coverage.
Collapse
Affiliation(s)
- Lei Han
- BGI Research, Hangzhou 310030, China
| | - Zhen Liu
- Lingang Laboratory, Shanghai 200031, China; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zehua Jing
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuxuan Liu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | - Junjie Lei
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kexin Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanfang Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Liu
- Lingang Laboratory, Shanghai 200031, China
| | - Zihan Wu
- Tencent AI Lab, Shenzhen 518057, China
| | - Qian Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI Research, Shenzhen 518083, China
| | - Xiaoxue Shi
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mingyuan Zheng
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - He Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yanqing Zhong
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Junkai Lin
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ruiyi Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Chen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinhua Wu
- Lingang Laboratory, Shanghai 200031, China
| | - Mingrui Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Biyu Ren
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Qian Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinxiang Song
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanbing Lu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanchun Tang
- BGI Research, Hangzhou 310030, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Nini Yuan
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Suhong Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yingjie An
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenqun Ding
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xing Sun
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanrong Wei
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuzhen Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yannong Dou
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun Zhao
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Luyao Han
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Junfeng Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiwen Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dan Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yinqi Bai
- BGI Research, Hangzhou 310030, China
| | - Yikai Liang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengni Chen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chun Xie
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Binshi Bo
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mei Li
- BGI Research, Shenzhen 518083, China
| | - Xinyan Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wang Ting
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiao Fang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xing Tan
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guolong Zuo
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yue Xie
- BGI Research, Shenzhen 518083, China
| | - Huanhuan Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Quyuan Tao
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jianfeng Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuyang Liu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingkun Hao
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingjing Wang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huiying Wen
- BGI Research, Hangzhou 310030, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiabing Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Hui Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yifan Sheng
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shui Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xuyin Jiang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guangling Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Congcong Wang
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ning Feng
- BGI Research, Shenzhen 518083, China
| | - Xin Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xiangjie Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Huateng Cao
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Huiwen Zheng
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Haorong Lu
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Zixian Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Bo Wang
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | | | - Qing Xie
- BGI Research, Shenzhen 518083, China
| | | | - Chuanyu Liu
- BGI Research, Shenzhen 518083, China; Shenzhen Proof-of-Concept Center of Digital Cytopathology, BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Chan Xu
- BGI Research, Qingdao 266555, China
| | - Luman Cui
- BGI Research, Shenzhen 518083, China
| | - Yuxiang Li
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Sha Liao
- BGI Research, Shenzhen 518083, China; BGI Research, Chongqing 401329, China; JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing 401329, China
| | - Ao Chen
- BGI Research, Shenzhen 518083, China; BGI Research, Chongqing 401329, China; JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing 401329, China; Department of Biology, University of Copenhagen, Copenhagen 2200, Denmark
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jian Wang
- BGI Research, Shenzhen 518083, China; China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201602, China
| | - Yidi Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jan Mulder
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden; Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden
| | | | - Xiaofei Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | | | - Xun Xu
- BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China.
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China.
| | - Zhiming Shen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201602, China.
| | - Wu Wei
- Lingang Laboratory, Shanghai 200031, China; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yan-Gang Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
4
|
Mascio G, Nicoletti F, Battaglia G, Notartomaso S. A type-5 metabotropic glutamate receptor-perineuronal net axis shapes the function of cortical GABAergic interneurons in chronic pain. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2025; 5:10. [PMID: 39985105 PMCID: PMC11846390 DOI: 10.1186/s44158-025-00228-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
Parvalbumin-positive (PV+) interneurons (basket and chandelier cells) regulate the firing rate of pyramidal neurons in the cerebral cortex and play a key role in the generation of network oscillations in the cerebral cortex. A growing body of evidence suggest that cortical PV+ interneurons become overactive in chronic pain and contribute to nociceptive sensitization by inhibiting a top-down analgesic pathway. Here, we provide further support to this hypothesis showing that intracortical infusion of the GABAA receptor antagonist, bicuculline, caused analgesia in a mouse model of chronic inflammatory pain, although it reduced pain thresholds in healthy mice. We propose that mGlu5 metabotropic glutamate receptors and perineuronal nets (PNNs) shape the activity of PV+ interneurons in chronic pain, generating a form of maladaptive plasticity that enhances behavioural pain responses. mGlu5 receptors might be locally targeted by drugs activated by light delivered in cortical regions of the pain matrix, whereas the density of PNNs enwrapping PV+ interneurons might be reduced by local activation of PNN-degrading enzyme, such as type-9 matrix metalloproteinase. These strategies, which may require invasive treatments, might be beneficial in the management of severe pain which is refractory to conventional pharmacological and non-pharmacological interventions.
Collapse
Affiliation(s)
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Battaglia
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
5
|
Auer S, Schicht M, Hoffmann L, Budday S, Frischknecht R, Blümcke I, Paulsen F. The Role of Perineuronal Nets in Physiology and Disease: Insights from Recent Studies. Cells 2025; 14:321. [PMID: 40072050 PMCID: PMC11898492 DOI: 10.3390/cells14050321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/15/2025] Open
Abstract
Perineuronal nets (PNNs) are specialized extracellular matrix structures that predominantly surround inhibitory neurons in the central nervous system (CNS). They have been identified as crucial regulators of synaptic plasticity and neuronal excitability. This literature review aims to summarize the current state of knowledge about PNNs, their molecular composition and structure, as well as their functional roles and involvement in neurological diseases. Furthermore, future directions in PNN research are proposed, and the therapeutic potential of targeting PNNs to develop novel treatment options for various neurological disorders is explored. This review emphasizes the importance of PNNs in CNS physiology and pathology and underscores the need for further research in this area.
Collapse
Affiliation(s)
- Sophia Auer
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Martin Schicht
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Lucas Hoffmann
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Partner of the European Reference Network (ERN) EpiCARE, 91054 Erlangen, Germany; (L.H.); (I.B.)
| | - Silvia Budday
- Institute of Continuum Mechanics and Biomechanics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany;
| | - Renato Frischknecht
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany;
| | - Ingmar Blümcke
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Partner of the European Reference Network (ERN) EpiCARE, 91054 Erlangen, Germany; (L.H.); (I.B.)
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| |
Collapse
|
6
|
Li X, Li N, Zhao P, Ren D, Luo B, Zhou T. Perineuronal Nets: From Structure to Neurological Disorders. Curr Med Chem 2025; 32:1685-1701. [PMID: 37946343 DOI: 10.2174/0109298673258290231009111633] [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: 04/17/2023] [Revised: 08/05/2023] [Accepted: 09/01/2023] [Indexed: 11/12/2023]
Abstract
Perineuronal nets (PNN) is condensed extracellular matrix (ECM) in the central nervous system (CNS), which surrounds cell soma, axon initial segments, and synapses. In the brain, most neurons surrounded by PNN are interneurons, especially the parvalbumin-expressing interneurons (PVI). The formation of PNN is involved in the PVI maturation as well as the onset and closure of critical periods for developmental plasticity end. Dysfunction of PVI can lead to some neurological disorders, such as schizophrenia, bipolar depression, and Alzheimer's disease. Similarly, PNN assembling abnormalities are often observed in human patients and animal disease models. PNN is thought to have a neuroprotective effect and interact with signaling molecules to regulate synaptic plasticity and neuronal activity. In this review, we provide an overview of the composition, structure, and functions of PNN. In addition, we highlight abnormal changes in PNN components in pathological conditions. Understanding the roles of different components of PNN will bring us a new perspective on brain plasticity and neurological disorders.
Collapse
Affiliation(s)
- Xianghe Li
- Queen Mary School of Nanchang University, Nanchang 330031, China
| | - Nuojin Li
- Queen Mary School of Nanchang University, Nanchang 330031, China
| | - Pingping Zhao
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Dongyan Ren
- Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Bin Luo
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Tian Zhou
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| |
Collapse
|
7
|
Nguyen QH, Tran HN, Jeong Y. Regulation of neuronal fate specification and connectivity of the thalamic reticular nucleus by the Ascl1-Isl1 transcriptional cascade. Cell Mol Life Sci 2024; 81:478. [PMID: 39625482 PMCID: PMC11615174 DOI: 10.1007/s00018-024-05523-6] [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: 06/28/2024] [Revised: 10/21/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024]
Abstract
The thalamic reticular nucleus (TRN) is an anatomical and functional hub that modulates the flow of information between the cerebral cortex and thalamus, and its dysfunction has been linked to sensory disturbance and multiple behavioral disorders. Therefore, understanding how TRN neurons differentiate and establish connectivity is crucial to clarify the basics of TRN functions. Here, we showed that the regulatory cascade of the transcription factors Ascl1 and Isl1 promotes the fate of TRN neurons and concomitantly represses the fate of non-TRN prethalamic neurons. Furthermore, we found that this cascade is necessary for the correct development of the two main axonal connections, thalamo-cortical projections and prethalamo-thalamic projections. Notably, the disruption of prethalamo-thalamic axons can cause the pathfinding defects of thalamo-cortical axons in the thalamus. Finally, forced Isl1 expression can rescue disruption of cell fate specification and prethalamo-thalamic projections in in vitro primary cultures of Ascl1-deficient TRN neurons, indicating that Isl1 is an essential mediator of Ascl1 function in TRN development. Together, our findings provide insights into the molecular mechanisms for TRN neuron differentiation and circuit formation.
Collapse
Affiliation(s)
- Quy-Hoai Nguyen
- Department of Genetics and Biotechnology, College of Life Sciences, Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Hong-Nhung Tran
- Department of Genetics and Biotechnology, College of Life Sciences, Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Yongsu Jeong
- Department of Genetics and Biotechnology, College of Life Sciences, Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Republic of Korea.
| |
Collapse
|
8
|
Segobin S, Haast RAM, Kumar VJ, Lella A, Alkemade A, Bach Cuadra M, Barbeau EJ, Felician O, Pergola G, Pitel AL, Saranathan M, Tourdias T, Hornberger M. A roadmap towards standardized neuroimaging approaches for human thalamic nuclei. Nat Rev Neurosci 2024; 25:792-808. [PMID: 39420114 DOI: 10.1038/s41583-024-00867-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 10/19/2024]
Abstract
The thalamus has a key role in mediating cortical-subcortical interactions but is often neglected in neuroimaging studies, which mostly focus on changes in cortical structure and activity. One of the main reasons for the thalamus being overlooked is that the delineation of individual thalamic nuclei via neuroimaging remains controversial. Indeed, neuroimaging atlases vary substantially regarding which thalamic nuclei are included and how their delineations were established. Here, we review current and emerging methods for thalamic nuclei segmentation in neuroimaging data and consider the limitations of existing techniques in terms of their research and clinical applicability. We address these challenges by proposing a roadmap to improve thalamic nuclei segmentation in human neuroimaging and, in turn, harmonize research approaches and advance clinical applications. We believe that a collective effort is required to achieve this. We hope that this will ultimately lead to the thalamic nuclei being regarded as key brain regions in their own right and not (as often currently assumed) as simply a gateway between cortical and subcortical regions.
Collapse
Affiliation(s)
- Shailendra Segobin
- Normandie University, UNICAEN, PSL Université Paris, EPHE, INSERM, U1077, CHU de Caen, Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, Caen, France.
| | - Roy A M Haast
- Aix-Marseille University, CRMBM CNRS UMR 7339, Marseille, France
- APHM, La Timone Hospital, CEMEREM, Marseille, France
| | | | - Annalisa Lella
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Bari, Italy
| | - Anneke Alkemade
- Integrative Model-based Cognitive Neuroscience Unit, Department of Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Meritxell Bach Cuadra
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Radiology Department, Lausanne University and University Hospital, Lausanne, Switzerland
| | - Emmanuel J Barbeau
- Centre de recherche Cerveau et Cognition (Cerco), UMR5549, CNRS - Université de Toulouse, Toulouse, France
| | - Olivier Felician
- Aix Marseille Université, INSERM INS UMR 1106, APHM, Marseille, France
| | - Giulio Pergola
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Bari, Italy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anne-Lise Pitel
- Normandie University, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", NeuroPresage Team, Cyceron, Caen, France
| | | | - Thomas Tourdias
- Neuroimagerie diagnostique et thérapeutique, CHU de Bordeaux, Bordeaux, France
- Neurocentre Magendie, University of Bordeaux, INSERM U1215, Bordeaux, France
| | | |
Collapse
|
9
|
Klimczak P, Alcaide J, Gramuntell Y, Castillo-Gómez E, Varea E, Perez-Rando M, Nacher J. Long-term effects of a double hit murine model for schizophrenia on parvalbumin expressing cells and plasticity-related molecules in the thalamic reticular nucleus and the habenula. Transl Psychiatry 2024; 14:450. [PMID: 39448557 PMCID: PMC11502763 DOI: 10.1038/s41398-024-03166-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
The exposure to aversive experiences during early-life affects brain maturation and induces changes in behavior. Additionally, when these experiences coincide with subtle neurodevelopmental alterations, they may contribute to the emergence of psychiatric disorders, such as schizophrenia. Studies in patients and animal models have identified changes in parvalbumin (PV) expressing inhibitory neurons, highlighting their significance in the etiology of this disorder. Most studies have been focused on the cortex, but PV+ neurons also provide inhibitory input to diencephalic regions, particularly to the thalamus (through cells in the thalamic reticular nucleus, TRN) and the habenula. Remarkably, alterations in both nuclei have been described in schizophrenia. Some of these changes in PV+ cells may be mediated by perineuronal nets (PNN), specialized regions of the extracellular matrix that often surround them and regulate their synaptic input and activity. Interestingly, the physiological maturation and integration of PV+ neurons, which involves the assembly of PNN, occurs during early postnatal life. Plasticity molecules associated to inhibitory neurons, such as PSA-NCAM, or NMDA receptors (NMDAR) can also influence the structure and function of these cells. Growing evidence also indicates that glial cells regulate the physiology of PV+ neurons by influencing their maturation and modulating their synaptic connectivity. To explore the impact of early-life aversive experiences and concomitant subtle neurodevelopmental alterations on diencephalic PV+ cells, we analyzed adult male mice subjected to a double-hit model (DHM) of schizophrenia, combining a single injection of an NMDAR antagonist at P7 and post-weaning social isolation. We observed that exploratory behavior, PV+ neurons and their associated PNN, as well as PSA-NCAM and NMDAR expression and glial cells, in the TRN and the habenula were affected by the DHM or one of its factors. To our knowledge, this is the first report on such alterations in these diencephalic structures in an animal model combining neurodevelopmental alterations and early-life stress during adolescence. Our findings complement previous work on PV+ neurons in cortical regions and underscore the importance of studying diencephalic inhibitory networks and their intricate interactions with aversive experiences and neurodevelopmental alterations during early life in the context of schizophrenia.
Collapse
Affiliation(s)
- Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| | - Emilio Varea
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain.
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain.
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain.
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain.
| |
Collapse
|
10
|
Puzzo CD, Martinez-Garcia RI, Liu H, Dyson LF, Gilbert WO, Cruikshank SJ. Integration of distinct cortical inputs to primary and higher order inhibitory cells of the thalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.12.618039. [PMID: 39416152 PMCID: PMC11482941 DOI: 10.1101/2024.10.12.618039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The neocortex controls its own sensory input in part through top-down inhibitory mechanisms. Descending corticothalamic projections drive GABAergic neurons of the thalamic reticular nucleus (TRN), which govern thalamocortical cell activity via inhibition. Neurons in sensory TRN are organized into primary and higher order (HO) subpopulations, with separate intrathalamic connections and distinct genetic and functional properties. Here, we investigated top-down neocortical control over primary and HO neurons of somatosensory TRN. Projections from layer 6 of somatosensory cortex evoked stronger and more state-dependent activity in primary than in HO TRN, driven by more robust synaptic inputs and potent T-type calcium currents. However, HO TRN received additional, physiologically distinct, inputs from motor cortex and layer 5 of S1. Thus, in a departure from the canonical focused sensory layer 6 innervation characteristic of primary TRN, HO TRN integrates broadly from multiple corticothalamic systems, with unique state-dependence, extending the range of mechanisms for top-down control.
Collapse
|
11
|
Alcaide J, Gramuntell Y, Klimczak P, Bueno-Fernandez C, Garcia-Verellen E, Guicciardini C, Sandi C, Castillo-Gómez E, Crespo C, Perez-Rando M, Nacher J. Long term effects of peripubertal stress on the thalamic reticular nucleus of female and male mice. Neurobiol Dis 2024; 200:106642. [PMID: 39173845 DOI: 10.1016/j.nbd.2024.106642] [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: 07/13/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024] Open
Abstract
Adverse experiences during infancy and adolescence have an important and enduring effect on the brain and are predisposing factors for mental disorders, particularly major depression. This impact is particularly notable in regions with protracted development, such as the prefrontal cortex. The inhibitory neurons of this cortical region are altered by peripubertal stress (PPS), particularly in female mice. In this study we have explored whether the inhibitory circuits of the thalamus are impacted by PPS in male and female mice. This diencephalic structure, as the prefrontal cortex, also completes its development during postnatal life and is affected by adverse experiences. The long-term changes induced by PPS were exclusively found in adult female mice. We have found that PPS increases depressive-like behavior and induces changes in parvalbumin-expressing (PV+) cells of the thalamic reticular nucleus (TRN). We observed reductions in the volume of the TRN, together with those of parameters related to structures/molecules that regulate the plasticity and connectivity of PV+ cells: perineuronal nets, matricellular structures surrounding PV+ neurons, and the polysialylated form of the neural cell adhesion molecule (PSA-NCAM). The expression of the GluN1, but not of GluN2C, NMDA receptor subunit was augmented in the TRN after PPS. An increase in the fluorescence intensity of PV+ puncta was also observed in the synaptic output of TRN neurons in the lateral posterior thalamic nucleus. These results demonstrate that the inhibitory circuits of the thalamus, as those of the prefrontal cortex, are vulnerable to the effects of aversive experiences during early life, particularly in females. This vulnerability is probably related to the protracted development of the TRN and might contribute to the development of psychiatric disorders.
Collapse
Affiliation(s)
- Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Clara Bueno-Fernandez
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Erica Garcia-Verellen
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Chiara Guicciardini
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| | - Carlos Crespo
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain.
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain.
| |
Collapse
|
12
|
Laurent N, Bellamy EL, Hristova D, Houston A. Ketogenic diets in clinical psychology: examining the evidence and implications for practice. Front Psychol 2024; 15:1468894. [PMID: 39391844 PMCID: PMC11464436 DOI: 10.3389/fpsyg.2024.1468894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction The application of ketogenic dietary interventions to mental health treatments is increasingly acknowledged within medical and psychiatric fields, yet its exploration in clinical psychology remains limited. This article discusses the potential implications of ketogenic diets, traditionally utilized for neurological disorders, within broader mental health practices. Methods This article presents a perspective based on existing ketogenic diet research on historical use, biological mechanisms, and therapeutic benefits. It examines the potential application of these diets in mental health treatment and their relevance to clinical psychology research and practice. Results The review informs psychologists of the therapeutic benefits of ketogenic diets and introduces to the psychology literature the underlying biological mechanisms involved, such as modulation of neurotransmitters, reduction of inflammation, and stabilization of brain energy metabolism, demonstrating their potential relevance to biopsychosocial practice in clinical psychology. Conclusion By considering metabolic therapies, clinical psychologists can broaden their scope of biopsychosocial clinical psychology practice. This integration provides a care model that incorporates knowledge of the ketogenic diet as a treatment option in psychiatric care. The article emphasizes the need for further research and training for clinical psychologists to support the effective implementation of this metabolic psychiatry intervention.
Collapse
|
13
|
Lisboa JRF, Costa O, Pakes GH, Colodete DAE, Gomes FV. Perineuronal net density in schizophrenia: A systematic review of postmortem brain studies. Schizophr Res 2024; 271:100-109. [PMID: 39018984 DOI: 10.1016/j.schres.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 05/07/2024] [Accepted: 07/07/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND The onset of schizophrenia is concurrent with multiple key processes of brain development, such as the maturation of inhibitory networks. Some of these processes are proposed to depend on the development of perineuronal nets (PNNs), a specialized extracellular matrix structure that surrounds preferentially parvalbumin-containing GABAergic interneurons (PVIs). PNNs are fundamental to the postnatal experience-dependent maturation of inhibitory brain circuits. PNN abnormalities have been proposed as a core pathophysiological finding in SCZ, being linked to widespread consequences on circuit disruptions underlying SCZ symptoms. OBJECTIVE Here, we systematically evaluate PNN density in postmortem brain studies of subjects with SCZ. METHODS A systematic search in 3 online databases (PubMed, Embase, and Scopus) and qualitative review analysis of case-control studies reporting on PNN density in the postmortem brain of subjects with SCZ were performed. RESULTS Results consisted of 7 studies that were included in the final analysis. The specific brain regions investigated in the studies varied, with most attention given to the dorsolateral prefrontal cortex (DLPFC; 3 studies) and amygdala (2 studies). Findings were mostly positive for reduced PNN density in SCZ, with 6 of the 7 studies reporting significant reductions and one reporting a tendency towards reduced PNN density. Overall, tissue processing methodologies were heterogeneous. CONCLUSIONS Despite few studies, PNN density was consistently reduced in SCZ across different brain regions. These findings support evidence that implicates deficits in PNN density in the pathophysiology of SCZ. However, more studies, preferably using similar methodological approaches as well as replication of findings, are needed.
Collapse
Affiliation(s)
- João Roberto F Lisboa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| | - Olga Costa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gustavo Henrique Pakes
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Debora Akemi E Colodete
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
14
|
O’Brien JT, Jalilvand SP, Suji NA, Jupelly RK, Phensy A, Mwirigi JM, Elahi H, Price TJ, Kroener S. Elevations in the Mitochondrial Matrix Protein Cyclophilin D Correlate With Reduced Parvalbumin Expression in the Prefrontal Cortex of Patients With Schizophrenia. Schizophr Bull 2024; 50:1197-1207. [PMID: 38412332 PMCID: PMC11349014 DOI: 10.1093/schbul/sbae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
BACKGROUND AND HYPOTHESIS Cognitive deficits in schizophrenia are linked to dysfunctions of the dorsolateral prefrontal cortex (DLPFC), including alterations in parvalbumin (PV)-expressing interneurons (PVIs). Redox dysregulation and oxidative stress may represent convergence points in the pathology of schizophrenia, causing dysfunction of GABAergic interneurons and loss of PV. Here, we show that the mitochondrial matrix protein cyclophilin D (CypD), a critical initiator of the mitochondrial permeability transition pore (mPTP) and modulator of the intracellular redox state, is altered in PVIs in schizophrenia. STUDY DESIGN Western blotting was used to measure CypD protein levels in postmortem DLPFC specimens of schizophrenic patients (n = 27) and matched comparison subjects with no known history of psychiatric or neurological disorders (n = 26). In a subset of this cohort, multilabel immunofluorescent confocal microscopy with unbiased stereological sampling methods were used to quantify (1) numbers of PVI across the cortical mantle (20 unaffected comparison, 14 schizophrenia) and (2) PV and CypD protein levels from PVIs in the cortical layers 2-4 (23 unaffected comparison, 18 schizophrenia). STUDY RESULTS In schizophrenic patients, the overall number of PVIs in the DLPFC was not significantly altered, but in individual PVIs of layers 2-4 PV protein levels decreased along a superficial-to-deep gradient when compared to unaffected comparison subjects. These laminar-specific PVI alterations were reciprocally linked to significant CypD elevations both in PVIs and total DLPFC gray matter. CONCLUSIONS Our findings support previously reported PVI anomalies in schizophrenia and suggest that CypD-mediated mPTP formation could be a potential contributor to PVI dysfunction in schizophrenia.
Collapse
Affiliation(s)
- John T O’Brien
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Sophia P Jalilvand
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Neha A Suji
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Rohan K Jupelly
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Aarron Phensy
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Juliet M Mwirigi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Hajira Elahi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Sven Kroener
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
15
|
Nabit BP, Taylor A, Winder DG. Thalamocortical mGlu8 Modulates Dorsal Thalamus Excitatory Transmission and Sensorimotor Activity. J Neurosci 2024; 44:e0119242024. [PMID: 38918065 PMCID: PMC11293446 DOI: 10.1523/jneurosci.0119-24.2024] [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: 12/11/2023] [Revised: 02/25/2024] [Accepted: 04/04/2024] [Indexed: 06/27/2024] Open
Abstract
Metabotropic glutamate receptor 8 (mGlu8) is a heterogeneously expressed and poorly understood glutamate receptor with potential pharmacological significance. The thalamic reticular nucleus (TRN) is a critical inhibitory modulator of the thalamocortical-corticothalamic (TC-CT) network and plays a crucial role in information processing throughout the brain, is implicated in a variety of psychiatric conditions, and is also a site of significant mGlu8 expression. Using both male and female mice, we determined via fluorescent in situ hybridization that parvalbumin-expressing cells in the TRN core and shell matrices (identified by spp1+ and ecel1+ expression, respectively), as well as the cortical layers involved in CT signaling, express grm8 mRNA. We then assayed the physiological and behavioral impacts of perturbing grm8 signaling in the TC circuit through conditional (adeno-associated virus-CRE mediated) and cell-type-specific constitutive deletion strategies. We show that constitutive parvalbumin grm8 knock-out (PV grm8 knock-out) mice exhibited (1) increased spontaneous excitatory drive onto dorsal thalamus relay cells and (2) impaired sensorimotor gating, measured via paired-pulse inhibition, but observed no differences in locomotion and thigmotaxis in repeated bouts of open field test (OFT). Conversely, we observed hyperlocomotive phenotypes and anxiolytic effects of AAV-mediated conditional knockdown of grm8 in the TRN (TRN grm8 knockdown) in repeated OFT. Our findings underscore a role for mGlu8 in regulating excitatory neurotransmission as well as anxiety-related locomotor behavior and sensorimotor gating, revealing potential therapeutic applications for various neuropsychiatric disorders and guiding future research endeavors into mGlu8 signaling and TRN function.
Collapse
Affiliation(s)
- Bretton P Nabit
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37203
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37203
| | - Anne Taylor
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37203
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37203
| | - Danny G Winder
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37203
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37203
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37203
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37203
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01655
| |
Collapse
|
16
|
Pecheva D, Smith DM, Casey BJ, Woodward LJ, Dale AM, Filippi CG, Watts R. Sex and mental health are related to subcortical brain microstructure. Proc Natl Acad Sci U S A 2024; 121:e2403212121. [PMID: 39042688 PMCID: PMC11295051 DOI: 10.1073/pnas.2403212121] [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: 02/18/2024] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Some mental health problems such as depression and anxiety are more common in females, while others such as autism and attention deficit/hyperactivity (AD/H) are more common in males. However, the neurobiological origins of these sex differences are poorly understood. Animal studies have shown substantial sex differences in neuronal and glial cell structure, while human brain imaging studies have shown only small differences, which largely reflect overall body and brain size. Advanced diffusion MRI techniques can be used to examine intracellular, extracellular, and free water signal contributions and provide unique insights into microscopic cellular structure. However, the extent to which sex differences exist in these metrics of subcortical gray matter structures implicated in psychiatric disorders is not known. Here, we show large sex-related differences in microstructure in subcortical regions, including the hippocampus, thalamus, and nucleus accumbens in a large sample of young adults. Unlike conventional T1-weighted structural imaging, large sex differences remained after adjustment for age and brain volume. Further, diffusion metrics in the thalamus and amygdala were associated with depression, anxiety, AD/H, and antisocial personality problems. Diffusion MRI may provide mechanistic insights into the origin of sex differences in behavior and mental health over the life course and help to bridge the gap between findings from experimental, epidemiological, and clinical mental health research.
Collapse
Affiliation(s)
- Diliana Pecheva
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA92093
| | - Diana M. Smith
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA92093
- Medical Scientist Training Program, University of California, San Diego, La Jolla, CA92093
| | - B. J. Casey
- Department of Neuroscience and Behavior, Barnard College, New York, NY10027
| | - Lianne J. Woodward
- Faculty of Health, University of Canterbury, Christchurch8140, New Zealand
| | - Anders M. Dale
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA92093
- Department of Radiology, University of California, San Diego, La Jolla, CA92093
- Department of Neurosciences, University of California, San Diego, La Jolla, CA92093
- Department of Psychiatry, University of California, San Diego, La Jolla, CA92093
| | - Christopher G. Filippi
- Department of Radiology, The Hospital for Sick Children and the SickKids Research Institute, Toronto, ON M5G 1E8, Canada
| | - Richard Watts
- Faculty of Health, University of Canterbury, Christchurch8140, New Zealand
| |
Collapse
|
17
|
Iwakura Y, Kobayashi Y, Namba H, Nawa H, Takei N. Epidermal Growth Factor Suppresses the Development of GABAergic Neurons Via the Modulation of Perineuronal Net Formation in the Neocortex of Developing Rodent Brains. Neurochem Res 2024; 49:1347-1358. [PMID: 38353896 DOI: 10.1007/s11064-024-04122-y] [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: 10/31/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 04/04/2024]
Abstract
Previously, we reported that epidermal growth factor (EGF) suppresses GABAergic neuronal development in the rodent cortex. Parvalbumin-positive GABAergic neurons (PV neurons) have a unique extracellular structure, perineuronal nets (PNNs). PNNs are formed during the development of PV neurons and are mainly formed from chondroitin sulfate (CS) proteoglycans (CSPGs). We examined the effect of EGF on CSPG production and PNN formation as a potential molecular mechanism for the inhibition of inhibiting GABAergic neuronal development by EGF. In EGF-overexpressing transgenic (EGF-Tg) mice, the number of PNN-positive PV neurons was decreased in the cortex compared with that in wild-type mice, as in our previous report. The amount of CS and neurocan was also lower in the cortex of EGF-Tg mice, with a similar decrease observed in EGF-treated cultured cortical neurons. PD153035, an EGF receptor (ErbB1) kinase inhibitor, prevented those mentioned above excess EGF-induced reduction in PNN. We explored the molecular mechanism underlying the effect of EGF on PNNs using fluorescent substrates for matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinases (ADAMs). EGF increased the enzyme activity of MMPs and ADAMs in cultured neurons. These enzyme activities were also increased in the EGF-Tg mice cortex. GM6001, a broad inhibitor of MMPs and ADAMs, also blocked EGF-induced PNN reductions. Therefore, EGF/EGF receptor signals may regulate PNN formation in the developing cortex.
Collapse
Affiliation(s)
- Yuriko Iwakura
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan.
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan.
| | - Yutaro Kobayashi
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Biochemistry, Graduate School of Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hisaaki Namba
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, 640-8156, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, 640-8156, Japan
| | - Nobuyuki Takei
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
| |
Collapse
|
18
|
Palmisano A, Pandit S, Smeralda CL, Demchenko I, Rossi S, Battelli L, Rivolta D, Bhat V, Santarnecchi E. The Pathophysiological Underpinnings of Gamma-Band Alterations in Psychiatric Disorders. Life (Basel) 2024; 14:578. [PMID: 38792599 PMCID: PMC11122172 DOI: 10.3390/life14050578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 05/26/2024] Open
Abstract
Investigating the biophysiological substrates of psychiatric illnesses is of great interest to our understanding of disorders' etiology, the identification of reliable biomarkers, and potential new therapeutic avenues. Schizophrenia represents a consolidated model of γ alterations arising from the aberrant activity of parvalbumin-positive GABAergic interneurons, whose dysfunction is associated with perineuronal net impairment and neuroinflammation. This model of pathogenesis is supported by molecular, cellular, and functional evidence. Proof for alterations of γ oscillations and their underlying mechanisms has also been reported in bipolar disorder and represents an emerging topic for major depressive disorder. Although evidence from animal models needs to be further elucidated in humans, the pathophysiology of γ-band alteration represents a common denominator for different neuropsychiatric disorders. The purpose of this narrative review is to outline a framework of converging results in psychiatric conditions characterized by γ abnormality, from neurochemical dysfunction to alterations in brain rhythms.
Collapse
Affiliation(s)
- Annalisa Palmisano
- Chair of Lifespan Developmental Neuroscience, Faculty of Psychology, TUD Dresden University of Technology, 01069 Dresden, Germany
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Department of Education, Psychology, and Communication, University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Siddhartha Pandit
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
| | - Carmelo L. Smeralda
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Siena Brain Investigation and Neuromodulation (SI-BIN) Laboratory, Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, 53100 Siena, Italy;
| | - Ilya Demchenko
- Interventional Psychiatry Program, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (I.D.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Simone Rossi
- Siena Brain Investigation and Neuromodulation (SI-BIN) Laboratory, Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, 53100 Siena, Italy;
| | - Lorella Battelli
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Center for Neuroscience and Cognitive Systems@UniTn, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Davide Rivolta
- Department of Education, Psychology, and Communication, University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Venkat Bhat
- Interventional Psychiatry Program, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (I.D.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Emiliano Santarnecchi
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Department of Neurology and Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
19
|
Paveliev M, Egorchev AA, Musin F, Lipachev N, Melnikova A, Gimadutdinov RM, Kashipov AR, Molotkov D, Chickrin DE, Aganov AV. Perineuronal Net Microscopy: From Brain Pathology to Artificial Intelligence. Int J Mol Sci 2024; 25:4227. [PMID: 38673819 PMCID: PMC11049984 DOI: 10.3390/ijms25084227] [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: 02/02/2024] [Revised: 03/31/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Perineuronal nets (PNN) are a special highly structured type of extracellular matrix encapsulating synapses on large populations of CNS neurons. PNN undergo structural changes in schizophrenia, epilepsy, Alzheimer's disease, stroke, post-traumatic conditions, and some other brain disorders. The functional role of the PNN microstructure in brain pathologies has remained largely unstudied until recently. Here, we review recent research implicating PNN microstructural changes in schizophrenia and other disorders. We further concentrate on high-resolution studies of the PNN mesh units surrounding synaptic boutons to elucidate fine structural details behind the mutual functional regulation between the ECM and the synaptic terminal. We also review some updates regarding PNN as a potential pharmacological target. Artificial intelligence (AI)-based methods are now arriving as a new tool that may have the potential to grasp the brain's complexity through a wide range of organization levels-from synaptic molecular events to large scale tissue rearrangements and the whole-brain connectome function. This scope matches exactly the complex role of PNN in brain physiology and pathology processes, and the first AI-assisted PNN microscopy studies have been reported. To that end, we report here on a machine learning-assisted tool for PNN mesh contour tracing.
Collapse
Affiliation(s)
- Mikhail Paveliev
- Neuroscience Center, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Anton A. Egorchev
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Foat Musin
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Nikita Lipachev
- Institute of Physics, Kazan Federal University, Kremlyovskaya 16a, Kazan 420008, Tatarstan, Russia; (N.L.); (A.V.A.)
| | - Anastasiia Melnikova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, Kazan 420015, Tatarstan, Russia;
| | - Rustem M. Gimadutdinov
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Aidar R. Kashipov
- Institute of Artificial Intelligence, Robotics and Systems Engineering, Kazan Federal University, Kremlyovskaya 18, Kazan 420008, Tatarstan, Russia; (A.R.K.); (D.E.C.)
| | - Dmitry Molotkov
- Biomedicum Imaging Unit, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland;
| | - Dmitry E. Chickrin
- Institute of Artificial Intelligence, Robotics and Systems Engineering, Kazan Federal University, Kremlyovskaya 18, Kazan 420008, Tatarstan, Russia; (A.R.K.); (D.E.C.)
| | - Albert V. Aganov
- Institute of Physics, Kazan Federal University, Kremlyovskaya 16a, Kazan 420008, Tatarstan, Russia; (N.L.); (A.V.A.)
| |
Collapse
|
20
|
Liang HB, He WY, Liu YP, Wang HB. Pain Comorbidities with Attention Deficit: A Narrative Review of Clinical and Preclinical Research. J Pain Res 2024; 17:1055-1065. [PMID: 38505503 PMCID: PMC10948333 DOI: 10.2147/jpr.s443915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 02/23/2024] [Indexed: 03/21/2024] Open
Abstract
A negative correlation exists between attention and pain. The cognitive impairments linked to pain can significantly impede a patient's healing process and everyday tasks, particularly for individuals experiencing persistent pain. Furthermore, it has been demonstrated that diversion can effectively decrease pain levels in individuals. The focus of this review is to analyze clinical trials and fundamental investigations regarding alterations in focus and persistent discomfort. Moreover, we investigated the common neuroanatomy associated with attention and pain. Furthermore, we examined the impact of various neuromodulators on the transmission of pain and processes related to attention, while also considering the potential neural mechanisms that contribute to the co-occurrence of pain and attention deficits. Further investigation in this field will enhance our comprehension of patient symptoms and the underlying pathophysiology, ultimately resulting in more objective approaches to treatment.
Collapse
Affiliation(s)
- Hong-Bin Liang
- Graduate School of Guangdong Medical University, Zhanjiang, Guangdong Province, People’s Republic of China
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, Guangdong Province, People’s Republic of China
| | - Wan-You He
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, Guangdong Province, People’s Republic of China
| | - Yan-Ping Liu
- College of Nursing, Shandong First Medical University (Shandong Academy of Medical Science), Jinan, Shandong Province, People’s Republic of China
| | - Han-Bing Wang
- Graduate School of Guangdong Medical University, Zhanjiang, Guangdong Province, People’s Republic of China
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, Guangdong Province, People’s Republic of China
| |
Collapse
|
21
|
Valeri J, Stiplosek C, O'Donovan SM, Sinclair D, Grant KA, Bollavarapu R, Platt DM, Stockmeier CA, Gisabella B, Pantazopoulos H. Extracellular matrix abnormalities in the hippocampus of subjects with substance use disorder. Transl Psychiatry 2024; 14:115. [PMID: 38402197 PMCID: PMC10894211 DOI: 10.1038/s41398-024-02833-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/26/2024] Open
Abstract
Contextual triggers are significant factors contributing to relapse in substance use disorders (SUD). Emerging evidence points to a critical role of extracellular matrix (ECM) molecules as mediators of reward memories. Chondroitin sulfate proteoglycans (CSPGs) are a subset of ECM molecules that form perineuronal nets (PNN) around inhibitory neurons. PNNs restrict synaptic connections and help maintain synapses. Rodent models suggest that modulation of PNNs may strengthen contextual reward memories in SUD. However, there is currently a lack of information regarding PNNs in the hippocampus of people with SUD as well as how comorbidity with major depressive disorder (MDD) may affect PNNs. We used postmortem hippocampal tissues from cohorts of human and nonhuman primates with or without chronic alcohol use to test the hypothesis that PNNs are increased in subjects with SUD. We used histochemical labeling and quantitative microscopy to examine PNNs, and qRT-PCR to examine gene expression for ECM molecules, synaptic markers and related markers. We identified increased densities of PNNs and CSPG-labeled glial cells in SUD, coinciding with decreased expression of the ECM protease matrix metalloproteinase 9 (Mmp9), and increased expression for the excitatory synaptic marker vesicle associated membrane protein 2 (Vamp2). Similar increases in PNNs were observed in monkeys with chronic alcohol self-administration. Subjects with MDD displayed changes opposite to SUD, and subjects with SUD and comorbid MDD had minimal changes in any of the outcome measures examined. Our findings demonstrate that PNNs are increased in SUD, possibly contributing to stabilizing contextual reward memories as suggested by preclinical studies. Our results also point to a previously unsuspected role for CSPG expression in glial cells in SUD. Evidence for increased hippocampal PNNs in SUD suggests that targeting PNNs to weaken contextual reward memories is a promising therapeutic approach for SUD, however comorbidity with MDD is a significant consideration.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charlotte Stiplosek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - David Sinclair
- Department of Neuroscience, University of Toledo, Toledo, OH, USA
| | | | - Ratna Bollavarapu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Donna M Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
22
|
Visocky V, Morris BJ, Dunlop J, Brandon N, Sakata S, Pratt JA. Site-specific inhibition of the thalamic reticular nucleus induces distinct modulations in sleep architecture. Eur J Neurosci 2024; 59:554-569. [PMID: 36623837 DOI: 10.1111/ejn.15908] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023]
Abstract
The thalamic reticular nucleus (TRN) is crucial for the modulation of sleep-related oscillations. The caudal and rostral subpopulations of the TRN exert diverse activities, which arise from their interconnectivity with all thalamic nuclei, as well as other brain regions. Despite the recent characterization of the functional and genetic heterogeneity of the TRN, the implications of this heterogeneity for sleep regulation have not been assessed. Here, using a combination of optogenetics and electrophysiology in C57BL/6 mice, we demonstrate that caudal and rostral TRN modulations are associated with changes in cortical alpha and delta oscillations and have distinct effects on sleep stability. Tonic silencing of the rostral TRN elongates sleep episodes, while tonic silencing of the caudal TRN fragments sleep. Overall, we show evidence of distinct roles exerted by the rostral and caudal TRN in sleep regulation and oscillatory activity.
Collapse
Affiliation(s)
- Vladimir Visocky
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Brian J Morris
- College of Medical, Veterinary and Life Sciences, Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | | | | | - Shuzo Sakata
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
23
|
Li J, Li Z, Wang X, Liu Y, Wang S, Wang X, Li Y, Qin L. The Thalamocortical Mechanism Underlying the Generation and Regulation of the Auditory Steady-State Responses in Awake Mice. J Neurosci 2024; 44:e1166232023. [PMID: 37945348 PMCID: PMC10851679 DOI: 10.1523/jneurosci.1166-23.2023] [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: 06/24/2023] [Revised: 09/28/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
The auditory steady-state response (ASSR) is a cortical oscillation induced by trains of 40 Hz acoustic stimuli. While the ASSR has been widely used in clinic measurement, the underlying neural mechanism remains poorly understood. In this study, we investigated the contribution of different stages of auditory thalamocortical pathway-medial geniculate body (MGB), thalamic reticular nucleus (TRN), and auditory cortex (AC)-to the generation and regulation of 40 Hz ASSR in C57BL/6 mice of both sexes. We found that the neural response synchronizing to 40 Hz sound stimuli was most prominent in the GABAergic neurons in the granular layer of AC and the ventral division of MGB (MGBv), which were regulated by optogenetic manipulation of TRN neurons. Behavioral experiments confirmed that disrupting TRN activity has a detrimental effect on the ability of mice to discriminate 40 Hz sounds. These findings revealed a thalamocortical mechanism helpful to interpret the results of clinical ASSR examinations.Significance Statement Our study contributes to clarifying the thalamocortical mechanisms underlying the generation and regulation of the auditory steady-state response (ASSR), which is commonly used in both clinical and neuroscience research to assess the integrity of auditory function. Combining a series of electrophysiological and optogenetic experiments, we demonstrate that the generation of cortical ASSR is dependent on the lemniscal thalamocortical projections originating from the ventral division of medial geniculate body to the GABAergic interneurons in the granule layer of the auditory cortex. Furthermore, the thalamocortical process for ASSR is strictly regulated by the activity of thalamic reticular nucleus (TRN) neurons. Behavioral experiments confirmed that dysfunction of TRN would cause a disruption of mice's behavioral performance in the auditory discrimination task.
Collapse
Affiliation(s)
- Jinhong Li
- Department of Physiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Zijie Li
- Department of Physiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Xueru Wang
- Department of Physiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Yunhan Liu
- Department of Physiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Shuai Wang
- Department of Physiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Xuejiao Wang
- Department of Physiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Yingna Li
- Department of Physiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Ling Qin
- Department of Physiology, China Medical University, Shenyang 110122, People's Republic of China
| |
Collapse
|
24
|
Valeri J, Stiplosek C, O’Donovan SM, Sinclair D, Grant K, Bollavarapu R, Platt DM, Stockmeier CA, Gisabella B, Pantazopoulos H. Extracellular Matrix Abnormalities in the Hippocampus of Subjects with Substance Use Disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.09.07.23295222. [PMID: 37732207 PMCID: PMC10508799 DOI: 10.1101/2023.09.07.23295222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Contextual triggers are significant factors contributing to relapse in substance use disorders (SUD). Emerging evidence points to a critical role of extracellular matrix (ECM) molecules as mediators of reward memories. Chondroitin sulfate proteoglycans (CSPGs) are a subset of ECM molecules that form perineuronal nets (PNN) around inhibitory neurons. PNNs restrict synaptic connections and help maintain synapses. Rodent models suggest that modulation of PNNs may strengthen contextual reward memories in SUD. However, there is currently a lack of information regarding PNNs in the hippocampus of people with SUD as well as how comorbidity with major depressive disorder (MDD) may affect PNNs. We used postmortem hippocampal tissues from cohorts of human and nonhuman primates with or without chronic alcohol use to test the hypothesis that PNNs are increased in subjects with SUD. We used histochemical labeling and quantitative microscopy to examine PNNs, and qRT-PCR to examine gene expression for ECM molecules, synaptic markers and related markers. We identified increased densities of PNNs and CSPG-labeled glial cells in SUD, coinciding with decreased expression of the ECM protease matrix metalloproteinase 9 (Mmp9), and increased expression for the excitatory synaptic marker vesicle associated membrane protein 2 (Vamp2). Similar increases in PNNs were observed in monkeys with chronic alcohol self-administration. Subjects with MDD displayed changes opposite to SUD, and subjects with SUD and comorbid MDD had minimal changes in any of the outcome measures examined. Our findings demonstrate that PNNs are increased in SUD, possibly contributing to stabilizing contextual reward memories as suggested by preclinical studies. Our results also point to a previously unsuspected role for CSPG expression in glial cells in SUD. Evidence for increased hippocampal PNNs in SUD suggests that targeting PNNs to weaken contextual reward memories is a promising therapeutic approach for SUD, however comorbidity with MDD is a significant consideration.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Charlotte Stiplosek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
| | | | - David Sinclair
- Department of Neuroscience, University of Toledo, Toledo, OH
| | | | - Ratna Bollavarapu
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Donna M. Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Craig A. Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
25
|
Villacres JE, Riveira N, Kim S, Colgin LL, Noebels JL, Lopez AY. Abnormal patterns of sleep and waking behaviors are accompanied by neocortical oscillation disturbances in an Ank3 mouse model of epilepsy-bipolar disorder comorbidity. Transl Psychiatry 2023; 13:403. [PMID: 38123552 PMCID: PMC10733341 DOI: 10.1038/s41398-023-02700-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
ANK3 is a leading bipolar disorder (BD) candidate gene in humans and provides a unique opportunity for studying epilepsy-BD comorbidity. Previous studies showed that deletion of Ank3-1b, a BD-associated variant of Ank3 in mice leads to increased firing threshold and diminished action potential dynamic range of parvalbumin (PV) interneurons and absence epilepsy, thus providing a biological mechanism linking epilepsy and BD. To explore the behavioral overlap of these disorders, we characterized behavioral patterns of Ank3-1b KO mice during overnight home-cage activity and examined network activity during these behaviors using paired video and EEG recordings. Since PV interneurons contribute to the generation of high-frequency gamma oscillations, we anticipated changes in the power of neocortical EEG signals in the gamma frequency range (> 25 Hz) during behavioral states related to human BD symptoms, including abnormal sleep, hyperactivity, and repetitive behaviors. Ank3-1b KO mice exhibited an overall increase in slow gamma (~25-45 Hz) power compared to controls, and slow gamma power correlated with seizure phenotype severity across behaviors. During sleep, increased slow gamma power correlated with decreased time spent in the rapid eye movement (REM) stage of sleep. Seizures were more common during REM sleep compared to non-REM (NREM) sleep. We also found that Ank3-1b KO mice were hyperactive and exhibited a repetitive behavior phenotype that co-occurred with increased slow gamma power. Our results identify a novel EEG biomarker associating Ank3 genetic variation with BD and epilepsy and suggest modulation of gamma oscillations as a potential therapeutic target.
Collapse
Affiliation(s)
- Juan E Villacres
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX, 78712-0805, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712-0805, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712-0805, USA
| | - Nicholas Riveira
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX, 78712-0805, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712-0805, USA
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX, 78712-0805, USA
| | - Sohmee Kim
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX, 78712-0805, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712-0805, USA
| | - Laura L Colgin
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX, 78712-0805, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712-0805, USA
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX, 78712-0805, USA
| | - Jeffrey L Noebels
- Department of Neurology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Angel Y Lopez
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX, 78712-0805, USA.
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712-0805, USA.
| |
Collapse
|
26
|
Dong Y, Zhao K, Qin X, Du G, Gao L. The mechanisms of perineuronal net abnormalities in contributing aging and neurological diseases. Ageing Res Rev 2023; 92:102092. [PMID: 37839757 DOI: 10.1016/j.arr.2023.102092] [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: 08/11/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
The perineuronal net (PNN) is a highly latticed extracellular matrix in the central nervous system, which is composed of hyaluronic acid, proteoglycan, hyaluronan and proteoglycan link protein (Hapln), and tenascin. PNN is predominantly distributed in GABAergic interneurons expressing Parvalbumin (PV) and plays a critical role in synaptic function, learning and memory, oxidative stress, and inflammation. In addition, PNN's structure and function are also modulated by a variety of factors, including protein tyrosine phosphatase σ (PTPσ), orthodenticle homeo-box 2 (Otx2), and erb-b2 receptor tyrosine kinase 4 (ErbB4). Glycosaminoglycan (GAG), a component of proteoglycan, also influences PNN through its sulfate mode. PNN undergoes abnormal changes during aging and in various neurological diseases, such as Alzheimer's disease, Parkinson's disease, schizophrenia, autism spectrum disorder, and multiple sclerosis. Nevertheless, there is limited report on the relationship between PNN and aging or age-related neurological diseases. This review elaborates on the mechanisms governing PNN regulation and summarizes how PNN abnormalities contribute to aging and neurological diseases, offering insights for potential treatments.
Collapse
Affiliation(s)
- Yixiao Dong
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Kunkun Zhao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Guanhua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Li Gao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China.
| |
Collapse
|
27
|
Zhang B, Li L, Tang X, Zeng J, Song Y, Hou Z, Ma T, Afewerky HK, Li H, Lu Y, He A, Li X. Distribution Patterns of Subgroups of Inhibitory Neurons Divided by Calbindin 1. Mol Neurobiol 2023; 60:7285-7296. [PMID: 37548854 DOI: 10.1007/s12035-023-03542-1] [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: 06/14/2023] [Accepted: 07/25/2023] [Indexed: 08/08/2023]
Abstract
The inhibitory neurons in the brain play an essential role in neural network firing patterns by releasing γ-aminobutyric acid (GABA) as the neurotransmitter. In the mouse brain, based on the protein molecular markers, inhibitory neurons are usually to be divided into three non-overlapping groups: parvalbumin (PV), neuropeptide somatostatin (SST), and vasoactive intestinal peptide (VIP)-expressing neurons. Each neuronal group exhibited unique properties in molecule, electrophysiology, circuitry, and function. Calbindin 1 (Calb1), a ubiquitous calcium-binding protein, often acts as a "divider" in excitatory neuronal classification. Based on Calb1 expression, the excitatory neurons from the same brain region can be classified into two subgroups with distinct properties. Besides excitatory neurons, Calb1 also expresses in part of inhibitory neurons. But, to date, little research focused on the intersectional relationship between inhibitory neuronal subtypes and Calb1. In this study, we genetically targeted Calb1-expression (Calb1+) and Calb1-lacking (Calb1-) subgroups of PV and SST neurons throughout the mouse brain by flexibly crossing transgenic mice relying on multi-recombinant systems, and the distribution patterns and electrophysiological properties of each subgroup were further demonstrated. Thus, this study provided novel insights and strategies into inhibitory neuronal classification.
Collapse
Affiliation(s)
- Bing Zhang
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lanfang Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaomei Tang
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinyu Zeng
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yige Song
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhenye Hou
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Henok Kessete Afewerky
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Youming Lu
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Aodi He
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xinyan Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
28
|
Denis D, Baran B, Mylonas D, Spitzer C, Raymond N, Talbot C, Kohnke E, Stickgold R, Keshavan M, Manoach DS. NREM sleep oscillations and their relations with sleep-dependent memory consolidation in early course psychosis and first-degree relatives. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564703. [PMID: 37961668 PMCID: PMC10634996 DOI: 10.1101/2023.10.30.564703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Sleep spindles are believed to mediate sleep-dependent memory consolidation, particularly when coupled to neocortical slow oscillations. Schizophrenia is characterized by a deficit in sleep spindles that correlates with reduced overnight memory consolidation. Here, we examined sleep spindle activity, slow oscillation-spindle coupling, and both motor procedural and verbal declarative memory consolidation in early course, minimally medicated psychosis patients and non-psychotic first-degree relatives. Using a four-night experimental procedure, we observed significant deficits in spindle density and amplitude in patients relative to controls that were driven by individuals with schizophrenia. Schizophrenia patients also showed reduced sleep-dependent consolidation of motor procedural memory, which correlated with spindle density. Contrary to expectations, there were no group differences in the consolidation of declarative memory on a word pairs task. Nor did the relatives of patients differ in spindle activity or memory consolidation compared with controls, however increased consistency in the timing of SO-spindle coupling were seen in both patient and relatives. Our results extend prior work by demonstrating correlated deficits in sleep spindles and sleep-dependent motor procedural memory consolidation in early course, minimally medicated patients with schizophrenia, but not in first-degree relatives. This is consistent with other work in suggesting that impaired sleep-dependent memory consolidation has some specificity for schizophrenia and is a core feature rather than reflecting the effects of medication or chronicity.
Collapse
Affiliation(s)
- Dan Denis
- Department of Psychology, University of York, York, UK
| | - Bengi Baran
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Dimitrios Mylonas
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | | | | | - Christine Talbot
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Erin Kohnke
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Robert Stickgold
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Matcheri Keshavan
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Dara S Manoach
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| |
Collapse
|
29
|
Carceller H, Gramuntell Y, Klimczak P, Nacher J. Perineuronal Nets: Subtle Structures with Large Implications. Neuroscientist 2023; 29:569-590. [PMID: 35872660 DOI: 10.1177/10738584221106346] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Perineuronal nets (PNNs) are specialized structures of the extracellular matrix that surround the soma and proximal dendrites of certain neurons in the central nervous system, particularly parvalbumin-expressing interneurons. Their appearance overlaps the maturation of neuronal circuits and the closure of critical periods in different regions of the brain, setting their connectivity and abruptly reducing their plasticity. As a consequence, the digestion of PNNs, as well as the removal or manipulation of their components, leads to a boost in this plasticity and can play a key role in the functional recovery from different insults and in the etiopathology of certain neurologic and psychiatric disorders. Here we review the structure, composition, and distribution of PNNs and their variation throughout the evolutive scale. We also discuss methodological approaches to study these structures. The function of PNNs during neurodevelopment and adulthood is discussed, as well as the influence of intrinsic and extrinsic factors on these specialized regions of the extracellular matrix. Finally, we review current data on alterations in PNNs described in diseases of the central nervous system (CNS), focusing on psychiatric disorders. Together, all the data available point to the PNNs as a promising target to understand the physiology and pathologic conditions of the CNS.
Collapse
Affiliation(s)
- Héctor Carceller
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
30
|
Zhang Y, Guo Z, Yang L, Cheng C, Gai C, Gao Y, Zhang Y, Sun H, Hu D. Possible Involvement of Perineuronal Nets in Anti-Depressant Effects of Electroacupuncture in Chronic-Stress-Induced Depression in Rats. Neurochem Res 2023; 48:3146-3159. [PMID: 37347359 DOI: 10.1007/s11064-023-03970-4] [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: 09/25/2022] [Revised: 05/29/2023] [Accepted: 06/12/2023] [Indexed: 06/23/2023]
Abstract
Acupuncture can alleviate depression-like behaviors. However, the neural mechanisms behind the anti-depressive effect remain unknown. Perineuronal net (PNN) abnormalities have been reported in multiple psychiatric disorders. This study investigated the modulation and neural mechanism of PNNs in the anti-depressant process of electroacupuncture (EA) at Baihui (GV20) and Yintang (GV29) points. A rat depression model was induced by chronic unpredicted mild stress (CUMS). The results revealed that CUMS, applied for four weeks, specifically reduces PNNs around parvalbumin (PV). In addition, EA and fluoxetine treatments reverse the decrease in PNNs+ cell density and the ratio of PV and PNN double-positive cells to PV+ neurons in the medial prefrontal cortex (mPFC) after CUMS. Furthermore, EA treatment can reverse the decrease in the protein expression of PNN components (aggrecan and brevican) in the mPFC caused by stress. After EA treatment, the decreased expression of GAD67, GLuA1, and PSD95 in the mPFC induced by CUMS for four weeks was also reversed. PNN degradation in mPFC brain areas potentially interferes with the anti-depressant benefits of EA in rats with depression induced by CUMS. EA treatment did not increase PNNs+ cell density and the ratio of PV and PNN double-positive cells to PV+ neurons after PNNs degradation in the mPFC brain region of rats. This finding indicated that the mechanism of acupuncture's anti-depressant effect may be based on reversing the CUMS-induced decline in PNN expression, the functional impairment of γ-aminobutyric acid (GABA) neurons, and the regulation of excitatory synaptic proteins expression.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyu Guo
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Luping Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Cuicui Cheng
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong Gai
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yushan Gao
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hongmei Sun
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Die Hu
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
31
|
Liu D, Fujihara K, Yanagawa Y, Mushiake H, Ohshiro T. Gad1 knock-out rats exhibit abundant spike-wave discharges in EEG, exacerbated with valproate treatment. Front Neurol 2023; 14:1243301. [PMID: 37830095 PMCID: PMC10566305 DOI: 10.3389/fneur.2023.1243301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/29/2023] [Indexed: 10/14/2023] Open
Abstract
Objective To elucidate the functional role of gamma-aminobutyric acid (GABA)-ergic inhibition in suppressing epileptic brain activities such as spike-wave discharge (SWD), we recorded electroencephalogram (EEG) in knockout rats for Glutamate decarboxylase 1 (Gad1), which encodes one of the two GABA-synthesizing enzymes in mammals. We also examined how anti-epileptic drug valproate (VPA) acts on the SWDs present in Gad1 rats and affects GABA synthesis in the reticular thalamic nucleus (RTN), which is known to play an essential role in suppressing SWD. Methods Chronic EEG recordings were performed in freely moving control rats and homozygous knockout Gad1 (-/-) rats. Buzzer tones (82 dB) were delivered to the rats during EEG monitoring to test whether acoustic stimulation could interrupt ongoing SWDs. VPA was administered orally to the rats, and the change in the number of SWDs was examined. The distribution of GABA in the RTN was examined immunohistochemically. Results SWDs were abundant in EEG from Gad1 (-/-) rats as young as 2 months old. Although SWDs were universally detected in older rats irrespective of their Gad1 genotype, SWD symptom was most severe in Gad1 (-/-) rats. Acoustic stimulation readily interrupted ongoing SWDs irrespective of the Gad1 genotype, whereas SWDs were more resistant to interruption in Gad1 (-/-) rats. VPA treatment alleviated SWD symptoms in control rats, however, counterintuitively exacerbated the symptoms in Gad1 (-/-) rats. The immunohistochemistry results indicated that GABA immunoreactivity was significantly reduced in the somata of RTN neurons in Gad1 (-/-) rats but not in their axons targeting the thalamus. VPA treatment greatly increased GABA immunoreactivity in the RTN neurons of Gad1 (-/-) rats, which is likely due to the intact GAD2, another GAD isozyme, in these neurons. Discussion Our results revealed two opposing roles of GABA in SWD generation: suppression and enhancement of SWD. To account for these contradictory roles, we propose a model in which GABA produced by GAD1 in the RTN neuronal somata is released extrasynaptically and mediates intra-RTN inhibition.
Collapse
Affiliation(s)
- Dongyu Liu
- Department of Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Kazuyuki Fujihara
- Department of Psychiatry and Neuroscience, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Hajime Mushiake
- Department of Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Tomokazu Ohshiro
- Department of Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
32
|
Karanikas E. The Gordian knot of the immune-redox systems' interactions in psychosis. Int Clin Psychopharmacol 2023; 38:285-296. [PMID: 37351570 DOI: 10.1097/yic.0000000000000481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
During the last decades the attempt to enlighten the pathobiological substrate of psychosis, from merely focusing on neurotransmitters, has expanded into new areas like the immune and redox systems. Indeed, the inflammatory hypothesis concerning psychosis etiopathology has exponentially grown with findings reflecting dysfunction/aberration of the immune/redox systems' effector components namely cytokines, chemokines, CRP, complement system, antibodies, pro-/anti-oxidants, oxidative stress byproducts just to name a few. Yet, we still lie far from comprehending the underlying cellular mechanisms, their causality directions, and the moderating/mediating parameters affecting these systems; let alone the inter-systemic (between immune and redox) interactions. Findings from preclinical studies on the stress field have provided evidence indicative of multifaceted interactions among the immune and redox components so tightly intertwined as a Gordian knot. Interestingly the literature concerning the interactions between these same systems in the context of psychosis appears minimal (if not absent) and ambiguous. This review attempts to draw a frame of the immune-redox systems' interactions starting from basic research on the stress field and expanding on clinical studies with cohorts with psychosis, hoping to instigate new avenues of research.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Department of Psychiatry, 424 General Military Hospital, Ring Road, Nea Efkarpia, Thessaloniki, Greece
| |
Collapse
|
33
|
Hirai S, Sakuma A, Kunii Y, Shimbo H, Hino M, Izumi R, Nagaoka A, Yabe H, Kojima R, Seki E, Arai N, Komori T, Okado H. Disease specific brain capillary angiopathy in schizophrenia, bipolar disorder, and Alzheimer's disease. J Psychiatr Res 2023; 163:74-79. [PMID: 37207434 DOI: 10.1016/j.jpsychires.2023.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/26/2023] [Accepted: 04/08/2023] [Indexed: 05/21/2023]
Abstract
Schizophrenia (SZ) and bipolar disorder (BD), which are both psychiatric disorders, share some common clinical evidence. We recently discovered that brain capillary angiopathy is another common feature of these psychiatric disorders using fibrin accumulation in vascular endothelial cells as an indicator. This study aimed to characterize the similarities and differences in cerebral capillary injuries in various brain diseases to provide new diagnostic methods for SZ and BD and to develop new therapeutic strategies. We evaluated whether discrepancies exist in the degree of vascular damage among SZ and BD and other brain disorders (amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), and Alzheimer's disease (AD)) using postmortem brains. Our results demonstrate that fibrin was strongly accumulated in the capillaries of the grey matter (GM) of brains of patients with SZ and AD and in the capillaries of the white matter (WM) in those of patients with SZ, BD, and AD when compared with control subjects without any psychiatric or neurological disease history. However, ALS and PD brains did not present a significant increase in the amount of accumulated fibrin, either in the capillaries of WM or GM. Furthermore, significant leakage of fibrin into the brain parenchyma, indicating a vascular physical disruption, was observed in the brains of patients with AD but not in the brains of other patients compared with control subjects. In conclusion, our work reveals that Fibrin-accumulation in the brain capillaries are observed in psychiatric disorders, such as SZ, BD, and AD. Furthermore, fibrin-accumulating, nonbreaking type angiopathy is characteristic of SZ and BD, even though there are regional differences between these diseases.
Collapse
Affiliation(s)
- Shinobu Hirai
- Brain Metabolic Regulation Group, Frontier Laboratory, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan.
| | - Atsuhiro Sakuma
- Brain Metabolic Regulation Group, Frontier Laboratory, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Yasuto Kunii
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan; Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Miyagi, 980-8573, Japan
| | - Hiroko Shimbo
- Brain Metabolic Regulation Group, Frontier Laboratory, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan; Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Mizuki Hino
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan; Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Miyagi, 980-8573, Japan
| | - Ryuta Izumi
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Atsuko Nagaoka
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Hirooki Yabe
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Rika Kojima
- Laboratory of Neuropathology, Tokyo Metropolitan Institute of Medical Sciences, Tokyo, 156-8506, Japan
| | - Erika Seki
- Laboratory of Neuropathology, Tokyo Metropolitan Institute of Medical Sciences, Tokyo, 156-8506, Japan
| | - Nobutaka Arai
- Laboratory of Neuropathology, Tokyo Metropolitan Institute of Medical Sciences, Tokyo, 156-8506, Japan
| | - Takashi Komori
- Department of Pathology and Laboratory Medicine, Tokyo Metropolitan Neurological Hospital, Tokyo, 183-0042, Japan
| | - Haruo Okado
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan.
| |
Collapse
|
34
|
Valeri J, Gisabella B, Pantazopoulos H. Dynamic regulation of the extracellular matrix in reward memory processes: a question of time. Front Cell Neurosci 2023; 17:1208974. [PMID: 37396928 PMCID: PMC10311570 DOI: 10.3389/fncel.2023.1208974] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Substance use disorders are a global health problem with increasing prevalence resulting in significant socioeconomic burden and increased mortality. Converging lines of evidence point to a critical role of brain extracellular matrix (ECM) molecules in the pathophysiology of substance use disorders. An increasing number of preclinical studies highlight the ECM as a promising target for development of novel cessation pharmacotherapies. The brain ECM is dynamically regulated during learning and memory processes, thus the time course of ECM alterations in substance use disorders is a critical factor that may impact interpretation of the current studies and development of pharmacological therapies. This review highlights the evidence for the involvement of ECM molecules in reward learning, including drug reward and natural reward such as food, as well as evidence regarding the pathophysiological state of the brain's ECM in substance use disorders and metabolic disorders. We focus on the information regarding time-course and substance specific changes in ECM molecules and how this information can be leveraged for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
35
|
Zhang S, Hu S, Dong W, Huang S, Jiao Z, Hu Z, Dai S, Yi Y, Gong X, Li K, Wang H, Xu D. Prenatal dexamethasone exposure induces anxiety- and depressive-like behavior of male offspring rats through intrauterine programming of the activation of NRG1-ErbB4 signaling in hippocampal PV interneurons. Cell Biol Toxicol 2023; 39:657-678. [PMID: 34189720 DOI: 10.1007/s10565-021-09621-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
Dexamethasone is a commonly used synthetic glucocorticoid in the clinic. As a compound that can cross the placental barrier to promote fetal lung maturation, dexamethasone is extensively used in pregnant women at risk of premature delivery. However, the use of glucocorticoids during pregnancy increases the risk of neurodevelopmental disorders. In the present study, we observed anxiety- and depressive-like behavior changes and hyperexcitability of hippocampal neurons in adult rat offspring with previous prenatal dexamethasone exposure (PDE); the observed changes were related to in utero damage of parvalbumin interneurons. A programmed change in neuregulin 1 (NRG1)-Erb-b2 receptor tyrosine kinase 4 (ErbB4) signaling was the key to the damage of parvalbumin interneurons in the hippocampus of PDE offspring. Anxiety- and depressive-like behavior, NRG1-ErbB4 signaling activation, and damage of parvalbumin interneurons in PDE offspring were aggravated after chronic stress. The intervention of NRG1-ErbB4 signaling contributed to the improvement in dexamethasone-mediated injury to parvalbumin interneurons. These results suggested that PDE might cause anxiety- and depressive-like behavior changes in male rat offspring through the programmed activation of NRG1-ErbB4 signaling, resulting in damage to parvalbumin interneurons and hyperactivity of the hippocampus. Intrauterine programming of neuregulin 1 (NRG1)-Erb-b2 receptor tyrosine kinase 4 (ERBB4) overactivation by dexamethasone mediates anxiety- and depressive-like behavior in male rat offspring.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shuwei Hu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wanting Dong
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Songqiang Huang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Zhexiao Jiao
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Zewen Hu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University, Wuhan, 430071, China
| | - Shiyun Dai
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yiwen Yi
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaohan Gong
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ke Li
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Dan Xu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
36
|
Dwir D, Khadimallah I, Xin L, Rahman M, Du F, Öngür D, Do KQ. Redox and Immune Signaling in Schizophrenia: New Therapeutic Potential. Int J Neuropsychopharmacol 2023; 26:309-321. [PMID: 36975001 PMCID: PMC10229853 DOI: 10.1093/ijnp/pyad012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/27/2023] [Indexed: 03/29/2023] Open
Abstract
Redox biology and immune signaling play major roles in the body, including in brain function. A rapidly growing literature also suggests that redox and immune abnormalities are implicated in neuropsychiatric conditions such as schizophrenia (SZ), bipolar disorder, autism, and epilepsy. In this article we review this literature, its implications for the pathophysiology of SZ, and the potential for development of novel treatment interventions targeting redox and immune signaling. Redox biology and immune signaling in the brain are complex and not fully understood; in addition, there are discrepancies in the literature, especially in patient-oriented studies. Nevertheless, it is clear that abnormalities arise in SZ from an interaction between genetic and environmental factors during sensitive periods of brain development, and these abnormalities disrupt local circuits and long-range connectivity. Interventions that correct these abnormalities may be effective in normalizing brain function in psychotic disorders, especially in early phases of illness.
Collapse
Affiliation(s)
- Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Route de Cery, 1008 Prilly-Lausanne, Switzerland
| | - Ines Khadimallah
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Route de Cery, 1008 Prilly-Lausanne, Switzerland
| | - Lijing Xin
- Center for Biomedical Imaging (CIBM), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Meredith Rahman
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Fei Du
- Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Dost Öngür
- Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Route de Cery, 1008 Prilly-Lausanne, Switzerland
| |
Collapse
|
37
|
Zeng C, Liao S, Pu W. Trait and state-related characteristics of thalamo-cortical circuit disruption in bipolar disorder: a prospective cross-sectional study. Front Psychiatry 2023; 14:1067819. [PMID: 37304427 PMCID: PMC10250647 DOI: 10.3389/fpsyt.2023.1067819] [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: 10/12/2022] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
OBJECTIVE The purpose of this study is to investigate the shared and distinct thalamic-cortical circuit between bipolar depression and remission, as well as to investigate the trait and state-related characteristics of the abnormal thalamic-cortical circuit in bipolar disorder. METHODS Resting-state functional magnetic resonance imaging was performed on 38 bipolar depression patients, 40 bipolar remission patients, and 39 gender-matched healthy controls (rsfMRI). The thalamic subregions were used as seed points to draw the functional connectivity of the entire brain, and then the shared and distinct thalamic-cortical circuits between bipolar depression and remission were compared. RESULTS When compared to the healthy group, both groups of patients had significantly lower functional connectivity between the rostral temporal thalamus and the lingual gyrus, the posterior parietal thalamus, the precuneus/cerebellum, and the occipital thalamus and the precuneus; however, functional connectivity between the premotor thalamus and the superior medial frontal was significantly lower in depression. CONCLUSION This study discovered that both bipolar depression and remission had abnormal sensorimotor-thalamic functional connectivity, implying that it is a trait-related characteristic of bipolar disorder; however, the decline in prefrontal-thalamic connectivity exists specifically in bipolar depression, implying that it is a state-related characteristic of bipolar disorder.
Collapse
Affiliation(s)
- Can Zeng
- Department of Psychology, Shaoguan University, Shaoguan, China
| | - SuQun Liao
- Department of Psychology, Shaoguan University, Shaoguan, China
| | - Weidan Pu
- Department of Clinical Psychology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
38
|
Zhang A, Wang Y. Zooming into finer brain structures in vitro. Cell Stem Cell 2023; 30:503-504. [PMID: 37146575 DOI: 10.1016/j.stem.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023]
Abstract
Regionalized organoids have recently been developed to resemble a particular brain region. However, generating organoids with even finer sub-regional resolution has proven challenging. In this issue of Cell Stem Cell, Kiral et al.1 report a new organoid model resembling the human ventral thalamus and thalamic reticular nucleus.
Collapse
Affiliation(s)
- Ai Zhang
- Aspen Neuroscience, Inc., 10835 Rd. to the Cure, San Diego, CA 92121, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA; Department of Neurological Sciences, Rush University Medical Center, 1620 W. Harrison St., Chicago, IL 60612, USA; Rush Graduate College, Rush University, 600 S. Paulina St., Chicago, IL 60612, USA.
| |
Collapse
|
39
|
Chandrabhatla AS, Pomeraniec IJ, Horgan TM, Wat EK, Ksendzovsky A. Landscape and future directions of machine learning applications in closed-loop brain stimulation. NPJ Digit Med 2023; 6:79. [PMID: 37106034 PMCID: PMC10140375 DOI: 10.1038/s41746-023-00779-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/17/2023] [Indexed: 04/29/2023] Open
Abstract
Brain stimulation (BStim) encompasses multiple modalities (e.g., deep brain stimulation, responsive neurostimulation) that utilize electrodes implanted in deep brain structures to treat neurological disorders. Currently, BStim is primarily used to treat movement disorders such as Parkinson's, though indications are expanding to include neuropsychiatric disorders like depression and schizophrenia. Traditional BStim systems are "open-loop" and deliver constant electrical stimulation based on manually-determined parameters. Advancements in BStim have enabled development of "closed-loop" systems that analyze neural biomarkers (e.g., local field potentials in the sub-thalamic nucleus) and adjust electrical modulation in a dynamic, patient-specific, and energy efficient manner. These closed-loop systems enable real-time, context-specific stimulation adjustment to reduce symptom burden. Machine learning (ML) has emerged as a vital component in designing these closed-loop systems as ML models can predict / identify presence of disease symptoms based on neural activity and adaptively learn to modulate stimulation. We queried the US National Library of Medicine PubMed database to understand the role of ML in developing closed-loop BStim systems to treat epilepsy, movement disorders, and neuropsychiatric disorders. Both neural and non-neural network ML algorithms have successfully been leveraged to create closed-loop systems that perform comparably to open-loop systems. For disorders in which the underlying neural pathophysiology is relatively well understood (e.g., Parkinson's, essential tremor), most work has involved refining ML models that can classify neural signals as aberrant or normal. The same is seen for epilepsy, where most current research has focused on identifying optimal ML model design and integrating closed-loop systems into existing devices. For neuropsychiatric disorders, where the underlying pathologic neural circuitry is still being investigated, research is focused on identifying biomarkers (e.g., local field potentials from brain nuclei) that ML models can use to identify onset of symptoms and stratify severity of disease.
Collapse
Affiliation(s)
- Anirudha S Chandrabhatla
- School of Medicine, University of Virginia Health Sciences Center, Charlottesville, VA, 22903, USA
| | - I Jonathan Pomeraniec
- Surgical Neurology Branch, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
- Department of Neurosurgery, University of Virginia Health Sciences Center, Charlottesville, VA, 22903, USA.
| | - Taylor M Horgan
- School of Medicine, University of Virginia Health Sciences Center, Charlottesville, VA, 22903, USA
| | - Elizabeth K Wat
- School of Medicine, University of Virginia Health Sciences Center, Charlottesville, VA, 22903, USA
| | - Alexander Ksendzovsky
- Department of Neurosurgery, University of Maryland Medical System, Baltimore, MD, 21201, USA
| |
Collapse
|
40
|
Seah C, Huckins LM, Brennand KJ. Stem Cell Models for Context-Specific Modeling in Psychiatric Disorders. Biol Psychiatry 2023; 93:642-650. [PMID: 36658083 DOI: 10.1016/j.biopsych.2022.09.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 01/21/2023]
Abstract
Genome-wide association studies reveal the complex polygenic architecture underlying psychiatric disorder risk, but there is an unmet need to validate causal variants, resolve their target genes(s), and explore their functional impacts on disorder-related mechanisms. Disorder-associated loci regulate transcription of target genes in a cell type- and context-specific manner, which can be measured through expression quantitative trait loci. In this review, we discuss methods and insights from context-specific modeling of genetically and environmentally regulated expression. Human induced pluripotent stem cell-derived cell type and organoid models have uncovered context-specific psychiatric disorder associations by investigating tissue-, cell type-, sex-, age-, and stressor-specific genetic regulation of expression. Techniques such as massively parallel reporter assays and pooled CRISPR (clustered regularly interspaced short palindromic repeats) screens make it possible to functionally fine-map genome-wide association study loci and validate their target genes at scale. Integration of disorder-associated contexts with these patient-specific human induced pluripotent stem cell models makes it possible to uncover gene by environment interactions that mediate disorder risk, which will ultimately improve our ability to diagnose and treat psychiatric disorders.
Collapse
Affiliation(s)
- Carina Seah
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.
| | - Kristen J Brennand
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
41
|
Gandhi T, Liu CC, Adeyelu TT, Canepa CR, Lee CC. Behavioral regulation by perineuronal nets in the prefrontal cortex of the CNTNAP2 mouse model of autism spectrum disorder. Front Behav Neurosci 2023; 17:1114789. [PMID: 36998537 PMCID: PMC10043266 DOI: 10.3389/fnbeh.2023.1114789] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
Autism spectrum disorders (ASDs) arise from altered development of the central nervous system, and manifest behaviorally as social interaction deficits and restricted and repetitive behaviors. Alterations to parvalbumin (PV) expressing interneurons have been implicated in the neuropathological and behavioral deficits in autism. In addition, perineuronal nets (PNNs), specialized extracellular matrix structures that enwrap the PV-expressing neurons, also may be altered, which compromises neuronal function and susceptibility to oxidative stress. In particular, the prefrontal cortex (PFC), which regulates several core autistic traits, relies on the normal organization of PNNs and PV-expressing cells, as well as other neural circuit elements. Consequently, we investigated whether PNNs and PV-expressing cells were altered in the PFC of the CNTNAP2 knockout mouse model of ASD and whether these contributed to core autistic-like behaviors in this model system. We observed an overexpression of PNNs, PV-expressing cells, and PNNs enwrapping PV-expressing cells in adult CNTNAP2 mice. Transient digestion of PNNs from the prefrontal cortex (PFC) by injection of chondroitinase ABC in CNTNAP2 mutant mice rescued some of the social interaction deficits, but not the restricted and repetitive behaviors. These findings suggest that the neurobiological regulation of PNNs and PVs in the PFC contribute to social interaction behaviors in neurological disorders including autism.
Collapse
Affiliation(s)
- Tanya Gandhi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Chin-Chi Liu
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Tolulope T. Adeyelu
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Cade R. Canepa
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
42
|
Do KQ. Bridging the gaps towards precision psychiatry: Mechanistic biomarkers for early detection and intervention. Psychiatry Res 2023; 321:115064. [PMID: 36716550 DOI: 10.1016/j.psychres.2023.115064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023]
Abstract
Early detection and intervention in schizophrenia, improving prognosis, requires mechanism-based biomarkers that capture circuitry dysfunction, allowing optimized patient stratification, disease monitoring and treatment. Dr. Do's translational research, bridging basic neuroscience and clinical psychiatry, tackles an urgent need to develop effective treatments that target mechanisms underlying cognitive deficits, a critical dimension of schizophrenia, currently not well treated. By adopting a reverse translation of validated circuitry relevant human endpoints, her research brought new insights in mechanism-based biomarker guided treatment of patients in early stages of psychosis. She showed that oxidative stress/redox dysregulation, in reciprocal interaction with dopamine imbalance, NMDAR hypofunction, neuroinflammation and mitochondrial bioenergetic dysfunction, may represent a "hub" on which both genetic and environmental risk factors converge during neurodevelopment. This leads to impairments of structural and functional connectivity in microcircuits, involving impaired parvalbumin fast-spiking GABA neurons, and macrocircuits, impacting myelination of fiber tracts, at the basis of neural synchronization abnormalities, as well as sensory and cognitive deficits. These unique insights led to successful proof-of-concept clinical trials, targeting oxidative stress through antioxidant-based strategies in patients at various disease stages, paving the way for precision medicine in psychiatry.
Collapse
Affiliation(s)
- Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
43
|
Alemán-Gómez Y, Baumgartner T, Klauser P, Cleusix M, Jenni R, Hagmann P, Conus P, Do KQ, Bach Cuadra M, Baumann PS, Steullet P. Multimodal Magnetic Resonance Imaging Depicts Widespread and Subregion Specific Anomalies in the Thalamus of Early-Psychosis and Chronic Schizophrenia Patients. Schizophr Bull 2023; 49:196-207. [PMID: 36065156 PMCID: PMC9810016 DOI: 10.1093/schbul/sbac113] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND HYPOTHESIS Although the thalamus has a central role in schizophrenia pathophysiology, contributing to sensory, cognitive, and sleep alterations, the nature and dynamics of the alterations occurring within this structure remain largely elusive. Using a multimodal magnetic resonance imaging (MRI) approach, we examined whether anomalies: (1) differ across thalamic subregions/nuclei, (2) are already present in the early phase of psychosis (EP), and (3) worsen in chronic schizophrenia (SCHZ). STUDY DESIGN T1-weighted and diffusion-weighted images were analyzed to estimate gray matter concentration (GMC) and microstructural parameters obtained from the spherical mean technique (intra-neurite volume fraction [VFINTRA)], intra-neurite diffusivity [DIFFINTRA], extra-neurite mean diffusivity [MDEXTRA], extra-neurite transversal diffusivity [TDEXTRA]) within 7 thalamic subregions. RESULTS Compared to age-matched controls, the thalamus of EP patients displays previously unreported widespread microstructural alterations (VFINTRA decrease, TDEXTRA increase) that are associated with similar alterations in the whole brain white matter, suggesting altered integrity of white matter fiber tracts in the thalamus. In both patient groups, we also observed more localized and heterogenous changes (either GMC decrease, MDEXTRA increase, or DIFFINTRA decrease) in mediodorsal, posterior, and ventral anterior parts of the thalamus in both patient groups, suggesting that the nature of the alterations varies across subregions. GMC and DIFFINTRA in the whole thalamus correlate with global functioning, while DIFFINTRA in the subregion encompassing the medial pulvinar is significantly associated with negative symptoms in SCHZ. CONCLUSION Our data reveals both widespread and more localized thalamic anomalies that are already present in the early phase of psychosis.
Collapse
Affiliation(s)
- Yasser Alemán-Gómez
- Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Prilly, Switzerland
| | - Thomas Baumgartner
- Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Paul Klauser
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Prilly, Switzerland
- Department of Psychiatry, Service of Child and Adolescent Psychiatry, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Martine Cleusix
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Prilly, Switzerland
| | - Raoul Jenni
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Prilly, Switzerland
| | - Patric Hagmann
- Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philippe Conus
- Department of Psychiatry, Service of General Psychiatry, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Kim Q Do
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Prilly, Switzerland
| | - Meritxell Bach Cuadra
- Medical Image Analysis Laboratory (MIAL), Centre d’Imagerie BioMédicale (CIBM), Switzerland
- Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philipp S Baumann
- Department of Psychiatry, Service of General Psychiatry, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Pascal Steullet
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Prilly, Switzerland
| |
Collapse
|
44
|
Wartchow KM, Scaini G, Quevedo J. Glial-Neuronal Interaction in Synapses: A Possible Mechanism of the Pathophysiology of Bipolar Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:191-208. [PMID: 36949311 DOI: 10.1007/978-981-19-7376-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Bipolar disorder (BD) is a severe and chronic psychiatric disorder that affects approximately 1-4% of the world population and is characterized by recurrent episodes of mania or hypomania and depression. BD is also associated with illnesses marked by immune activation, such as metabolic syndrome, obesity, type 2 diabetes mellitus, and cardiovascular diseases. Indeed, a connection has been suggested between neuroinflammation and peripheral inflammatory markers in the pathophysiology of BD, which can be associated with the modulation of many dysfunctional processes, including synaptic plasticity, neurotransmission, neurogenesis, neuronal survival, apoptosis, and even cognitive/behavioral functioning. Rising evidence suggests that synaptic dysregulations, especially glutamatergic system dysfunction, are directly involved in mood disorders. It is becoming clear that dysregulations in connection and structural changes of glial cells play a central role in the BD pathophysiology. This book chapter highlighted the latest findings that support the theory of synaptic dysfunction in BD, providing an overview of the alterations in neurotransmitters release, astrocytic uptake, and receptor signaling, as well as the role of inflammation on glial cells in mood disorders. Particular emphasis is given to the alterations in presynaptic and postsynaptic neurons and glial cells, all cellular elements of the "tripartite synapse," compromising the neurotransmitters system, excitatory-inhibitory balance, and neurotrophic states of local networks in mood disorders. Together, these studies provide a foundation of knowledge about the exact role of the glial-neuronal interaction in mood disorders.
Collapse
Affiliation(s)
- Krista M Wartchow
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Giselli Scaini
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - João Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
45
|
Yu BJ, Oz RS, Sethi S. Ketogenic diet as a metabolic therapy for bipolar disorder: Clinical developments. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2023. [DOI: 10.1016/j.jadr.2022.100457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
46
|
Hádinger N, Bősz E, Tóth B, Vantomme G, Lüthi A, Acsády L. Region-selective control of the thalamic reticular nucleus via cortical layer 5 pyramidal cells. Nat Neurosci 2023; 26:116-130. [PMID: 36550291 PMCID: PMC9829539 DOI: 10.1038/s41593-022-01217-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
Corticothalamic pathways, responsible for the top-down control of the thalamus, have a canonical organization such that every cortical region sends output from both layer 6 (L6) and layer 5 (L5) to the thalamus. Here we demonstrate a qualitative, region-specific difference in the organization of mouse corticothalamic pathways. Specifically, L5 pyramidal cells of the frontal cortex, but not other cortical regions, establish monosynaptic connections with the inhibitory thalamic reticular nucleus (TRN). The frontal L5-TRN pathway parallels the L6-TRN projection but has distinct morphological and physiological features. The exact spike output of the L5-contacted TRN cells correlated with the level of cortical synchrony. Optogenetic perturbation of the L5-TRN connection disrupted the tight link between cortical and TRN activity. L5-driven TRN cells innervated thalamic nuclei involved in the control of frontal cortex activity. Our data show that frontal cortex functions require a highly specialized cortical control over intrathalamic inhibitory processes.
Collapse
Affiliation(s)
- Nóra Hádinger
- Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, Hungary.
| | - Emília Bősz
- Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Boglárka Tóth
- Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, Hungary
| | - Gil Vantomme
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - László Acsády
- Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
47
|
Gene set enrichment analysis of pathophysiological pathways highlights oxidative stress in psychosis. Mol Psychiatry 2022; 27:5135-5143. [PMID: 36131045 PMCID: PMC9763118 DOI: 10.1038/s41380-022-01779-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 01/14/2023]
Abstract
Polygenic risk prediction remains an important aim of genetic association studies. Currently, the predictive power of schizophrenia polygenic risk scores (PRSs) is not large enough to allow highly accurate discrimination between cases and controls and thus is not adequate for clinical integration. Since PRSs are rarely used to reveal biological functions or to validate candidate pathways, to fill this gap, we investigated whether their predictive ability could be improved by building genome-wide (GW-PRSs) and pathway-specific PRSs, using distance- or expression quantitative trait loci (eQTLs)- based mapping between genetic variants and genes. We focused on five pathways (glutamate, oxidative stress, GABA/interneurons, neuroimmune/neuroinflammation and myelin) which belong to a critical hub of schizophrenia pathophysiology, centred on redox dysregulation/oxidative stress. Analyses were first performed in the Lausanne Treatment and Early Intervention in Psychosis Program (TIPP) study (n = 340, cases/controls: 208/132), a sample of first-episode of psychosis patients and matched controls, and then validated in an independent study, the epidemiological and longitudinal intervention program of First-Episode Psychosis in Cantabria (PAFIP) (n = 352, 224/128). Our results highlighted two main findings. First, GW-PRSs for schizophrenia were significantly associated with early psychosis status. Second, oxidative stress was the only significantly associated pathway that showed an enrichment in both the TIPP (p = 0.03) and PAFIP samples (p = 0.002), and exclusively when gene-variant linking was done using eQTLs. The results suggest that the predictive accuracy of polygenic risk scores could be improved with the inclusion of information from functional annotations, and through a focus on specific pathways, emphasizing the need to build and study functionally informed risk scores.
Collapse
|
48
|
The role of thalamic group II mGlu receptors in health and disease. Neuronal Signal 2022; 6:NS20210058. [PMID: 36561092 PMCID: PMC9760452 DOI: 10.1042/ns20210058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 12/25/2022] Open
Abstract
The thalamus plays a pivotal role in the integration and processing of sensory, motor, and cognitive information. It is therefore important to understand how the thalamus operates in states of both health and disease. In the present review, we discuss the function of the Group II metabotropic glutamate (mGlu) receptors within thalamic circuitry, and how they may represent therapeutic targets in treating disease states associated with thalamic dysfunction.
Collapse
|
49
|
John U, Patro N, Patro I. Perineuronal nets: Cruise from a honeycomb to the safety nets. Brain Res Bull 2022; 190:179-194. [DOI: 10.1016/j.brainresbull.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/17/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
|
50
|
Czekus C, Steullet P, Orero López A, Bozic I, Rusterholz T, Bandarabadi M, Do KQ, Gutierrez Herrera C. Alterations in TRN-anterodorsal thalamocortical circuits affect sleep architecture and homeostatic processes in oxidative stress vulnerable Gclm -/- mice. Mol Psychiatry 2022; 27:4394-4406. [PMID: 35902628 PMCID: PMC9734061 DOI: 10.1038/s41380-022-01700-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Schizophrenia is associated with alterations of sensory integration, cognitive processing and both sleep architecture and sleep oscillations in mouse models and human subjects, possibly through changes in thalamocortical dynamics. Oxidative stress (OxS) damage, including inflammation and the impairment of fast-spiking gamma-aminobutyric acid neurons have been hypothesized as a potential mechanism responsible for the onset and development of schizophrenia. Yet, the link between OxS and perturbation of thalamocortical dynamics and sleep remains unclear. Here, we sought to investigate the effects of OxS on sleep regulation by characterizing the dynamics of thalamocortical networks across sleep-wake states in a mouse model with a genetic deletion of the modifier subunit of glutamate-cysteine ligase (Gclm knockout, KO) using high-density electrophysiology in freely-moving mice. We found that Gcml KO mice exhibited a fragmented sleep architecture and impaired sleep homeostasis responses as revealed by the increased NREM sleep latencies, decreased slow-wave activities and spindle rate after sleep deprivation. These changes were associated with altered bursting activity and firing dynamics of neurons from the thalamic reticularis nucleus, anterior cingulate and anterodorsal thalamus. Administration of N-acetylcysteine (NAC), a clinically relevant antioxidant, rescued the sleep fragmentation and spindle rate through a renormalization of local neuronal dynamics in Gclm KO mice. Collectively, these findings provide novel evidence for a link between OxS and the deficits of frontal TC network dynamics as a possible mechanism underlying sleep abnormalities and impaired homeostatic responses observed in schizophrenia.
Collapse
Affiliation(s)
- Christina Czekus
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Site de Cery, CH-1008, Prilly-Lausanne, Switzerland
| | - Albert Orero López
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland
| | - Ivan Bozic
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Thomas Rusterholz
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland
| | - Mojtaba Bandarabadi
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Site de Cery, CH-1008, Prilly-Lausanne, Switzerland
| | - Carolina Gutierrez Herrera
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland.
- Department for Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|