1
|
Zheng XX, Wang F, Ding H, Li HT, Yang XJ, Li XC, Dou ZW, Hu WC, Han WJ, Li ZZ, Li YC, Chu WG, Yuan H, Wu SX, Xie RG, Luo C. cGMP-dependent protein kinase I in the dorsal hippocampus protects against synaptic plasticity and cognitive deficit induced by chronic pain. Pain 2025:00006396-990000000-00888. [PMID: 40310865 DOI: 10.1097/j.pain.0000000000003624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 03/07/2025] [Indexed: 05/03/2025]
Abstract
ABSTRACT Patients with chronic pain often experience an exacerbated pain response and complain of memory deficits. However, the mechanistic link between pain and cognitive function remains unclear. The dorsal hippocampus (dHPC), a well-defined region responsible for learning and memory, displays maladaptive plasticity upon injury, which involves the activation of N-methyl-d-aspartic acid receptors. Mounting evidence has shown that cyclic guanosine cGMP-dependent protein kinase I (PKG-I) serves as a key downstream target of the N-methyl-d-aspartic acid receptors-NO-cGMP signaling pathway, regulating neuronal plasticity, pain hypersensitivity, and pain-related affective disorders. Despite these advances, it has remained elusive whether and how PKG-I in the dHPC contributes to hippocampal plasticity, as well as to chronic pain and pain-related cognitive deficits. In this study, we disclosed the crucial role of PKG-I in the dHPC in chronic pain and pain-related cognitive deficits. Following nerve injury, mice exhibited mechanical allodynia and thermal hyperalgesia, along with pain-related cognitive impairments; these changes were accompanied by the downregulation of PKG-I at both mRNA and protein levels in the dHPC. Overexpression of PKG-I in the dHPC alleviated pain hypersensitivity and associated cognitive deficits. Further mechanistic analysis revealed that PKG-I contributes to modulating Ca2+ mobilization in hippocampal pyramidal neurons, which brings about the production and secretion of a brain-derived neurotrophic factor in the dHPC. The resultant increase of the brain-derived neurotrophic factor in turn enhanced hippocampal neuronal excitability and synaptic plasticity and thus relieved pain hypersensitivity and pain-related cognitive impairment. Our findings extended the functional capability of hippocampal PKG-I on chronic pain and pain-related cognitive impairment. Hippocampal PKG-I may represent a novel therapeutic target for the treatment of chronic pain and pain-related memory deficits.
Collapse
Affiliation(s)
- Xing-Xing Zheng
- College of Life Sciences, Northwest University, Xi'an, China
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Fei Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hui Ding
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hai-Tao Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- The Fourteenth Squadron of the Fourth Regiment, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Xin-Jiang Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiang-Chen Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- The Third Squadron of the First Regiment, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhi-Wei Dou
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Class 2018, The Twenty-fourth Squadron of the Sixth Brigade, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Wen-Chao Hu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Class 2018, The Twenty-fourth Squadron of the Sixth Brigade, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Wen-Juan Han
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhen-Zhen Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ying-Chun Li
- College of Life Sciences, Northwest University, Xi'an, China
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wen-Guang Chu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hua Yuan
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
2
|
Wang F, Tian ZC, Ding H, Yang XJ, Wang FD, Ji RX, Xu L, Cao ZX, Ma SB, Zhang M, Cui YT, Cong XY, Chu WG, Li ZZ, Han WJ, Gao YH, Yu YW, Zhao XH, Wang WT, Xie RG, Wu SX, Luo C. A sensory-motor-sensory circuit underlies antinociception ignited by primary motor cortex in mice. Neuron 2025:S0896-6273(25)00246-6. [PMID: 40239652 DOI: 10.1016/j.neuron.2025.03.027] [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: 12/06/2023] [Revised: 02/05/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025]
Abstract
Sensory-motor integration is crucial in the processing of chronic pain. The primary motor cortex (M1) is emerging as a promising target for chronic pain treatment. However, it remains elusive how nociceptive sensory inputs influence M1 activity and how rectifying M1 defects, in turn, regulates pain processing at cellular and network levels. We show that injury/inflammation leads to hypoactivity of M1Glu pyramidal neurons by excitation-inhibition imbalance between the primary somatosensory cortex (S1) and the M1. The impaired M1 output further weakens inputs to excitatory parvalbumin neurons of the lateral hypothalamus (LHPV) and impairs the descending inhibitory system, hence exacerbating spinal nociceptive sensitivity. When rectifying M1 defects with repetitive transcranial magnetic stimulation (rTMS), the imbalance of the S1-M1 microcircuitry can be effectively reversed, which aids in restoring the ability of the M1 to trigger the descending inhibitory system, thereby alleviating nociceptive hypersensitivity. Thus, a sensory-motor-sensory loop is identified for pain-related interactions between the sensory and motor systems and can be potentially exploited for treating chronic pain.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046, China; Shaanxi Province Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Zhi-Cheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Hui Ding
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xin-Jiang Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Department of Rehabilitation and Physical Therapy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Fu-Dong Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ruo-Xin Ji
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Lei Xu
- The Sixteenth Squadron of Fourth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Zi-Xuan Cao
- The Twenty-Second Squadron of Sixth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Sui-Bin Ma
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ming Zhang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ya-Ting Cui
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiang-Yu Cong
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Guang Chu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Zhen-Zhen Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Juan Han
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yong-Heng Gao
- Department of Respiration, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan-Wang Yu
- Shaanxi Province Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiang-Hui Zhao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Ting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
3
|
Yu Y, Zhao Y, Feng J, Lian N, Yu J, Yang Y, Yao J, Yu Y. Repeated Exposure to Sevoflurane in Neonatal Mice Induces Cognitive and Synaptic Impairments in a TTLL6-Mediated Tubulin Polyglutamylation Manner. CNS Neurosci Ther 2025; 31:e70376. [PMID: 40202105 PMCID: PMC11979716 DOI: 10.1111/cns.70376] [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: 12/09/2024] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 04/10/2025] Open
Abstract
AIMS Repeated sevoflurane exposure during the neonatal stage may induce Tau phosphorylation, dendritic spine loss, and neurocognitive impairment in the developing brain. Tubulin tyrosine ligase like-6 (TTLL6), which aggregates in dendrites due to Tau missorting, regulates microtubule stability via α-tubulin polyglutamylation. Meanwhile, Spastin modulates dendritic spine formation by severing microtubules. We hypothesize that repeated sevoflurane treatment impairs dendritic spine remodeling in neonatal mice by enhancing TTLL6-mediated tubulin polyglutamylation and increasing Spastin expression, leading to cognitive dysfunction in their pre-adolescent stage. METHODS Six-day-old wild type (WT), TTLL6 brain conditional knockout (TTLL6CKO), TTLL6-flox (TTLL6CON) and Tau-knockout mice were treated with 3% sevoflurane for 2 h daily on postnatal days (P) 6, 8, and 10. Levels of Tau, phosphorylated Tau (pTau), TTLL6, polyglutamylated tubulin, ATP, Spastin, PSD95, Tau-TTLL6 interaction, Tau-TTLL6 missorting, dendritic spine remodeling, and behavioral alterations were compared across these groups. RESULTS Repeated sevoflurane exposure during brain development in neonatal mice could reduce dendritic spine density, synapse number, PSD95, and ATP levels, while increasing pTau, polyglutamylated tubulin, Tau-TTLL6 missorting from axons to the somatodendritic compartment, and Spastin levels, leading to cognitive impairment later in their pre-adolescent stage (P30). However, these changes were ameliorated in the TTLL6CKO mice. CONCLUSIONS Repeated neonatal sevoflurane exposure results in synaptic impairment through TTLL6-mediated tubulin polyglutamylation and increased Spastin expression, causing pre-adolescent cognitive dysfunction in mice. This process is initiated by Tau phosphorylation and missorting from axons to somatodendritic compartments.
Collapse
Affiliation(s)
- Yang Yu
- Department of AnaesthesiaTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnaesthesiologyTianjinChina
| | - Yue Zhao
- Department of AnaesthesiologyShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Jingyu Feng
- Department of AnaesthesiaTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnaesthesiologyTianjinChina
| | - Naqi Lian
- Department of AnaesthesiaTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnaesthesiologyTianjinChina
| | - Jiafeng Yu
- Department of AnaesthesiaTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnaesthesiologyTianjinChina
| | - Yongyan Yang
- Department of AnaesthesiaTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnaesthesiologyTianjinChina
| | - Junyan Yao
- Department of AnaesthesiologyShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Yonghao Yu
- Department of AnaesthesiaTianjin Medical University General HospitalTianjinChina
- Tianjin Institute of AnaesthesiologyTianjinChina
| |
Collapse
|
4
|
Kim GW, Cha M, Ong HTM, Yoo J, Jeon YH, Lee SW, Oh SY, Kang MJ, Kim Y, Kwon SH. HDAC6 and USP9X Control Glutamine Metabolism by Stabilizing GS to Promote Glioblastoma Tumorigenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501553. [PMID: 40162736 DOI: 10.1002/advs.202501553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Glioblastoma (GBM) is the most common and the deadliest brain cancer. Glutamine anabolism mediated by glutamine synthetase (GS) is beneficial for GBM cell growth, especially under glutamine deprivation. However, the molecular mechanism underlying GS homeostasis in GBM remains undisclosed. Here, it is reported that histone deacetylase 6 (HDAC6) promotes GS deacetylation, stabilizing it via ubiquitin-mediated pathway. It is found that deubiquitination of GS is modulated by ubiquitin-specific peptidase 9, X-linked (USP9X). USP9X stabilizes GS by removing its K48-linked polyubiquitination on lysine 91 and 103. Accordingly, targeting HDAC6 and USP9X in vitro and in vivo represses GBM tumorigenesis by decreasing GS stability. Metabolic analysis shows that silencing HDAC6 and USP9X disrupts de novo nucleotide synthesis, thereby attenuating GBM cell growth. Furthermore, GS modulation by targeting HDAC6 and USP9X restrains the self-renewal capacity. These results suggest that HDAC6 and USP9X are crucial epigenetic enzymes that promote GBM tumorigenesis by modulating glutamine metabolism.
Collapse
Affiliation(s)
- Go Woon Kim
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - Minhae Cha
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - Hien Thi My Ong
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jung Yoo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - Yu Hyun Jeon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - Sang Wu Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - Soo Yeon Oh
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - Min-Jung Kang
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Youngsoo Kim
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| |
Collapse
|
5
|
Yoon S, Penzes P. Roles of ANK2/ankyrin-B in neurodevelopmental disorders: Isoform functions and implications for autism spectrum disorder and epilepsy. Curr Opin Neurobiol 2025; 90:102938. [PMID: 39631164 PMCID: PMC11839328 DOI: 10.1016/j.conb.2024.102938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
The ANK2 gene, encoding ankyrin-B, is a high-confidence risk factor for neurodevelopmental disorders (NDDs). Evidence from exome sequencing studies have repeatedly implicated rare variants in ANK2 in autism spectrum disorder. Recently, the functions of ankyrin-B isoforms on neuronal phenotypes have been investigated using a number of techniques including electrophysiology, proteomic screens and behavioral analysis using animal models with loss of distinct Ank2 isoforms or with targeted loss of Ank2 in different cell types and time points during brain development. ANK2 variants and their pathophysiology could provide valuable insights into the molecular mechanisms underlying NDDs. In this review, we focus on recently reported studies to help understand the pathological mechanisms of ANK2 loss and how it may facilitate the development of treatments for NDDs in the future.
Collapse
Affiliation(s)
- Sehyoun Yoon
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Northwestern University, Center for Autism and Neurodevelopment, Chicago, IL, 60611, USA
| |
Collapse
|
6
|
Yang ZH, Cai X, Zhang CY, Zhang Q, Li M, Ding ZL, Guo Y, Ma G, Yang CH, Guo L, Chang H, Wang C, Li M, Xiao X. NEK4 modulates circadian fluctuations of emotional behaviors and synaptogenesis in male mice. Nat Commun 2024; 15:9180. [PMID: 39448584 PMCID: PMC11502819 DOI: 10.1038/s41467-024-53585-8] [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: 03/07/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
GWASs have linked the 3p21.1 locus, which is associated with the expression levels of NEK4, to bipolar disorder. Here, we use integrative analyses of GWAS statistics and eQTL annotations to establish that elevated NEK4 expression in the hippocampus is associated with an increased risk of bipolar disorder. To further study this association, we generate transgenic male mice that conditionally overexpress NEK4 in the pyramidal neurons of the adult forebrain, or use AAV to overexpress NEK4 in the dorsal hippocampus. Compared to the control mice, male mice of both strains exhibit a shift from a diurnal anxiety state to a nocturnal normal or anxiolytic-like state. Overexpression of NEK4 also affects the circadian fluctuations in dendritic spine morphology and synaptic structure. Furthermore, we show that treatment with lithium ameliorates the effects of NEK4 overexpression in male mice. We then perform phosphoproteomic analyses to demonstrate that the diurnal and nocturnal phosphoproteomic profiles of male control and NEK4 overexpressing mice are different. These results suggest that male mice with different NEK4 expression levels may recapitulate some of the core features observed in patients with bipolar disorder, indicating that interruption of the homeostatic dynamics of synapses may underlie the emotional swings in bipolar disorder.
Collapse
Affiliation(s)
- Zhi-Hui Yang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xin Cai
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qing Zhang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Miao Li
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhong-Li Ding
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yingqi Guo
- Institutional Center for Shared Technologies and Facilities of Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Guolan Ma
- Institutional Center for Shared Technologies and Facilities of Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chao-Hao Yang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Lei Guo
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Hong Chang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chuang Wang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Ming Li
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Xiao Xiao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
7
|
Lukin J, Smith CM, De Rubeis S. Emerging X-linked genes associated with neurodevelopmental disorders in females. Curr Opin Neurobiol 2024; 88:102902. [PMID: 39167997 PMCID: PMC11392613 DOI: 10.1016/j.conb.2024.102902] [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: 04/16/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
A significant source of risk for neurodevelopmental disorders (NDDs), including intellectual disability (ID) and autism spectrum disorder (ASD), lies in genes located on the X chromosome. Males can be particularly vulnerable to X-linked variation because of hemizygosity, and male-specific segregation in pedigrees has guided earlier gene discovery for X-linked recessive conditions. More recently, X-linked disorders disproportionally affecting females, with complex inheritance patterns and/or presenting with sex differences, have surfaced. Here, we discuss the genetics and neurobiology of X-linked genes that are paradigmatic to understand NDDs in females. Integrating genetic, clinical, and functional data will be key to understand how X-linked variation contributes to the risk architecture of NDDs.
Collapse
Affiliation(s)
- Jeronimo Lukin
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Corinne M Smith
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
8
|
Zenge C, Ordureau A. Ubiquitin system mutations in neurological diseases. Trends Biochem Sci 2024; 49:875-887. [PMID: 38972780 PMCID: PMC11455613 DOI: 10.1016/j.tibs.2024.06.011] [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: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Neuronal ubiquitin balance impacts the fate of countless cellular proteins, and its disruption is associated with various neurological disorders. The ubiquitin system is critical for proper neuronal cell state transitions and the clearance of misfolded or aggregated proteins that threaten cellular integrity. This article reviews the state of and recent advancements in our understanding of the disruptions to components of the ubiquitin system, in particular E3 ligases and deubiquitylases, in neurodevelopmental and neurodegenerative diseases. Specific focus is on enzymes with recent progress in their characterization, including identifying enzyme-substrate pairs, the use of stem cell and animal models, and the development of therapeutics for ubiquitin-related diseases.
Collapse
Affiliation(s)
- Colin Zenge
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
9
|
Nagata N, Kurosaka H, Higashi K, Yamaguchi M, Yamamoto S, Inubushi T, Nagata M, Ishihara Y, Yonei A, Miyashita Y, Asano Y, Sakai N, Sakata Y, Kawabata S, Yamashiro T. Characteristic craniofacial defects associated with a novel USP9X truncation mutation. Hum Genome Var 2024; 11:21. [PMID: 38755172 PMCID: PMC11099082 DOI: 10.1038/s41439-024-00277-w] [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: 01/29/2024] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 05/18/2024] Open
Abstract
Germline loss-of-function mutations in USP9X have been reported to cause a wide spectrum of congenital anomalies. Here, we report a Japanese girl with a novel heterozygous nonsense mutation in USP9X who exhibited intellectual disability with characteristic craniofacial abnormalities, including hypotelorism, brachycephaly, hypodontia, micrognathia, severe dental crowding, and an isolated submucous cleft palate. Our findings provide further evidence that disruptions in USP9X contribute to a broad range of congenital craniofacial abnormalities.
Collapse
Affiliation(s)
- Namiki Nagata
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Suita, Japan.
| | - Kotaro Higashi
- Department of Microbiology, Osaka University Graduate School of Dentistry, Suita, Japan
- Department of Removable Prosthodontics and Gerodontology, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Masaya Yamaguchi
- Department of Microbiology, Osaka University Graduate School of Dentistry, Suita, Japan
- Bioinformatics Research Unit, Osaka University Graduate School of Dentistry, Suita, Japan
- Bioinformatics Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Suita, Japan
| | - Sayuri Yamamoto
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Toshihiro Inubushi
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Miho Nagata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasuki Ishihara
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ayumi Yonei
- Department of Genetic Counseling, Osaka University Hospital, Osaka, Japan
| | - Yohei Miyashita
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Norio Sakai
- Child Healthcare and Genetic Science Laboratory, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shigetada Kawabata
- Department of Microbiology, Osaka University Graduate School of Dentistry, Suita, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Suita, Japan
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Suita, Japan
| |
Collapse
|
10
|
Tang Y, Wu J, Liu C, Gan L, Chen H, Sun YL, Liu J, Tao YX, Zhu T, Chen C. Schwann cell-derived extracellular vesicles promote memory impairment associated with chronic neuropathic pain. J Neuroinflammation 2024; 21:99. [PMID: 38632655 PMCID: PMC11025217 DOI: 10.1186/s12974-024-03081-z] [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: 01/24/2024] [Accepted: 03/30/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND The pathogenesis of memory impairment, a common complication of chronic neuropathic pain (CNP), has not been fully elucidated. Schwann cell (SC)-derived extracellular vesicles (EVs) contribute to remote organ injury. Here, we showed that SC-EVs may mediate pathological communication between SCs and hippocampal neurons in the context of CNP. METHODS We used an adeno-associated virus harboring the SC-specific promoter Mpz and expressing the CD63-GFP gene to track SC-EVs transport. microRNA (miRNA) expression profiles of EVs and gain-of-function and loss-of-function regulatory experiments revealed that miR-142-5p was the main cargo of SC-EVs. Next, luciferase reporter gene and phenotyping experiments confirmed the direct targets of miR-142-5p. RESULTS The contents and granule sizes of plasma EVs were significantly greater in rats with chronic sciatic nerve constriction injury (CCI)than in sham rats. Administration of the EV biogenesis inhibitor GW4869 ameliorated memory impairment in CCI rats and reversed CCI-associated dendritic spine damage. Notably, during CCI stress, SC-EVs could be transferred into the brain through the circulation and accumulate in the hippocampal CA1-CA3 regions. miR-142-5p was the main cargo wrapped in SC-EVs and mediated the development of CCI-associated memory impairment. Furthermore, α-actinin-4 (ACTN4), ELAV-like protein 4 (ELAVL4) and ubiquitin-specific peptidase 9 X-linked (USP9X) were demonstrated to be important downstream target genes for miR-142-5p-mediated regulation of dendritic spine damage in hippocampal neurons from CCI rats. CONCLUSION Together, these findings suggest that SCs-EVs and/or their cargo miR-142-5p may be potential therapeutic targets for memory impairment associated with CNP.
Collapse
Affiliation(s)
- Yidan Tang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiahui Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Changliang Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lu Gan
- Research Laboratory of Emergency Medicine, West China Hospital, Emergency Medicine and National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hai Chen
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ya-Lan Sun
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Chan Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
11
|
Murphy KE, Duncan B, Sperringer JE, Zhang E, Haberman V, Wyatt EV, Maness P. Ankyrin B promotes developmental spine regulation in the mouse prefrontal cortex. Cereb Cortex 2023; 33:10634-10648. [PMID: 37642601 PMCID: PMC10560577 DOI: 10.1093/cercor/bhad311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Postnatal regulation of dendritic spine formation and refinement in cortical pyramidal neurons is critical for excitatory/inhibitory balance in neocortical networks. Recent studies have identified a selective spine pruning mechanism in the mouse prefrontal cortex mediated by class 3 Semaphorins and the L1 cell adhesion molecules, neuron-glia related cell adhesion molecule, Close Homolog of L1, and L1. L1 cell adhesion molecules bind Ankyrin B, an actin-spectrin adaptor encoded by Ankyrin2, a high-confidence gene for autism spectrum disorder. In a new inducible mouse model (Nex1Cre-ERT2: Ank2flox: RCE), Ankyrin2 deletion in early postnatal pyramidal neurons increased spine density on apical dendrites in prefrontal cortex layer 2/3 of homozygous and heterozygous Ankyrin2-deficient mice. In contrast, Ankyrin2 deletion in adulthood had no effect on spine density. Sema3F-induced spine pruning was impaired in cortical neuron cultures from Ankyrin B-null mice and was rescued by re-expression of the 220 kDa Ankyrin B isoform but not 440 kDa Ankyrin B. Ankyrin B bound to neuron-glia related CAM at a cytoplasmic domain motif (FIGQY1231), and mutation to FIGQH inhibited binding, impairing Sema3F-induced spine pruning in neuronal cultures. Identification of a novel function for Ankyrin B in dendritic spine regulation provides insight into cortical circuit development, as well as potential molecular deficiencies in autism spectrum disorder.
Collapse
Affiliation(s)
- Kelsey E Murphy
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Bryce Duncan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Justin E Sperringer
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Erin Zhang
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Victoria Haberman
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Elliott V Wyatt
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Patricia Maness
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| |
Collapse
|
12
|
Sha Z, Warrier V, Bethlehem RA, Schultz LM, Merikangas A, Sun KY, Gur RC, Gur RE, Shinohara RT, Seidlitz J, Almasy L, Andreassen OA, Alexander-Bloch AF. The overlapping genetic architecture of psychiatric disorders and cortical brain structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.05.561040. [PMID: 37873315 PMCID: PMC10592957 DOI: 10.1101/2023.10.05.561040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Both psychiatric vulnerability and cortical structure are shaped by the cumulative effect of common genetic variants across the genome. However, the shared genetic underpinnings between psychiatric disorders and brain structural phenotypes, such as thickness and surface area of the cerebral cortex, remains elusive. In this study, we employed pleiotropy-informed conjunctional false discovery rate analysis to investigate shared loci across genome-wide association scans of regional cortical thickness, surface area, and seven psychiatric disorders in approximately 700,000 individuals of European ancestry. Aggregating regional measures, we identified 50 genetic loci shared between psychiatric disorders and surface area, as well as 26 genetic loci shared with cortical thickness. Risk alleles exhibited bidirectional effects on both cortical thickness and surface area, such that some risk alleles for each disorder increased regional brain size while other risk alleles decreased regional brain size. Due to bidirectional effects, in many cases we observed extensive pleiotropy between an imaging phenotype and a psychiatric disorder even in the absence of a significant genetic correlation between them. The impact of genetic risk for psychiatric disorders on regional brain structure did exhibit a consistent pattern across highly comorbid psychiatric disorders, with 80% of the genetic loci shared across multiple disorders displaying consistent directions of effect. Cortical patterning of genetic overlap revealed a hierarchical genetic architecture, with the association cortex and sensorimotor cortex representing two extremes of shared genetic influence on psychiatric disorders and brain structural variation. Integrating multi-scale functional annotations and transcriptomic profiles, we observed that shared genetic loci were enriched in active genomic regions, converged on neurobiological and metabolic pathways, and showed differential expression in postmortem brain tissue from individuals with psychiatric disorders. Cumulatively, these findings provide a significant advance in our understanding of the overlapping polygenic architecture between psychopathology and cortical brain structure.
Collapse
Affiliation(s)
- Zhiqiang Sha
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | | | - Laura M. Schultz
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Alison Merikangas
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin Y. Sun
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - Russell T. Shinohara
- Penn Statistics in Imaging and Visualization Endeavor (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Dr, Philadelphia, PA 19104, United States
- Center for Biomedical Image Computing and Analytics (CBICA), Department of Radiology, Perelman School of Medicine, United States
| | - Jakob Seidlitz
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ole A. Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Aaron F. Alexander-Bloch
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| |
Collapse
|
13
|
Ma Q, Ruan H, Dai H, Yao WD. USP48/USP31 Is a Nuclear Deubiquitinase that Potently Regulates Synapse Remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558317. [PMID: 37781625 PMCID: PMC10541093 DOI: 10.1101/2023.09.19.558317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Deubiquitinases present locally at synapses regulate synaptic development, function, and plasticity. It remains largely unknown, however, whether deubiquitinases localized outside of the synapse control synapse remodeling. Here we identify ubiquitin specific protease 48 (USP48; formerly USP31) as a nuclear deubiquitinase mediating robust synapse removal. USP48 is expressed primarily during the first postnatal week in the rodent brain and is virtually restricted to nuclei, mediated by a conserved, 13-amino acid nuclear localization signal. When exogenously expressed, USP48, in a deubiquitinase and nuclear localization-dependent manner, induces striking filopodia elaboration, marked spine loss, and significantly reduced synaptic protein clustering in vitro, and erases ~70% of functional synapses in vivo. USP48 interacts with the transcription factor NF-κB, deubiquitinates NF-κB subunit p65 and promotes its stability and activation, and up-regulates NF-κB target genes known to inhibit synaptogenesis. Depleting NF-κB prevents USP48-dependent spine pruning. These findings identify a novel nucleus-enriched deubiquitinase that plays critical roles in synapse remodeling.
Collapse
Affiliation(s)
- Qi Ma
- Departments of Psychiatry and Neuroscience, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Hongyu Ruan
- Departments of Psychiatry and Neuroscience, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Huihui Dai
- Departments of Psychiatry and Neuroscience, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Wei-Dong Yao
- Departments of Psychiatry and Neuroscience, State University of New York, Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
14
|
Fréal A, Jamann N, Ten Bos J, Jansen J, Petersen N, Ligthart T, Hoogenraad CC, Kole MH. Sodium channel endocytosis drives axon initial segment plasticity. SCIENCE ADVANCES 2023; 9:eadf3885. [PMID: 37713493 PMCID: PMC10881073 DOI: 10.1126/sciadv.adf3885] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/15/2023] [Indexed: 09/17/2023]
Abstract
Activity-dependent plasticity of the axon initial segment (AIS) endows neurons with the ability to adapt action potential output to changes in network activity. Action potential initiation at the AIS highly depends on the clustering of voltage-gated sodium channels, but the molecular mechanisms regulating their plasticity remain largely unknown. Here, we developed genetic tools to label endogenous sodium channels and their scaffolding protein, to reveal their nanoscale organization and longitudinally image AIS plasticity in hippocampal neurons in slices and primary cultures. We find that N-methyl-d-aspartate receptor activation causes both long-term synaptic depression and rapid internalization of AIS sodium channels within minutes. The clathrin-mediated endocytosis of sodium channels at the distal AIS increases the threshold for action potential generation. These data reveal a fundamental mechanism for rapid activity-dependent AIS reorganization and suggests that plasticity of intrinsic excitability shares conserved features with synaptic plasticity.
Collapse
Affiliation(s)
- Amélie Fréal
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Nora Jamann
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jolijn Ten Bos
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jacqueline Jansen
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Naomi Petersen
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Thijmen Ligthart
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Casper C. Hoogenraad
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Department of Neuroscience, Genentech Inc, South San Francisco, CA, USA
| | - Maarten H. P. Kole
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
15
|
Murphy KE, Duncan BW, Sperringer JE, Zhang EY, Haberman VA, Wyatt EV, Maness PF. Ankyrin B Promotes Developmental Spine Regulation in the Mouse Prefrontal Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548527. [PMID: 37503187 PMCID: PMC10369899 DOI: 10.1101/2023.07.11.548527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Postnatal regulation of dendritic spine formation and refinement in cortical pyramidal neurons is critical for excitatory/inhibitory balance in neocortical networks. Recent studies have identified a selective spine pruning mechanism in the mouse prefrontal cortex (PFC) mediated by class 3 Semaphorins and the L1-CAM cell adhesion molecules Neuron-glia related CAM (NrCAM), Close Homolog of L1 (CHL1), and L1. L1-CAMs bind Ankyrin B (AnkB), an actin-spectrin adaptor encoded by Ankyrin2 ( ANK2 ), a high confidence gene for autism spectrum disorder (ASD). In a new inducible mouse model (Nex1Cre-ERT2: Ank2 flox : RCE), Ank2 deletion in early postnatal pyramidal neurons increased spine density on apical dendrites in PFC layer 2/3 of homozygous and heterozygous Ank2 -deficient mice. In contrast, Ank2 deletion in adulthood had no effect on spine density. Sema3F-induced spine pruning was impaired in cortical neuron cultures from AnkB-null mice and was rescued by re-expression of the 220 kDa AnkB isoform but not 440 kDa AnkB. AnkB bound to NrCAM at a cytoplasmic domain motif (FIGQY 1231 ), and mutation to FIGQH inhibited binding, impairing Sema3F-induced spine pruning in neuronal cultures. Identification of a novel function for AnkB in dendritic spine regulation provides insight into cortical circuit development, as well as potential molecular deficiencies in ASD.
Collapse
|
16
|
Xiang Y, Li X, Cai M, Cai D. USP9X promotes lipopolysaccharide-stimulated acute lung injury by deubiquitination of NLRP3. Cell Biol Int 2023; 47:394-405. [PMID: 36525374 DOI: 10.1002/cbin.11932] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/31/2022] [Accepted: 09/25/2022] [Indexed: 12/23/2022]
Abstract
Alveolar epithelial cells (AECs) function as a vital defense barrier avoiding the invasion of exogenous agents and preserving the functional and structural integrity of lung tissues, while damage/breakdown of this airway epithelial barrier is frequently associated with the pathogenesis of acute lung injury (ALI). NOD-like receptor family, pyrindomain-containing 3 (NLRP3) inflammasome activation-associated pyroptosis is involved in the development of ALI. Yet, how the activity of NLRP3 inflammasome is regulated in the context of ALI remains unknown. Herein we hypothesized that USP9X, an important deubiquitinase, participates in modulating the activation of NLRP3 inflammasome, thereby affecting the phenotypes in a lipopolysaccharide (LPS)-stimulated AEC model. Human pulmonary AECs were subjected to LPS/adenosine triphosphate (ATP) treatment to induce NLRP3 inflammasome activation and cell pyroptosis. Knockdown and overexpression of USP9X were applied to validate the function of USP9X. Inhibitors of proteinase and protein synthesis, as well as approach of co-immunoprecipitation coupled with Western blot, were utilized to explore the molecular mechanism. LPS/ATP challenge resulted in pronouncedly increased pyroptosis of AECs, activation of NLRP3 inflammasome and release of interleukin (IL)-1β and IL-18 cytokines, while downregulation of USP9X could reverse these alterations. USP9X was found to have marked impact on NLRP3 protein instead of mRNA level. Furthermore, increased ubiquitination of NLRP3 was observed upon downregulating USP9X. Additionally, the inhibitory effect of USP9X downregulation was reversed by NLRP3 overexpression, while the promoting impact of USP9X overexpression was dampened by NLRP3 inhibitor in terms of cell pyroptosis and cytokine secretion. USP9X modulated the activity of NLRP3 inflammasome and pyroptosis of AECs via its deubiquitination function.
Collapse
Affiliation(s)
- Yijin Xiang
- Development Project of Shanghai Peak Disciplines-Integrative Medicine, Department of Integrative Medicine, Zhongshan Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Xiangting Li
- Development Project of Shanghai Peak Disciplines-Integrative Medicine, Department of Integrative Medicine, Zhongshan Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Min Cai
- Development Project of Shanghai Peak Disciplines-Integrative Medicine, Department of Integrative Medicine, Zhongshan Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Dingfang Cai
- Development Project of Shanghai Peak Disciplines-Integrative Medicine, Department of Integrative Medicine, Zhongshan Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Impaired OTUD7A-dependent Ankyrin regulation mediates neuronal dysfunction in mouse and human models of the 15q13.3 microdeletion syndrome. Mol Psychiatry 2023; 28:1747-1769. [PMID: 36604605 DOI: 10.1038/s41380-022-01937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023]
Abstract
Copy number variations (CNVs) are associated with psychiatric and neurodevelopmental disorders (NDDs), and most, including the recurrent 15q13.3 microdeletion disorder, have unknown disease mechanisms. We used a heterozygous 15q13.3 microdeletion mouse model and patient iPSC-derived neurons to reveal developmental defects in neuronal maturation and network activity. To identify the underlying molecular dysfunction, we developed a neuron-specific proximity-labeling proteomics (BioID2) pipeline, combined with patient mutations, to target the 15q13.3 CNV genetic driver OTUD7A. OTUD7A is an emerging independent NDD risk gene with no known function in the brain, but has putative deubiquitinase function. The OTUD7A protein-protein interaction network included synaptic, axonal, and cytoskeletal proteins and was enriched for ASD and epilepsy risk genes (Ank3, Ank2, SPTAN1, SPTBN1). The interactions between OTUD7A and Ankyrin-G (Ank3) and Ankyrin-B (Ank2) were disrupted by an epilepsy-associated OTUD7A L233F variant. Further investigation of Ankyrin-G in mouse and human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed protein instability, increased polyubiquitination, and decreased levels in the axon initial segment, while structured illumination microscopy identified reduced Ankyrin-G nanodomains in dendritic spines. Functional analysis of human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed shared and distinct impairments to axonal growth and intrinsic excitability. Importantly, restoring OTUD7A or Ankyrin-G expression in 15q13.3 microdeletion neurons led to a reversal of abnormalities. These data reveal a critical OTUD7A-Ankyrin pathway in neuronal development, which is impaired in the 15q13.3 microdeletion syndrome, leading to neuronal dysfunction. Furthermore, our study highlights the utility of targeting CNV genes using cell type-specific proteomics to identify shared and unexplored disease mechanisms across NDDs.
Collapse
|
18
|
Matutino Santos P, Pereira Campos G, Nascimento C. Endo-Lysosomal and Autophagy Pathway and Ubiquitin-Proteasome System in Mood Disorders: A Review Article. Neuropsychiatr Dis Treat 2023; 19:133-151. [PMID: 36684613 PMCID: PMC9849791 DOI: 10.2147/ndt.s376380] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/08/2022] [Indexed: 01/15/2023] Open
Abstract
Mood disorders are disabling conditions that cause significant functional impairment. Due to the clinical heterogeneity and complex nature of these disorders, diagnostic and treatment strategies face challenges. The etiology of mood disorders is multifactorial, involving genetic and environmental aspects that are associated with specific biological pathways including inflammation, oxidative stress, and neuroprotection. Alterations in these pathways may reduce the cell's ability to recover from stress conditions occurring during mood episodes. The endo-lysosomal and autophagy pathway (ELAP) and the ubiquitin-proteasome system (UPS) play critical roles in protein homeostasis, impacting neuroplasticity and neurodevelopment. Thus, emerging evidence has suggested a role for these pathways in mental disorders. In the case of neurodegenerative diseases (NDDs), a deeper understanding in the role of ELAP and UPS has been critical to discover new treatment targets. Since it is suggested that NDDs and mood disorders share clinical symptomatology and risk factors, it has been hypothesized that there might be common underlying molecular pathways. Here, we review the importance of the ELAP and UPS for the central nervous system and for mood disorders. Finally, we discuss potential translational strategies for the diagnosis and treatment of major depressive disorder and bipolar disorder associated with these pathways.
Collapse
Affiliation(s)
- Petala Matutino Santos
- Center for Mathematics, Computing and Cognition (CMCC), Federal University of ABC (UFABC), São Paulo, Brazil
| | - Giovanna Pereira Campos
- Center for Mathematics, Computing and Cognition (CMCC), Federal University of ABC (UFABC), São Paulo, Brazil
| | - Camila Nascimento
- Department of Psychiatry, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
19
|
Elu N, Osinalde N, Ramirez J, Presa N, Rodriguez JA, Prieto G, Mayor U. Identification of substrates for human deubiquitinating enzymes (DUBs): An up-to-date review and a case study for neurodevelopmental disorders. Semin Cell Dev Biol 2022; 132:120-131. [PMID: 35042675 DOI: 10.1016/j.semcdb.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 12/15/2022]
Abstract
Similar to the reversal of kinase-mediated protein phosphorylation by phosphatases, deubiquitinating enzymes (DUBs) oppose the action of E3 ubiquitin ligases and reverse the ubiquitination of proteins. A total of 99 human DUBs, classified in 7 families, allow in this way for a precise control of cellular function and homeostasis. Ubiquitination regulates a myriad of cellular processes, and is altered in many pathological conditions. Thus, ubiquitination-regulating enzymes are increasingly regarded as potential candidates for therapeutic intervention. In this context, given the predicted easier pharmacological control of DUBs relative to E3 ligases, a significant effort is now being directed to better understand the processes and substrates regulated by each DUB. Classical studies have identified specific DUB substrate candidates by traditional molecular biology techniques in a case-by-case manner. Lately, single experiments can identify thousands of ubiquitinated proteins at a specific cellular context and narrow down which of those are regulated by a given DUB, thanks to the development of new strategies to isolate and enrich ubiquitinated material and to improvements in mass spectrometry detection capabilities. Here we present an overview of both types of studies, discussing the criteria that, in our view, need to be fulfilled for a protein to be considered as a high-confidence substrate of a given DUB. Applying these criteria, we have manually reviewed the relevant literature currently available in a systematic manner, and identified 650 high-confidence substrates of human DUBs. We make this information easily accessible to the research community through an updated version of the DUBase website (https://ehubio.ehu.eus/dubase/). Finally, in order to illustrate how this information can contribute to a better understanding of the physiopathological role of DUBs, we place a special emphasis on a subset of these enzymes that have been associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nagore Elu
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Nerea Osinalde
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Juanma Ramirez
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Natalia Presa
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Jose Antonio Rodriguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Gorka Prieto
- Department of Communications Engineering, University of the Basque Country (UPV/EHU), Bilbao 48013, Spain
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain; Ikerbasque, Basque Foundation for Science, Bilbao 48013, Spain.
| |
Collapse
|
20
|
A patient with mosaic USP9X gene variant. Eur J Med Genet 2022; 65:104638. [DOI: 10.1016/j.ejmg.2022.104638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/10/2022] [Accepted: 10/01/2022] [Indexed: 11/18/2022]
|
21
|
Ochneva A, Zorkina Y, Abramova O, Pavlova O, Ushakova V, Morozova A, Zubkov E, Pavlov K, Gurina O, Chekhonin V. Protein Misfolding and Aggregation in the Brain: Common Pathogenetic Pathways in Neurodegenerative and Mental Disorders. Int J Mol Sci 2022; 23:14498. [PMID: 36430976 PMCID: PMC9695177 DOI: 10.3390/ijms232214498] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
Mental disorders represent common brain diseases characterized by substantial impairments of social and cognitive functions. The neurobiological causes and mechanisms of psychopathologies still have not been definitively determined. Various forms of brain proteinopathies, which include a disruption of protein conformations and the formation of protein aggregates in brain tissues, may be a possible cause behind the development of psychiatric disorders. Proteinopathies are known to be the main cause of neurodegeneration, but much less attention is given to the role of protein impairments in psychiatric disorders' pathogenesis, such as depression and schizophrenia. For this reason, the aim of this review was to discuss the potential contribution of protein illnesses in the development of psychopathologies. The first part of the review describes the possible mechanisms of disruption to protein folding and aggregation in the cell: endoplasmic reticulum stress, dysfunction of chaperone proteins, altered mitochondrial function, and impaired autophagy processes. The second part of the review addresses the known proteins whose aggregation in brain tissue has been observed in psychiatric disorders (amyloid, tau protein, α-synuclein, DISC-1, disbindin-1, CRMP1, SNAP25, TRIOBP, NPAS3, GluA1, FABP, and ankyrin-G).
Collapse
Affiliation(s)
- Aleksandra Ochneva
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Yana Zorkina
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Abramova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Pavlova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Valeriya Ushakova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
- Department of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Anna Morozova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Eugene Zubkov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Konstantin Pavlov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Gurina
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Vladimir Chekhonin
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- National University of Science and Technology “MISiS”, Leninskiy Avenue 4, 119049 Moscow, Russia
| |
Collapse
|
22
|
Jolly LA, Kumar R, Penzes P, Piper M, Gecz J. The DUB Club: Deubiquitinating Enzymes and Neurodevelopmental Disorders. Biol Psychiatry 2022; 92:614-625. [PMID: 35662507 PMCID: PMC10084722 DOI: 10.1016/j.biopsych.2022.03.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/28/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023]
Abstract
Protein ubiquitination is a widespread, multifunctional, posttranslational protein modification, best known for its ability to direct protein degradation via the ubiquitin proteasome system (UPS). Ubiquitination is also reversible, and the human genome encodes over 90 deubiquitinating enzymes (DUBs), many of which appear to target specific subsets of ubiquitinated proteins. This review focuses on the roles of DUBs in neurodevelopmental disorders (NDDs). We present the current genetic evidence connecting 12 DUBs to a range of NDDs and the functional studies implicating at least 19 additional DUBs as candidate NDD genes. We highlight how the study of DUBs in NDDs offers critical insights into the role of protein degradation during brain development. Because one of the major known functions of a DUB is to antagonize the UPS, loss of function of DUB genes has been shown to culminate in loss of abundance of its protein substrates. The identification and study of NDD DUB substrates in the developing brain is revealing that they regulate networks of proteins that themselves are encoded by NDD genes. We describe the new technologies that are enabling the full resolution of DUB protein networks in the developing brain, with the view that this knowledge can direct the development of new therapeutic paradigms. The fact that the abundance of many NDD proteins is regulated by the UPS presents an exciting opportunity to combat NDDs caused by haploinsufficiency, because the loss of abundance of NDD proteins can be potentially rectified by antagonizing their UPS-based degradation.
Collapse
Affiliation(s)
- Lachlan A Jolly
- University of Adelaide and Robinson Research Institute, Adelaide, South Australia, Australia.
| | - Raman Kumar
- University of Adelaide and Robinson Research Institute, Adelaide, South Australia, Australia
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Michael Piper
- School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Jozef Gecz
- University of Adelaide and Robinson Research Institute, Adelaide, South Australia, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
23
|
Li H, Wei M, Ye T, Liu Y, Qi D, Cheng X. Identification of the molecular subgroups in Alzheimer's disease by transcriptomic data. Front Neurol 2022; 13:901179. [PMID: 36204002 PMCID: PMC9530954 DOI: 10.3389/fneur.2022.901179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAlzheimer's disease (AD) is a heterogeneous pathological disease with genetic background accompanied by aging. This inconsistency is present among molecular subtypes, which has led to diagnostic ambiguity and failure in drug development. We precisely distinguished patients of AD at the transcriptome level.MethodsWe collected 1,240 AD brain tissue samples collected from the GEO dataset. Consensus clustering was used to identify molecular subtypes, and the clinical characteristics were focused on. To reveal transcriptome differences among subgroups, we certificated specific upregulated genes and annotated the biological function. According to RANK METRIC SCORE in GSEA, TOP10 was defined as the hub gene. In addition, the systematic correlation between the hub gene and “A/T/N” was analyzed. Finally, we used external data sets to verify the diagnostic value of hub genes.ResultsWe identified three molecular subtypes of AD from 743 AD samples, among which subtypes I and III had high-risk factors, and subtype II had protective factors. All three subgroups had higher neuritis plaque density, and subgroups I and III had higher clinical dementia scores and neurofibrillary tangles than subgroup II. Our results confirmed a positive association between neurofibrillary tangles and dementia, but not neuritis plaques. Subgroup I genes clustered in viral infection, hypoxia injury, and angiogenesis. Subgroup II showed heterogeneity in synaptic pathology, and we found several essential beneficial synaptic proteins. Due to presenilin one amplification, Subgroup III was a risk subgroup suspected of familial AD, involving abnormal neurogenic signals, glial cell differentiation, and proliferation. Among the three subgroups, the highest combined diagnostic value of the hub genes were 0.95, 0.92, and 0.83, respectively, indicating that the hub genes had sound typing and diagnostic ability.ConclusionThe transcriptome classification of AD cases played out the pathological heterogeneity of different subgroups. It throws daylight on the personalized diagnosis and treatment of AD.
Collapse
Affiliation(s)
- He Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Meiqi Wei
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianyuan Ye
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiduan Liu
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongmei Qi
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaorui Cheng
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Xiaorui Cheng
| |
Collapse
|
24
|
Roles and mechanisms of ankyrin-G in neuropsychiatric disorders. Exp Mol Med 2022; 54:867-877. [PMID: 35794211 PMCID: PMC9356056 DOI: 10.1038/s12276-022-00798-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 12/20/2022] Open
Abstract
Ankyrin proteins act as molecular scaffolds and play an essential role in regulating cellular functions. Recent evidence has implicated the ANK3 gene, encoding ankyrin-G, in bipolar disorder (BD), schizophrenia (SZ), and autism spectrum disorder (ASD). Within neurons, ankyrin-G plays an important role in localizing proteins to the axon initial segment and nodes of Ranvier or to the dendritic shaft and spines. In this review, we describe the expression patterns of ankyrin-G isoforms, which vary according to the stage of brain development, and consider their functional differences. Furthermore, we discuss how posttranslational modifications of ankyrin-G affect its protein expression, interactions, and subcellular localization. Understanding these mechanisms leads us to elucidate potential pathways of pathogenesis in neurodevelopmental and psychiatric disorders, including BD, SZ, and ASD, which are caused by rare pathogenic mutations or changes in the expression levels of ankyrin-G in the brain. Mutations affecting the production, distribution, or function of the ankyrin-G protein may contribute to a variety of different neuropsychiatric disorders. Ankyrin-G is typically observed at the synapses between neurons, and contributes to intercellular adhesion and signaling along with other important functions. Peter Penzes and colleagues at Northwestern University, Chicago, USA, review the biology of this protein and identify potential mechanisms by which ankyrin-G mutations might impair healthy brain development. Mutations in the gene encoding this protein are strongly linked with bipolar disorder, but have also been tentatively connected to autism spectrum disorders and schizophrenia. The authors highlight physiologically important interactions with a diverse array of other brain proteins, which can in turn be modulated by various chemical modifications to ankyrin-G, and conclude that drugs that influence these modifications could have potential therapeutic value.
Collapse
|
25
|
Phenotypes, mechanisms and therapeutics: insights from bipolar disorder GWAS findings. Mol Psychiatry 2022; 27:2927-2939. [PMID: 35351989 DOI: 10.1038/s41380-022-01523-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 12/25/2022]
Abstract
Genome-wide association studies (GWAS) have reported substantial genomic loci significantly associated with clinical risk of bipolar disorder (BD), and studies combining techniques of genetics, neuroscience, neuroimaging, and pharmacology are believed to help tackle clinical problems (e.g., identifying novel therapeutic targets). However, translating findings of psychiatric genetics into biological mechanisms underlying BD pathogenesis remains less successful. Biological impacts of majority of BD GWAS risk loci are obscure, and the involvement of many GWAS risk genes in this illness is yet to be investigated. It is thus necessary to review the progress of applying BD GWAS risk genes in the research and intervention of the disorder. A comprehensive literature search found that a number of such risk genes had been investigated in cellular or animal models, even before they were highlighted in BD GWAS. Intriguingly, manipulation of many BD risk genes (e.g., ANK3, CACNA1C, CACNA1B, HOMER1, KCNB1, MCHR1, NCAN, SHANK2 etc.) resulted in altered murine behaviors largely restoring BD clinical manifestations, including mania-like symptoms such as hyperactivity, anxiolytic-like behavior, as well as antidepressant-like behavior, and these abnormalities could be attenuated by mood stabilizers. In addition to recapitulating phenotypic characteristics of BD, some GWAS risk genes further provided clues for the neurobiology of this illness, such as aberrant activation and functional connectivity of brain areas in the limbic system, and modulated dendritic spine morphogenesis as well as synaptic plasticity and transmission. Therefore, BD GWAS risk genes are undoubtedly pivotal resources for modeling this illness, and might be translational therapeutic targets in the future clinical management of BD. We discuss both promising prospects and cautions in utilizing the bulk of useful resources generated by GWAS studies. Systematic integrations of findings from genetic and neuroscience studies are called for to promote our understanding and intervention of BD.
Collapse
|
26
|
Yoon S, Myczek K, Penzes P. cAMP Signaling-Mediated Phosphorylation of Diacylglycerol Lipase α Regulates Interaction With Ankyrin-G and Dendritic Spine Morphology. Biol Psychiatry 2021; 90:263-274. [PMID: 34099188 PMCID: PMC8384113 DOI: 10.1016/j.biopsych.2021.03.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Diacylglycerol lipase α (DAGLα), a major biosynthetic enzyme for endogenous cannabinoid signaling, has emerged as a risk gene in multiple psychiatric disorders. However, its role in the regulation of dendritic spine plasticity is unclear. METHODS DAGLα wild-type or point mutants were overexpressed in primary cortical neurons or human embryonic kidney 293T cells. The effects of mutated variants on interaction, dendritic spine morphology, and dynamics were examined by proximity ligation assay or fluorescence recovery after photobleaching. Behavioral tests and immunohistochemistry were performed with ankyrin-G conditional knockout and wild-type male mice. RESULTS DAGLα modulated dendritic spine size and density, but the effects of changes in its protein level versus enzymatic activity were different, implicating either a 2-arachidonoylglycerol (2-AG)-dependent or -independent mechanism. The 2-AG-independent effects were mediated by the interaction of DAGLα with ankyrin-G, a multifunctional scaffold protein implicated in psychiatric disorders. Using superresolution microscopy, we observed that they colocalized in distinct nanodomains, which correlated with spine size. In situ proximity ligation assay combined with structured illumination microscopy revealed that DAGLα phosphorylation upon forskolin treatment enhanced the interaction with ankyrin-G in spines, leading to increased spine size and decreased DAGLα surface diffusion. Ankyrin-G conditional knockout mice showed significantly decreased DAGLα-positive neurons in the forebrain. In mice, ankyrin-G was required for forskolin-dependent reversal of depression-related behavior. CONCLUSIONS Taken together, ANK3 and DAGLA, both neuropsychiatric disorder genes, interact in a complex to regulate spine morphology. These data reveal novel synaptic signaling mechanisms and potential therapeutic avenues.
Collapse
Affiliation(s)
- Sehyoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kristoffer Myczek
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Psychiatry and Behavioral Sciences, and Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
27
|
Historical perspective and progress on protein ubiquitination at glutamatergic synapses. Neuropharmacology 2021; 196:108690. [PMID: 34197891 DOI: 10.1016/j.neuropharm.2021.108690] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/07/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
Transcription-translation coupling leads to the production of proteins that are key for controlling essential neuronal processes that include neuronal development and changes in synaptic strength. Although these events have been a prevailing theme in neuroscience, the regulation of proteins via posttranslational signaling pathways are equally relevant for these neuronal processes. Ubiquitin is one type of posttranslational modification that covalently attaches to its targets/substrates. Ubiquitination of proteins play a key role in multiple signaling pathways, the predominant being removal of its substrates by a large molecular machine called the proteasome. Here, I review 40 years of progress on ubiquitination in the nervous system at glutamatergic synapses focusing on axon pathfinding, synapse formation, presynaptic release, dendritic spine formation, and regulation of postsynaptic glutamate receptors. Finally, I elucidate emerging themes in ubiquitin biology that may challenge our current understanding of ubiquitin signaling in the nervous system.
Collapse
|
28
|
Zaccard CR, Kirchenbuechler D, Yoon S, Arvanitis C, Penzes P. Protocol for live enhanced resolution confocal imaging of dendritic spinule dynamics in primary mouse cortical neuron culture. STAR Protoc 2021; 2:100427. [PMID: 33899014 PMCID: PMC8056271 DOI: 10.1016/j.xpro.2021.100427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Dendritic spinules are fine membranous protrusions of neuronal spines that play a role in synaptic plasticity, but their nanoscale requires resolution beyond conventional confocal microscopy, hindering live studies. Here, we describe how to track individual spinules in live dissociated cortical pyramidal neurons utilizing fluorescence labeling, optimized confocal imaging parameters, and post-acquisition iterative 3D deconvolution, employing NIS Elements software. This approach enables investigations of spinule structural dynamics and function without using super-resolution microscopy, which involves special fluorophores and/or high laser power. For complete details on the use and execution of this protocol, please refer to Zaccard et al. (2020).
Collapse
Affiliation(s)
- Colleen R. Zaccard
- Department of Physiology, Northwestern University, Chicago, IL 60611, USA
| | | | - Sehyoun Yoon
- Department of Physiology, Northwestern University, Chicago, IL 60611, USA
| | - Constadina Arvanitis
- Center for Advanced Microscopy, Northwestern University, Chicago, IL 60611, USA
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
29
|
Yoon S, Parnell E, Penzes P. TGF-β-Induced Phosphorylation of Usp9X Stabilizes Ankyrin-G and Regulates Dendritic Spine Development and Maintenance. Cell Rep 2021; 31:107685. [PMID: 32460012 PMCID: PMC7324065 DOI: 10.1016/j.celrep.2020.107685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/02/2020] [Accepted: 05/04/2020] [Indexed: 12/30/2022] Open
Abstract
Signaling by the cytokine transforming growth factor β (TGF-β) has been implicated in a multitude of biological functions; however, TGF-β signaling, particularly in the CNS, remains largely unexplored. ANK3 variants (encoding ankyrin-G) are associated with bipolar disorder, intellectual disability, and autism spectrum disorder, while mutations in USP9X, which encodes a deubiquitinase, are associated with X-linked intellectual disability and autism in humans. Here, we show that TGF-β signaling promotes Usp9X phosphorylation, which enhances its interaction with ankyrin-G and stabilizes ankyrin-G in spines, leading to spine enlargement. Using in situ proximity ligation combined with structured illumination superresolution microscopy, we characterize the postsynaptic spatial organization of phosphorylation-dependent regulation of Usp9X/ankyrin-G interactions in dendrites and its quantitative relationship with spine morphology and number. These data reveal a cytokine-mediated mechanism regulating protein stability in spines and suggest a role for deubiquitination and TGF-β signaling in neurodevelopmental disorder pathogenesis and treatment. Yoon et al. show that phosphorylation of a deubiquitinating enzyme by a cytokine enhances the stabilization of synaptic scaffolding protein during dendritic spine development, and its alterations result in deficient synaptic structural maintenance, with relevance for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sehyoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Euan Parnell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Northwestern University, Center for Autism and Neurodevelopment, Chicago, IL 60611, USA.
| |
Collapse
|
30
|
Alò R, Olivito I, Fazzari G, Zizza M, Di Vito A, Avolio E, Mandalà M, Bruno R, Barni T, Canonaco M, Facciolo RM. Correlation of distinct behaviors to the modified expression of cerebral Shank1,3 and BDNF in two autistic animal models. Behav Brain Res 2021; 404:113165. [PMID: 33577886 DOI: 10.1016/j.bbr.2021.113165] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/27/2020] [Accepted: 02/02/2021] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder featuring altered neuronal circuitry and consequently impaired social interactions, restrictive interests plus repetitive stereotypic activities. In the present study, differentiated behaviors of valproic (VPA) and propionic (PPA) acid-mediated autism rats were correlated to cerebral scaffolding proteins (Shank1,3) and BDNF expression variations. Sprague-Dawley offspring that received VPA during pregnancy displayed a notably diminished permanence (-78 %, p < 0.01) in the light chamber of light dark (LD) test, reduced exploratory tasks, i.e. grooming (-90 %) and rearing (-65 %). Moreover, they executed extremely greater climbing intervals (+300 %, p < 0.001) in novel cage (NC) test, plus exhibited an extremely reduced (-331 %) discrimination index in novel object recognition (NOR) test when compared to controls. PPA-treated postnatal days (PND) 12-16 rats also displayed anxiety-like behaviors, although in a less evident manner, as indicated by a moderate time (+55 %; p < 0.05) spent in dark chamber along with notable and moderate decreases in digging (-78 %) plus grooming (-52 %), respectively. Contextually, VPA- more than PPA supplied opposite Shank1,3 expression changes in cerebellum (CB; -62 %; +78 %), dorsomedial prefrontal cortex (DM-PFC; +95 % -76 %), respectively, while resulting extremely upregulated in hippocampus (HIP; +125 % - +155 %). Even BDNF resulted to be substantially and notably diminished in HIP (-85 %) and DM-PFC (-72 %), respectively, of VPA rats while it was only moderately reduced (-35 % to -45 %) in these same areas of PPA rats. The early altered brain-specific expression levels accounting for different behavioral performances may provide useful diagnostic indications and constitute valuable therapeutic strategies for autistic patients.
Collapse
Affiliation(s)
- Raffaella Alò
- Comparative Anatomy & Cytology, Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy
| | - Ilaria Olivito
- Comparative Anatomy & Cytology, Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy
| | - Gilda Fazzari
- Comparative Anatomy & Cytology, Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy
| | - Merylin Zizza
- Comparative Anatomy & Cytology, Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy
| | - Anna Di Vito
- Dept. of Clinical and Experimental Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Ennio Avolio
- Comparative Anatomy & Cytology, Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy; Health Center srl, Biomedical and Nutritional Center, via Sabotino 66, 87100 Cosenza, Italy
| | - Maurizio Mandalà
- Vascular Physiology Lab., Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy
| | - Rosalinda Bruno
- Dept. of Pharmacy and Science of Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende-Cosenza, Italy
| | - Tullio Barni
- Dept. of Clinical and Experimental Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Marcello Canonaco
- Comparative Anatomy & Cytology, Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy.
| | - Rosa Maria Facciolo
- Comparative Anatomy & Cytology, Dept. of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende-Cosenza, Italy
| |
Collapse
|
31
|
Basar MA, Beck DB, Werner A. Deubiquitylases in developmental ubiquitin signaling and congenital diseases. Cell Death Differ 2021; 28:538-556. [PMID: 33335288 PMCID: PMC7862630 DOI: 10.1038/s41418-020-00697-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Metazoan development from a one-cell zygote to a fully formed organism requires complex cellular differentiation and communication pathways. To coordinate these processes, embryos frequently encode signaling information with the small protein modifier ubiquitin, which is typically attached to lysine residues within substrates. During ubiquitin signaling, a three-step enzymatic cascade modifies specific substrates with topologically unique ubiquitin modifications, which mediate changes in the substrate's stability, activity, localization, or interacting proteins. Ubiquitin signaling is critically regulated by deubiquitylases (DUBs), a class of ~100 human enzymes that oppose the conjugation of ubiquitin. DUBs control many essential cellular functions and various aspects of human physiology and development. Recent genetic studies have identified mutations in several DUBs that cause developmental disorders. Here we review principles controlling DUB activity and substrate recruitment that allow these enzymes to regulate ubiquitin signaling during development. We summarize key mechanisms of how DUBs control embryonic and postnatal differentiation processes, highlight developmental disorders that are caused by mutations in particular DUB members, and describe our current understanding of how these mutations disrupt development. Finally, we discuss how emerging tools from human disease genetics will enable the identification and study of novel congenital disease-causing DUBs.
Collapse
Affiliation(s)
- Mohammed A Basar
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David B Beck
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
- Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Achim Werner
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
32
|
Deubiquitylating enzymes in neuronal health and disease. Cell Death Dis 2021; 12:120. [PMID: 33483467 PMCID: PMC7822931 DOI: 10.1038/s41419-020-03361-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022]
Abstract
Ubiquitylation and deubiquitylation play a pivotal role in protein homeostasis (proteostasis). Proteostasis shapes the proteome landscape in the human brain and its impairment is linked to neurodevelopmental and neurodegenerative disorders. Here we discuss the emerging roles of deubiquitylating enzymes in neuronal function and survival. We provide an updated perspective on the genetics, physiology, structure, and function of deubiquitylases in neuronal health and disease. ![]()
Collapse
|
33
|
Structural Insights into Ankyrin Repeat-Containing Proteins and Their Influence in Ubiquitylation. Int J Mol Sci 2021; 22:ijms22020609. [PMID: 33435370 PMCID: PMC7826745 DOI: 10.3390/ijms22020609] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Ankyrin repeat (AR) domains are considered the most abundant repeat motif found in eukaryotic proteins. AR domains are predominantly known to mediate specific protein-protein interactions (PPIs) without necessarily recognizing specific primary sequences, nor requiring strict conformity within its own primary sequence. This promiscuity allows for one AR domain to recognize and bind to a variety of intracellular substrates, suggesting that AR-containing proteins may be involved in a wide array of functions. Many AR-containing proteins serve a critical role in biological processes including the ubiquitylation signaling pathway (USP). There is also strong evidence that AR-containing protein malfunction are associated with several neurological diseases and disorders. In this review, the structure and mechanism of key AR-containing proteins are discussed to suggest and/or identify how each protein utilizes their AR domains to support ubiquitylation and the cascading pathways that follow upon substrate modification.
Collapse
|
34
|
Zhang C, Xiao X, Li T, Li M. Translational genomics and beyond in bipolar disorder. Mol Psychiatry 2021; 26:186-202. [PMID: 32424235 DOI: 10.1038/s41380-020-0782-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 02/08/2023]
Abstract
Genome-wide association studies (GWAS) have revealed multiple genomic loci conferring risk of bipolar disorder (BD), providing hints for its underlying pathobiology. However, there are still remaining questions to answer. For example, discordance exists between BD heritability estimated with earlier epidemiological evidence and that calculated based on common GWAS variations. Where is the "missing heritability"? How can we explain the biology of the disease based on genetic findings? In this review, we summarize the accomplishments and limitations of current BD GWAS, and discuss potential reasons for the "missing heritability." In addition, progresses of research for the biological mechanisms underlying BD genetic risk using brain tissues, reprogrammed cells, and model animals are reviewed. While our knowledge of BD genetic basis is significantly promoted by these efforts, the complexities of gene regulation in the genome, the spatial-temporal heterogeneity during brain development, and the limitations of different experimental models should always be considered. Notably, several genes have been widely studied given their relatively well-characterized involvement in BD (e.g., CACAN1C and ANK3), and findings of these genes are summarized to both outline possible biological mechanisms of BD and describe examples of translating GWAS discoveries into the pathophysiology.
Collapse
Affiliation(s)
- Chen Zhang
- Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Tao Li
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China. .,West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
35
|
Yoon S, Piguel NH, Khalatyan N, Dionisio LE, Savas JN, Penzes P. Homer1 promotes dendritic spine growth through ankyrin-G and its loss reshapes the synaptic proteome. Mol Psychiatry 2021; 26:1775-1789. [PMID: 33398084 PMCID: PMC8254828 DOI: 10.1038/s41380-020-00991-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 11/24/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Homer1 is a synaptic scaffold protein that regulates glutamatergic synapses and spine morphogenesis. HOMER1 knockout (KO) mice show behavioral abnormalities related to psychiatric disorders, and HOMER1 has been associated with psychiatric disorders such as addiction, autism disorder (ASD), schizophrenia (SZ), and depression. However, the mechanisms by which it promotes spine stability and its global function in maintaining the synaptic proteome has not yet been fully investigated. Here, we used computational approaches to identify global functions for proteins containing the Homer1-interacting PPXXF motif within the postsynaptic compartment. Ankyrin-G was one of the most topologically important nodes in the postsynaptic peripheral membrane subnetwork, and we show that one of the PPXXF motifs, present in the postsynaptically-enriched 190 kDa isoform of ankyrin-G (ankyrin-G 190), is recognized by the EVH1 domain of Homer1. We use proximity ligation combined with super-resolution microscopy to map the interaction of ankyrin-G and Homer1 to distinct nanodomains within the spine head and correlate them with spine head size. This interaction motif is critical for ankyrin-G 190's ability to increase spine head size, and for the maintenance of a stable ankyrin-G pool in spines. Intriguingly, lack of Homer1 significantly upregulated the abundance of ankyrin-G, but downregulated Shank3 in cortical crude plasma membrane fractions. In addition, proteomic analysis of the cortex in HOMER1 KO and wild-type (WT) mice revealed a global reshaping of the postsynaptic proteome, surprisingly characterized by extensive upregulation of synaptic proteins. Taken together, we show that Homer1 and its protein interaction motif have broad global functions within synaptic protein-protein interaction networks. Enrichment of disease risk factors within these networks has important implications for neurodevelopmental disorders including bipolar disorder, ASD, and SZ.
Collapse
Affiliation(s)
- Sehyoun Yoon
- grid.16753.360000 0001 2299 3507Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Nicolas H. Piguel
- grid.16753.360000 0001 2299 3507Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Natalia Khalatyan
- grid.16753.360000 0001 2299 3507Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Leonardo E. Dionisio
- grid.16753.360000 0001 2299 3507Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA ,grid.19006.3e0000 0000 9632 6718Present Address: Neuroscience Interdepartmental Program, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Jeffrey N. Savas
- grid.16753.360000 0001 2299 3507Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Northwestern University, Center for Autism and Neurodevelopment, Chicago, IL, 60611, USA.
| |
Collapse
|
36
|
Jolly LA, Parnell E, Gardner AE, Corbett MA, Pérez-Jurado LA, Shaw M, Lesca G, Keegan C, Schneider MC, Griffin E, Maier F, Kiss C, Guerin A, Crosby K, Rosenbaum K, Tanpaiboon P, Whalen S, Keren B, McCarrier J, Basel D, Sadedin S, White SM, Delatycki MB, Kleefstra T, Küry S, Brusco A, Sukarova-Angelovska E, Trajkova S, Yoon S, Wood SA, Piper M, Penzes P, Gecz J. Missense variant contribution to USP9X-female syndrome. NPJ Genom Med 2020; 5:53. [PMID: 33298948 PMCID: PMC7725775 DOI: 10.1038/s41525-020-00162-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023] Open
Abstract
USP9X is an X-chromosome gene that escapes X-inactivation. Loss or compromised function of USP9X leads to neurodevelopmental disorders in males and females. While males are impacted primarily by hemizygous partial loss-of-function missense variants, in females de novo heterozygous complete loss-of-function mutations predominate, and give rise to the clinically recognisable USP9X-female syndrome. Here we provide evidence of the contribution of USP9X missense and small in-frame deletion variants in USP9X-female syndrome also. We scrutinise the pathogenicity of eleven such variants, ten of which were novel. Combined application of variant prediction algorithms, protein structure modelling, and assessment under clinically relevant guidelines universally support their pathogenicity. The core phenotype of this cohort overlapped with previous descriptions of USP9X-female syndrome, but exposed heightened variability. Aggregate phenotypic information of 35 currently known females with predicted pathogenic variation in USP9X reaffirms the clinically recognisable USP9X-female syndrome, and highlights major differences when compared to USP9X-male associated neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lachlan A Jolly
- University of Adelaide and Robinson Research Institute, Adelaide, SA, 5005, Australia.
| | - Euan Parnell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Il, USA
| | - Alison E Gardner
- University of Adelaide and Robinson Research Institute, Adelaide, SA, 5005, Australia
| | - Mark A Corbett
- University of Adelaide and Robinson Research Institute, Adelaide, SA, 5005, Australia
| | - Luis A Pérez-Jurado
- University of Adelaide and Robinson Research Institute, Adelaide, SA, 5005, Australia
- Women's and Children's Hospital, Adelaide, SA, 5006, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
- Hospital del Mar Research Institute (IMIM), Network Research Centre for Rare Diseases (CIBERER) and Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Marie Shaw
- University of Adelaide and Robinson Research Institute, Adelaide, SA, 5005, Australia
| | - Gaetan Lesca
- Institut Neuromyogène, métabolisme énergétique et développement durable, CNRS UMR 5310, INSERM U1217, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Service de Génétique, Hospices Civils de Lyon, Lyon, France
| | - Catherine Keegan
- Division of Genetics, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Michael C Schneider
- Section of Neurology, Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Emily Griffin
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Felicitas Maier
- Dr. von Hauner Children's Hospital, LMU - Ludwig-Maximilians-Universität Munich, University of Munich Medical Center, Munich, Germany
| | - Courtney Kiss
- Kingston Health Sciences Centre, Kingston, ON, K7L 2V7, Canada
| | - Andrea Guerin
- Division of Medical Genetics, Department of Pediatrics, Kingston General Hospital, Kingston, ON, Canada
| | - Kathleen Crosby
- Division of Genetics and Metabolism, Children's National Hospital, Washington, DC, USA
| | - Kenneth Rosenbaum
- Division of Genetics and Metabolism, Children's National Hospital, Washington, DC, USA
| | - Pranoot Tanpaiboon
- Division of Genetics and Metabolism, Children's National Hospital, Washington, DC, USA
| | - Sandra Whalen
- Unité Fonctionnelle de génétique clinique, Hôpital Armand Trousseau, Assistance publique-Hôpitaux de Paris, Centre de Référence Maladies Rares des anomalies du développement et syndromes malformatifs, Paris, France
| | - Boris Keren
- Hôpital de la Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Julie McCarrier
- Division of Genetics, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Donald Basel
- Division of Genetics, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Simon Sadedin
- Victorian Clinical Genetics Service, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Susan M White
- Victorian Clinical Genetics Service, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Martin B Delatycki
- Victorian Clinical Genetics Service, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Tjitske Kleefstra
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6500, HB, the Netherlands
| | - Sébastien Küry
- Service de Génétique Médicale, CHU Nantes, 44093, Nantes, France
- l'Institut du Thorax, INSERM, CNRS, UNIV Nantes, 44007, Nantes, France
| | - Alfredo Brusco
- Department of Medical Sciences, University of Turin, Torino, Italy
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Elena Sukarova-Angelovska
- Department of Endocronology and Genetics, University Clinic for Children's Diseases, Medical Faculty, University Sv. Kiril i Metodij, Skopje, Republic of Macedonia
| | - Slavica Trajkova
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - Sehoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Il, USA
| | - Stephen A Wood
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
| | - Michael Piper
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Il, USA
| | - Jozef Gecz
- University of Adelaide and Robinson Research Institute, Adelaide, SA, 5005, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
| |
Collapse
|
37
|
Wigerius M, Quinn D, Fawcett JP. Emerging roles for angiomotin in the nervous system. Sci Signal 2020; 13:13/655/eabc0635. [PMID: 33109746 DOI: 10.1126/scisignal.abc0635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Angiomotins are a family of molecular scaffolding proteins that function to organize contact points (called tight junctions in vertebrates) between adjacent cells. Some angiomotin isoforms bind to the actin cytoskeleton and are part of signaling pathways that influence cell morphology and migration. Others cooperate with components of the Hippo signaling pathway and the associated networks to control organ growth. The 130-kDa isoform, AMOT-p130, has critical roles in neural stem cell differentiation, dendritic patterning, and synaptic maturation-attributes that are essential for normal brain development and are consistent with its association with autism. Here, we review and discuss the evidence that supports a role for AMOT-p130 in neuronal development in the central nervous system.
Collapse
Affiliation(s)
- Michael Wigerius
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | - Dylan Quinn
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - James P Fawcett
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada. .,Department of Surgery, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
38
|
Chen D, Ren K, Liu H, Mao H, Li Z, Mo H, Xie S, Shi Y, Chen Q, Wang W. A Whole-Brain Cell-Type-Specific Sparse Neuron Labeling Method and Its Application in a Shank3 Autistic Mouse Model. Front Cell Neurosci 2020; 14:145. [PMID: 32581718 PMCID: PMC7291601 DOI: 10.3389/fncel.2020.00145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Single neurons, as the basic unit of the brain, consist of a cell body and processes, including dendrites and axons. Even neurons of the same type show various subtle process characteristics to fit into the diverse neural circuits. Different cell types of neurons form complicated circuits in the brain. Therefore, detailed neuronal morphology is required to understand normal neuronal function and pathological mechanisms, such as those that occur in autism. Here, we developed a strategy to sparsely label the same type of neurons throughout the whole brain and tested its application in an autistic animal model—Shank3 knockout (KO) mice. To achieve this, we designed an adeno-associated virus (AAV) that expresses Cre recombinase-dependent regular and membrane-targeted enhanced green fluorescent protein (EGFP) under a human synapsin 1 promoter and verified it in several Cre transgenic mice. We could sparsely label the projection neurons in multiple brain areas by retro-ocular injection of the virus into CaMKIIα-Cre mice. Then, we analyzed the morphology of the projection neurons in Shank3 KO mice with this method. We found differential dendritic complexity and dendritic spine changes in projection neurons in Shank3 KO mice crossed with CaMKIIα-Cre mice compared with littermate control mice in the striatum, cortex, and hippocampus. By combining this method with various Cre mouse lines crossed with mouse models of disease, we can screen the morphological traits of distinct types of neurons throughout the whole brain that will help us to understand the exact role of the specific cell types of neurons not only in autism spectrum disorder (ASD) mouse models but also in other psychiatric disorder mouse models.
Collapse
Affiliation(s)
- Di Chen
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Keke Ren
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Haiying Liu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Honghui Mao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zongyan Li
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huiming Mo
- Department of Physiology, Medical College of Yan'an University, Yan'an, China
| | - Shengjun Xie
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yiwu Shi
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Chen
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|