1
|
Alves VC, Carro E, Figueiro-Silva J. Unveiling DNA methylation in Alzheimer's disease: a review of array-based human brain studies. Neural Regen Res 2024; 19:2365-2376. [PMID: 38526273 PMCID: PMC11090417 DOI: 10.4103/1673-5374.393106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/05/2023] [Indexed: 03/26/2024] Open
Abstract
The intricacies of Alzheimer's disease pathogenesis are being increasingly illuminated by the exploration of epigenetic mechanisms, particularly DNA methylation. This review comprehensively surveys recent human-centered studies that investigate whole genome DNA methylation in Alzheimer's disease neuropathology. The examination of various brain regions reveals distinctive DNA methylation patterns that associate with the Braak stage and Alzheimer's disease progression. The entorhinal cortex emerges as a focal point due to its early histological alterations and subsequent impact on downstream regions like the hippocampus. Notably, ANK1 hypermethylation, a protein implicated in neurofibrillary tangle formation, was recurrently identified in the entorhinal cortex. Further, the middle temporal gyrus and prefrontal cortex were shown to exhibit significant hypermethylation of genes like HOXA3, RHBDF2, and MCF2L, potentially influencing neuroinflammatory processes. The complex role of BIN1 in late-onset Alzheimer's disease is underscored by its association with altered methylation patterns. Despite the disparities across studies, these findings highlight the intricate interplay between epigenetic modifications and Alzheimer's disease pathology. Future research efforts should address methodological variations, incorporate diverse cohorts, and consider environmental factors to unravel the nuanced epigenetic landscape underlying Alzheimer's disease progression.
Collapse
Affiliation(s)
- Victoria Cunha Alves
- Neurodegenerative Diseases Group, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
- Network Center for Biomedical Research, Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
- Neurotraumatology and Subarachnoid Hemorrhage Group, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Eva Carro
- Network Center for Biomedical Research, Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research Into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Joana Figueiro-Silva
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Escobedo G, Wu Y, Ogawa Y, Ding X, Rasband MN. An evolutionarily conserved AnkyrinG-dependent motif clusters axonal K2P K+ channels. J Cell Biol 2024; 223:e202401140. [PMID: 39078369 PMCID: PMC11289519 DOI: 10.1083/jcb.202401140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
The evolution of ion channel clustering at nodes of Ranvier enabled the development of complex vertebrate nervous systems. At mammalian nodes, the K+ leak channels TRAAK and TREK-1 underlie membrane repolarization. Despite the molecular similarities between nodes and the axon initial segment (AIS), TRAAK and TREK-1 are reportedly node-specific, suggesting a unique clustering mechanism. However, we show that TRAAK and TREK-1 are enriched at both nodes and AIS through a common mechanism. We identified a motif near the C-terminus of TRAAK that is necessary and sufficient for its clustering. The motif first evolved among cartilaginous fish. Using AnkyrinG (AnkG) conditional knockout mice, CRISPR/Cas9-mediated disruption of AnkG, co-immunoprecipitation, and surface recruitment assays, we show that TRAAK forms a complex with AnkG and that AnkG is necessary for TRAAK's AIS and nodal clustering. In contrast, TREK-1's clustering requires TRAAK. Our results expand the repertoire of AIS and nodal ion channel clustering mechanisms and emphasize AnkG's central role in assembling excitable domains.
Collapse
Affiliation(s)
- Gabriel Escobedo
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yu Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yuki Ogawa
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
3
|
Huang J, Sun C, Zhu Q, Wu G, Cao Y, Shi J, He S, Jiang L, Liao J, Li L, Zhong C, Lu Y. Phenotyping of FGF12A V52H mutation in mouse implies a complex FGF12 network. Neurobiol Dis 2024; 200:106637. [PMID: 39142611 DOI: 10.1016/j.nbd.2024.106637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/26/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024] Open
Abstract
Pathogenic missense mutation of the FGF12 gene is responsible for a variable disease phenotypic spectrum. Disease-specific therapies require precise dissection of the relationship between different mutations and phenotypes. The lack of a proper animal model hinders the investigation of related diseases, such as early-onset epileptic encephalopathy. Here, an FGF12AV52H mouse model was generated using CRISPR/Cas9 technology, which altered the A isoform without affecting the B isoform. The FGF12AV52H mice exhibited seizure susceptibility, while no spontaneous seizures were observed. The increased excitability in dorsal hippocampal CA3 neurons was confirmed by patch-clamp recordings. Furthermore, immunostaining showed that the balance of excitatory/inhibitory neurons in the hippocampus of the FGF12AV52H mice was perturbed. The increases in inhibitory SOM+ neurons and excitatory CaMKII+ neurons were heterogeneous. Moreover, the locomotion, anxiety levels, risk assessment behavior, social behavior, and cognition of the FGF12AV52H mice were investigated by elevated plus maze, open field, three-chamber sociability, and novel object tests, respectively. Cognition deficit, impaired risk assessment, and social behavior with normal social indexes were observed, implying complex consequences of V52H FGF12A in mice. Together, these data suggest that the function of FGF12A in neurons can be immediate or long-term and involves modulation of ion channels and the differentiation and maturation of neurons. The FGF12AV52H mouse model increases the understanding of the function of FGF12A, and it is of great importance for revealing the complex network of the FGF12 gene in physiological and pathological processes.
Collapse
Affiliation(s)
- Jianyu Huang
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Chongyang Sun
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhu
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Ge Wu
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Cao
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiarui Shi
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyu He
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Luyao Jiang
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jianxiang Liao
- Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Lin Li
- Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen 518038, China.
| | - Cheng Zhong
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Yi Lu
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
4
|
Ma D, Gu C. Discovering functional interactions among schizophrenia-risk genes by combining behavioral genetics with cell biology. Neurosci Biobehav Rev 2024; 167:105897. [PMID: 39278606 DOI: 10.1016/j.neubiorev.2024.105897] [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: 06/15/2024] [Revised: 08/26/2024] [Accepted: 09/13/2024] [Indexed: 09/18/2024]
Abstract
Despite much progress in identifying risk genes for polygenic brain disorders, their core pathogenic mechanisms remain poorly understood. In particular, functions of many proteins encoded by schizophrenia risk genes appear diverse and unrelated, complicating the efforts to establish the causal relationship between genes and behavior. Using various mouse lines, recent studies indicate that alterations of parvalbumin-positive (PV+) GABAergic interneurons can lead to schizophrenia-like behavior. PV+ interneurons display fast spiking and contribute to excitation-inhibition balance and network oscillations via feedback and feedforward inhibition. Here, we first summarize different lines of genetically modified mice that display motor, cognitive, emotional, and social impairments used to model schizophrenia and related mental disorders. We highlight ten genes, encoding either a nuclear, cytosolic, or membrane protein. Next, we discuss their functional relationship in regulating fast spiking and other aspects of PV+ interneurons and in the context of other domains of schizophrenia. Future investigations combining behavioral genetics and cell biology should elucidate functional relationships among risk genes to identify the core pathogenic mechanisms underlying polygenic brain disorders.
Collapse
Affiliation(s)
- Di Ma
- Ohio State Biochemistry Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Chen Gu
- Ohio State Biochemistry Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
5
|
Stankewich MC, Peters LL, Morrow JS. The loss of βΙ spectrin alters synaptic size and composition in the ja/ja mouse. Front Neurosci 2024; 18:1415115. [PMID: 39165342 PMCID: PMC11333264 DOI: 10.3389/fnins.2024.1415115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024] Open
Abstract
Introduction Deletion or mutation of members of the spectrin gene family contributes to many neurologic and neuropsychiatric disorders. While each spectrinopathy may generate distinct neuropathology, the study of βΙ spectrin's role (Sptb) in the brain has been hampered by the hematologic consequences of its loss. Methods Jaundiced mice (ja/ja) that lack βΙ spectrin suffer a rapidly fatal hemolytic anemia. We have used exchange transfusion of newborn ja/ja mice to blunt their hemolytic pathology, enabling an examination of βΙ spectrin deficiency in the mature mouse brain by ultrastructural and biochemical analysis. Results βΙ spectrin is widely utilized throughout the brain as the βΙΣ2 isoform; it appears by postnatal day 8, and concentrates in the CA1,3 region of the hippocampus, dentate gyrus, cerebellar granule layer, cortical layer 2, medial habenula, and ventral thalamus. It is present in a subset of dendrites and absent in white matter. Without βΙ spectrin there is a 20% reduction in postsynaptic density size in the granule layer of the cerebellum, a selective loss of ankyrinR in cerebellar granule neurons, and a reduction in the level of the postsynaptic adhesion molecule NCAM. While we find no substitution of another spectrin for βΙ at dendrites or synapses, there is curiously enhanced βΙV spectrin expression in the ja/ja brain. Discussion βΙΣ2 spectrin appears to be essential for refining postsynaptic structures through interactions with ankyrinR and NCAM. We speculate that it may play additional roles yet to be discovered.
Collapse
Affiliation(s)
- Michael C. Stankewich
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | | | - Jon S. Morrow
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
- Department Molecular Cellular and Developmental Biology, Yale University, New Haven, CT, United States
| |
Collapse
|
6
|
Argall AD, Sucharski-Argall HC, Comisford LG, Jurs SJ, Seminetta JT, Wallace MJ, Crawford CA, Takenaka SS, Han M, El Refaey M, Hund TJ, Mohler PJ, Koenig SN. Novel Identification of Ankyrin-R in Cardiac Fibroblasts and a Potential Role in Heart Failure. Int J Mol Sci 2024; 25:8403. [PMID: 39125973 PMCID: PMC11313496 DOI: 10.3390/ijms25158403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Altered ankyrin-R (AnkR; encoded by ANK1) expression is associated with diastolic function, left ventricular remodeling, and heart failure with preserved ejection fraction (HFpEF). First identified in erythrocytes, the role of AnkR in other tissues, particularly the heart, is less studied. Here, we identified the expression of both canonical and small isoforms of AnkR in the mouse myocardium. We demonstrate that cardiac myocytes primarily express small AnkR (sAnkR), whereas cardiac fibroblasts predominantly express canonical AnkR. As canonical AnkR expression in cardiac fibroblasts is unstudied, we focused on expression and localization in these cells. AnkR is expressed in both the perinuclear and cytoplasmic regions of fibroblasts with considerable overlap with the trans-Golgi network protein 38, TGN38, suggesting a potential role in trafficking. To study the role of AnkR in fibroblasts, we generated mice lacking AnkR in activated fibroblasts (Ank1-ifKO mice). Notably, Ank1-ifKO mice fibroblasts displayed reduced collagen compaction, supportive of a novel role of AnkR in normal fibroblast function. At the whole animal level, in response to a heart failure model, Ank1-ifKO mice displayed an increase in fibrosis and T-wave inversion compared with littermate controls, while preserving cardiac ejection fraction. Collagen type I fibers were decreased in the Ank1-ifKO mice, suggesting a novel function of AnkR in the maturation of collagen fibers. In summary, our findings illustrate the novel expression of AnkR in cardiac fibroblasts and a potential role in cardiac function in response to stress.
Collapse
Affiliation(s)
- Aaron D. Argall
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Holly C. Sucharski-Argall
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Luke G. Comisford
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Sallie J. Jurs
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Jack T. Seminetta
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Michael J. Wallace
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Casey A. Crawford
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Sarah S. Takenaka
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Mei Han
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Mona El Refaey
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Thomas J. Hund
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
- Department of Biomedical Engineering, College of Engineering, Ohio State University, Columbus, OH 43210, USA
| | - Peter J. Mohler
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Sara N. Koenig
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA; (A.D.A.); (H.C.S.-A.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
7
|
van der Heijden ME, Brown AM, Kizek DJ, Sillitoe RV. Cerebellar nuclei cells produce distinct pathogenic spike signatures in mouse models of ataxia, dystonia, and tremor. eLife 2024; 12:RP91483. [PMID: 39072369 DOI: 10.7554/elife.91483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
The cerebellum contributes to a diverse array of motor conditions, including ataxia, dystonia, and tremor. The neural substrates that encode this diversity are unclear. Here, we tested whether the neural spike activity of cerebellar output neurons is distinct between movement disorders with different impairments, generalizable across movement disorders with similar impairments, and capable of causing distinct movement impairments. Using in vivo awake recordings as input data, we trained a supervised classifier model to differentiate the spike parameters between mouse models for ataxia, dystonia, and tremor. The classifier model correctly assigned mouse phenotypes based on single-neuron signatures. Spike signatures were shared across etiologically distinct but phenotypically similar disease models. Mimicking these pathophysiological spike signatures with optogenetics induced the predicted motor impairments in otherwise healthy mice. These data show that distinct spike signatures promote the behavioral presentation of cerebellar diseases.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Amanda M Brown
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Dominic J Kizek
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Roy V Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, United States
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| |
Collapse
|
8
|
van der Heijden ME, Brown AM, Kizek DJ, Sillitoe RV. Cerebellar nuclei cells produce distinct pathogenic spike signatures in mouse models of ataxia, dystonia, and tremor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.07.539767. [PMID: 37214855 PMCID: PMC10197583 DOI: 10.1101/2023.05.07.539767] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The cerebellum contributes to a diverse array of motor conditions including ataxia, dystonia, and tremor. The neural substrates that encode this diversity are unclear. Here, we tested whether the neural spike activity of cerebellar output neurons is distinct between movement disorders with different impairments, generalizable across movement disorders with similar impairments, and capable of causing distinct movement impairments. Using in vivo awake recordings as input data, we trained a supervised classifier model to differentiate the spike parameters between mouse models for ataxia, dystonia, and tremor. The classifier model correctly assigned mouse phenotypes based on single neuron signatures. Spike signatures were shared across etiologically distinct but phenotypically similar disease models. Mimicking these pathophysiological spike signatures with optogenetics induced the predicted motor impairments in otherwise healthy mice. These data show that distinct spike signatures promote the behavioral presentation of cerebellar diseases.
Collapse
|
9
|
Ding X, Wu Y, Rodriguez V, Ricco E, Okoh JT, Liu Y, Kraushaar DC, Rasband MN. Age-dependent regulation of axoglial interactions and behavior by oligodendrocyte AnkyrinG. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587609. [PMID: 38617359 PMCID: PMC11014615 DOI: 10.1101/2024.04.01.587609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The bipolar disorder (BD) risk gene ANK3 encodes the scaffolding protein AnkyrinG (AnkG). In neurons, AnkG regulates polarity and ion channel clustering at axon initial segments and nodes of Ranvier. Disruption of neuronal AnkG causes BD-like phenotypes in mice. During development, AnkG is also expressed at comparable levels in oligodendrocytes and facilitates the efficient assembly of paranodal junctions. However, the physiological roles of glial AnkG in the mature nervous system, and its contributions to BD-like phenotypes, remain unexplored. Here, we generated oligodendroglia-specific AnkG conditional knockout mice and observed the destabilization of axoglial interactions in aged but not young adult mice. In addition, these mice exhibited profound histological, electrophysiological, and behavioral pathophysiologies. Unbiased translatomic profiling revealed potential compensatory machineries. These results highlight the critical functions of glial AnkG in maintaining proper axoglial interactions throughout aging and suggests a previously unrecognized contribution of oligodendroglial AnkG to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yu Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Victoria Rodriguez
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - Emily Ricco
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - James T. Okoh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yanhong Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Daniel C. Kraushaar
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
10
|
Kumar P, Goettemoeller AM, Espinosa-Garcia C, Tobin BR, Tfaily A, Nelson RS, Natu A, Dammer EB, Santiago JV, Malepati S, Cheng L, Xiao H, Duong DD, Seyfried NT, Wood LB, Rowan MJM, Rangaraju S. Native-state proteomics of Parvalbumin interneurons identifies unique molecular signatures and vulnerabilities to early Alzheimer's pathology. Nat Commun 2024; 15:2823. [PMID: 38561349 PMCID: PMC10985119 DOI: 10.1038/s41467-024-47028-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Dysfunction in fast-spiking parvalbumin interneurons (PV-INs) may represent an early pathophysiological perturbation in Alzheimer's Disease (AD). Defining early proteomic alterations in PV-INs can provide key biological and translationally-relevant insights. We used cell-type-specific in-vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state PV-IN proteomes. PV-IN proteomic signatures include high metabolic and translational activity, with over-representation of AD-risk and cognitive resilience-related proteins. In bulk proteomes, PV-IN proteins were associated with cognitive decline in humans, and with progressive neuropathology in humans and the 5xFAD mouse model of Aβ pathology. PV-IN CIBOP in early stages of Aβ pathology revealed signatures of increased mitochondria and metabolism, synaptic and cytoskeletal disruption and decreased mTOR signaling, not apparent in whole-brain proteomes. Furthermore, we demonstrated pre-synaptic defects in PV-to-excitatory neurotransmission, validating our proteomic findings. Overall, in this study we present native-state proteomes of PV-INs, revealing molecular insights into their unique roles in cognitive resiliency and AD pathogenesis.
Collapse
Affiliation(s)
- Prateek Kumar
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Annie M Goettemoeller
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, USA
| | - Claudia Espinosa-Garcia
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Brendan R Tobin
- Georgia W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Ali Tfaily
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Ruth S Nelson
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Aditya Natu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eric B Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Juliet V Santiago
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, USA
| | - Sneha Malepati
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lihong Cheng
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
| | - Hailian Xiao
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
| | - Duc D Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Nicholas T Seyfried
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Levi B Wood
- Georgia W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
- School of Chemical and Biological Engineering, GeoInsrgia titute of Technology, Atlanta, GA, 30322, USA
| | - Matthew J M Rowan
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Srikant Rangaraju
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA.
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
11
|
Sert O, Ding X, Zhang C, Mi R, Hoke A, Rasband MN. Postsynaptic β1 spectrin maintains Na + channels at the neuromuscular junction. J Physiol 2024; 602:1127-1145. [PMID: 38441922 PMCID: PMC10942750 DOI: 10.1113/jp285894] [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: 11/01/2023] [Accepted: 02/13/2024] [Indexed: 03/16/2024] Open
Abstract
Spectrins function together with actin as obligatory subunits of the submembranous cytoskeleton. Spectrins maintain cell shape, resist mechanical forces, and stabilize ion channel and transporter protein complexes through binding to scaffolding proteins. Recently, pathogenic variants of SPTBN4 (β4 spectrin) were reported to cause both neuropathy and myopathy. Although the role of β4 spectrin in neurons is mostly understood, its function in skeletal muscle, another excitable tissue subject to large forces, is unknown. Here, using a muscle specific β4 spectrin conditional knockout mouse, we show that β4 spectrin does not contribute to muscle function. In addition, we show β4 spectrin is not present in muscle, indicating the previously reported myopathy associated with pathogenic SPTBN4 variants is neurogenic in origin. More broadly, we show that α2, β1 and β2 spectrins are found in skeletal muscle, with α2 and β1 spectrins being enriched at the postsynaptic neuromuscular junction (NMJ). Surprisingly, using muscle specific conditional knockout mice, we show that loss of α2 and β2 spectrins had no effect on muscle health, function or the enrichment of β1 spectrin at the NMJ. Muscle specific deletion of β1 spectrin also had no effect on muscle health, but, with increasing age, resulted in the loss of clustered NMJ Na+ channels. Together, our results suggest that muscle β1 spectrin functions independently of an associated α spectrin to maintain Na+ channel clustering at the postsynaptic NMJ. Furthermore, despite repeated exposure to strong forces and in contrast to neurons, muscles do not require spectrin cytoskeletons to maintain cell shape or integrity. KEY POINTS: The myopathy found in pathogenic human SPTBN4 variants (where SPTBN4 is the gene encoding β4 spectrin) is neurogenic in origin. β1 spectrin plays essential roles in maintaining the density of neuromuscular junction Nav1.4 Na+ channels. By contrast to the canonical view of spectrin organization and function, we show that β1 spectrin can function independently of an associated α spectrin. Despite the large mechanical forces experienced by muscle, we show that spectrins are not required for muscle cell integrity. This is in stark contrast to red blood cells and the axons of neurons.
Collapse
Affiliation(s)
- Ozlem Sert
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA 77030
| | - Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA 77030
| | - Chuansheng Zhang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA 77030
| | - Ruifa Mi
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Ahmet Hoke
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA 77030
| |
Collapse
|
12
|
Page CE, Coutellier L. Kv3.1 Voltage-gated Potassium Channels Modulate Anxiety-like Behaviors in Female Mice. Neuroscience 2024; 538:68-79. [PMID: 38157976 PMCID: PMC10872248 DOI: 10.1016/j.neuroscience.2023.12.011] [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: 09/21/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Inhibitory parvalbumin (PV) interneurons regulate the activity of neural circuits within brain regions involved in emotional processing, including the prefrontal cortex (PFC). Recently, rodent studies have implicated a stress-induced increase in prefrontal PV neuron activity in the development of anxiety behaviors, particularly in females. However, the mechanisms through which stress increases activity of prefrontal PV neurons remain unknown. The fast-spiking properties of PV neurons in part come from their expression of voltage-gated potassium (K+) ion channels, particularly Kv3.1 channels. We therefore suggest that stress-induced changes in Kv3.1 channels contribute to the appearance of an anxious phenotype following chronic stress in female mice. Here, we first showed that unpredictable chronic mild stress (UCMS) increased expression of Kv3.1 channels on prefrontal PV neurons in female mice, a potential mechanism underlying the previously observed hyperactivity of these neurons after stress. We then showed that female mice deficient in Kv3.1 channels displayed resilience to UCMS-induced anxiety-like behaviors. Altogether, our findings implicate Kv3.1 channels in the development of anxiety-like behaviors following UCMS, particularly in females, providing a novel mechanism to understand sex-specific vulnerabilities to stress-induced psychopathologies.
Collapse
Affiliation(s)
- Chloe E Page
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Laurence Coutellier
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States; Department of Psychology, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
13
|
Ogawa Y, Lim BC, George S, Oses-Prieto JA, Rasband JM, Eshed-Eisenbach Y, Hamdan H, Nair S, Boato F, Peles E, Burlingame AL, Van Aelst L, Rasband MN. Antibody-directed extracellular proximity biotinylation reveals that Contactin-1 regulates axo-axonic innervation of axon initial segments. Nat Commun 2023; 14:6797. [PMID: 37884508 PMCID: PMC10603070 DOI: 10.1038/s41467-023-42273-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Axon initial segment (AIS) cell surface proteins mediate key biological processes in neurons including action potential initiation and axo-axonic synapse formation. However, few AIS cell surface proteins have been identified. Here, we use antibody-directed proximity biotinylation to define the cell surface proteins in close proximity to the AIS cell adhesion molecule Neurofascin. To determine the distributions of the identified proteins, we use CRISPR-mediated genome editing for insertion of epitope tags in the endogenous proteins. We identify Contactin-1 (Cntn1) as an AIS cell surface protein. Cntn1 is enriched at the AIS through interactions with Neurofascin and NrCAM. We further show that Cntn1 contributes to assembly of the AIS extracellular matrix, and regulates AIS axo-axonic innervation by inhibitory basket cells in the cerebellum and inhibitory chandelier cells in the cortex.
Collapse
Affiliation(s)
- Yuki Ogawa
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Brian C Lim
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shanu George
- Division of Neuroscience, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Joshua M Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yael Eshed-Eisenbach
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hamdan Hamdan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Physiology and Immunology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Supna Nair
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Francesco Boato
- Division of Neuroscience, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Elior Peles
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Linda Van Aelst
- Division of Neuroscience, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
14
|
Miao HT, Song RX, Xin Y, Wang LY, Lv JM, Liu NN, Wu ZY, Zhang W, Li Y, Zhang DX, Zhang LM. Spautin-1 Protects Against Mild TBI-Induced Anxiety-Like Behavior in Mice via Immunologically Silent Apoptosis. Neuromolecular Med 2023; 25:336-349. [PMID: 36745326 DOI: 10.1007/s12017-023-08737-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
Anxiety is reportedly one of the most common mental changes after traumatic brain injury (TBI). Perineuronal nets (PNNs) produced by astrocytes in the lateral hypothalamus (LHA) that surround gamma-aminobutyric acid-ergic (GABAergic) neurons have been associated with anxiety. The potent anti-tumor effects of Spautin-1, a novel autophagy inhibitor, have been documented in malignant melanoma; moreover, the inhibition of autophagy is reported to mitigate anxiety disorders. However, little is known about the ability of spautin-1 to alleviate anxiety. In this study, we sought to investigate whether spautin-1 could alleviate anxiety-like behaviors post-TBI by reducing the loss of PNNs in the LHA. A mild TBI was established in mice through Feeney's weight-drop model. Then, Spautin-1 (20 mmol/2 μl) was immediately administered into the left lateral ventricle. Behavioral and pathological changes were assessed at 24 h, 7 days, 30 days, 31 days and 32 days after TBI by the neurological severity scores (NSS), open field test (OFT), elevated plus-maze (EPM) test, western blot, immunofluorescence assays and electron microscopy. Spautin-1 significantly reversed TBI-induced decreased time in the central zone during OFT and in the open-arm during the EPM test. Spautin-1 also increased PNNs around GABAergic neurons indicated by WFA- plus GAD2- positive A2-type astrocytes and attenuated M1-type microglia in the LHA 32 days after TBI compared to TBI alone. Moreover, compared to mice that only underwent TBI, spautin-1 downregulated autophagic vacuoles, abnormal organelles, the expression of Beclin 1, USP13, phospho-TBK1, and phospho-IRF3 and upregulated the levels of cleaved caspase-3, -7 and -9, but failed to increase TUNEL-positive cells in the LHA at 24 h. Spautin-1 alleviated anxiety-like behavior in mice exposed to mild TBI; this protective mechanism may be associated with decreased PNNs loss around GABAergic neurons via immunologically silent apoptosis induced by the caspase cascade.
Collapse
Affiliation(s)
- Hui-Tao Miao
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Rong-Xin Song
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Yue Xin
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Lu-Ying Wang
- Department of Anesthesia and Trauma Research, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Jin-Meng Lv
- Department of Anesthesia and Trauma Research, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Na-Na Liu
- Department of Pediatric, Cangzhou Central Hospital, Cangzhou, China
| | - Zhi-You Wu
- Department of Neurosurgery, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Wei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China.
| |
Collapse
|
15
|
Yang JJ, Huang RC. Afterhyperpolarization potential modulated by local [K +] o in K + diffusion-restricted extracellular space in the central clock of suprachiasmatic nucleus. Biomed J 2023; 46:100551. [PMID: 35863667 PMCID: PMC10345224 DOI: 10.1016/j.bj.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/31/2022] [Accepted: 07/14/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Intercellular coupling is essential for the suprachiasmatic nucleus (SCN) to serve as a coherent central clock. Synaptic release of neurotransmitters and neuropeptides is critical for synchronizing SCN neurons. However, intercellular coupling via non-synaptic mechanisms has also been demonstrated. In particular, the abundant perikaryal appositions with morphological specializations in the narrow extracellular space (ECS) may hinder molecular diffusion to allow for ion accumulation or depletion. METHODS The SCN neurons were recorded in the whole-cell current-clamp mode, with pipette filled with high (26 mM)-Na+ or low (6 mM)-Na+ solution. RESULTS Cells recorded with high-Na+ pipette solution could fire spontaneous action potentials (AP) with peak AHP more negative than the calculated value of K+ equilibrium potential (EK) and with peak AP more positive than calculated ENa. Cells recorded with low-Na+ pipette solution could also have peak AHP more negative than calculated EK. In contrast, the resting membrane potential (RMP) was always less negative to calculated EK. The distribution and the averaged amplitude of peak AHP, peak AP, or RMP was similar between cells recorded with high-Na+ and low-Na+ solution pipette. In a number of cells, the peak AHP could increase from more positive to become more negative than calculated EK spontaneously or after treatments to hyperpolarize the RMP. TTX blocked the Na+ -dependent APs and tetraethylammonium (TEA), but not Ba2+ or Cd2+, markedly reduced the peak AHP. Perforated-patch cells could also but rarely fire APs with peak AHP more negative than calculated EK. CONCLUSION The result of peak AHP negative to calculated EK indicates that local [K+]o sensed by the TEA-sensitive AHP K+ channels must be lower than bulk [K+]o, most likely due to K+ clearance from K+ diffusion-restricted ECS by the Na+/K+-ATPase. The K+ diffusion-restricted ECS may allow for K+-mediated ionic interactions among neurons to regulate SCN excitability.
Collapse
Affiliation(s)
- Jyh-Jeen Yang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Rong-Chi Huang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| |
Collapse
|
16
|
Kumar P, Goettemoeller AM, Espinosa-Garcia C, Tobin BR, Tfaily A, Nelson RS, Natu A, Dammer EB, Santiago JV, Malepati S, Cheng L, Xiao H, Duong D, Seyfried NT, Wood LB, Rowan MJ, Rangaraju S. Native-state proteomics of Parvalbumin interneurons identifies novel molecular signatures and metabolic vulnerabilities to early Alzheimer's disease pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541038. [PMID: 37292756 PMCID: PMC10245729 DOI: 10.1101/2023.05.17.541038] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One of the earliest pathophysiological perturbations in Alzheimer's Disease (AD) may arise from dysfunction of fast-spiking parvalbumin (PV) interneurons (PV-INs). Defining early protein-level (proteomic) alterations in PV-INs can provide key biological and translationally relevant insights. Here, we use cell-type-specific in vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state proteomes of PV interneurons. PV-INs exhibited proteomic signatures of high metabolic, mitochondrial, and translational activity, with over-representation of causally linked AD genetic risk factors. Analyses of bulk brain proteomes indicated strong correlations between PV-IN proteins with cognitive decline in humans, and with progressive neuropathology in humans and mouse models of Aβ pathology. Furthermore, PV-IN-specific proteomes revealed unique signatures of increased mitochondrial and metabolic proteins, but decreased synaptic and mTOR signaling proteins in response to early Aβ pathology. PV-specific changes were not apparent in whole-brain proteomes. These findings showcase the first native state PV-IN proteomes in mammalian brain, revealing a molecular basis for their unique vulnerabilities in AD.
Collapse
|
17
|
Lorenzo DN, Edwards RJ, Slavutsky AL. Spectrins: molecular organizers and targets of neurological disorders. Nat Rev Neurosci 2023; 24:195-212. [PMID: 36697767 PMCID: PMC10598481 DOI: 10.1038/s41583-022-00674-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 01/26/2023]
Abstract
Spectrins are cytoskeletal proteins that are expressed ubiquitously in the mammalian nervous system. Pathogenic variants in SPTAN1, SPTBN1, SPTBN2 and SPTBN4, four of the six genes encoding neuronal spectrins, cause neurological disorders. Despite their structural similarity and shared role as molecular organizers at the cell membrane, spectrins vary in expression, subcellular localization and specialization in neurons, and this variation partly underlies non-overlapping disease presentations across spectrinopathies. Here, we summarize recent progress in discerning the local and long-range organization and diverse functions of neuronal spectrins. We provide an overview of functional studies using mouse models, which, together with growing human genetic and clinical data, are helping to illuminate the aetiology of neurological spectrinopathies. These approaches are all critical on the path to plausible therapeutic solutions.
Collapse
Affiliation(s)
- Damaris N Lorenzo
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Reginald J Edwards
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anastasia L Slavutsky
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
18
|
Ogawa Y, Lim BC, George S, Oses-Prieto JA, Rasband JM, Eshed-Eisenbach Y, Nair S, Boato F, Peles E, Burlingame AL, Van Aelst L, Rasband MN. Antibody-directed extracellular proximity biotinylation reveals Contactin-1 regulates axo-axonic innervation of axon initial segments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531378. [PMID: 36945454 PMCID: PMC10028829 DOI: 10.1101/2023.03.06.531378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Axon initial segment (AIS) cell surface proteins mediate key biological processes in neurons including action potential initiation and axo-axonic synapse formation. However, few AIS cell surface proteins have been identified. Here, we used antibody-directed proximity biotinylation to define the cell surface proteins in close proximity to the AIS cell adhesion molecule Neurofascin. To determine the distributions of the identified proteins, we used CRISPR-mediated genome editing for insertion of epitope tags in the endogenous proteins. We found Contactin-1 (Cntn1) among the previously unknown AIS proteins we identified. Cntn1 is enriched at the AIS through interactions with Neurofascin and NrCAM. We further show that Cntn1 contributes to assembly of the AIS-extracellular matrix, and is required for AIS axo-axonic innervation by inhibitory basket cells in the cerebellum and inhibitory chandelier cells in the cortex.
Collapse
Affiliation(s)
- Yuki Ogawa
- Baylor College of Medicine, Department of Neuroscience, Houston, TX, USA
| | - Brian C. Lim
- Baylor College of Medicine, Department of Neuroscience, Houston, TX, USA
| | - Shanu George
- Cold Spring Harbor Laboratory, Division of Neuroscience, Cold Spring Harbor, NY, USA
| | - Juan A. Oses-Prieto
- University of California San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA, USA
| | - Joshua M. Rasband
- Baylor College of Medicine, Department of Neuroscience, Houston, TX, USA
| | - Yael Eshed-Eisenbach
- Weizmann Institute of Science, Department of Molecular Cell Biology, Rehovot, Israel
| | - Supna Nair
- University of California San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA, USA
| | - Francesco Boato
- Cold Spring Harbor Laboratory, Division of Neuroscience, Cold Spring Harbor, NY, USA
| | - Elior Peles
- Weizmann Institute of Science, Department of Molecular Cell Biology, Rehovot, Israel
| | - Alma L. Burlingame
- University of California San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA, USA
| | - Linda Van Aelst
- Cold Spring Harbor Laboratory, Division of Neuroscience, Cold Spring Harbor, NY, USA
| | - Matthew N. Rasband
- Baylor College of Medicine, Department of Neuroscience, Houston, TX, USA
| |
Collapse
|
19
|
Bauer S, Chen CY, Jonson M, Kaczmarczyk L, Magadi SS, Jackson WS. Cerebellar granule neurons induce Cyclin D1 before the onset of motor symptoms in Huntington's disease mice. Acta Neuropathol Commun 2023; 11:17. [PMID: 36670467 PMCID: PMC9854201 DOI: 10.1186/s40478-022-01500-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/16/2022] [Indexed: 01/21/2023] Open
Abstract
Although Huntington's disease (HD) is classically defined by the selective vulnerability of striatal projection neurons, there is increasing evidence that cerebellar degeneration modulates clinical symptoms. However, little is known about cell type-specific responses of cerebellar neurons in HD. To dissect early disease mechanisms in the cerebellum and cerebrum, we analyzed translatomes of neuronal cell types from both regions in a new HD mouse model. For this, HdhQ200 knock-in mice were backcrossed with the calm 129S4 strain, to constrain experimental noise caused by variable hyperactivity of mice in a C57BL/6 background. Behavioral and neuropathological characterization showed that these S4-HdhQ200 mice had very mild behavioral abnormalities starting around 12 months of age that remained mild up to 18 months. By 9 months, we observed abundant Huntingtin-positive neuronal intranuclear inclusions (NIIs) in the striatum and cerebellum. The translatome analysis of GABAergic cells of the cerebrum further confirmed changes typical of HD-induced striatal pathology. Surprisingly, we observed the strongest response with 626 differentially expressed genes in glutamatergic neurons of the cerebellum, a population consisting primarily of granule cells, commonly considered disease resistant. Our findings suggest vesicular fusion and exocytosis, as well as differentiation-related pathways are affected in these neurons. Furthermore, increased expression of cyclin D1 (Ccnd1) in the granular layer and upregulated expression of polycomb group complex protein genes and cell cycle regulators Cbx2, Cbx4 and Cbx8 point to a putative role of aberrant cell cycle regulation in cerebellar granule cells in early disease.
Collapse
Affiliation(s)
- Susanne Bauer
- grid.5640.70000 0001 2162 9922Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Room 463.10.30, Linköping, Sweden
| | - Chwen-Yu Chen
- grid.5640.70000 0001 2162 9922Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Room 463.10.30, Linköping, Sweden
| | - Maria Jonson
- grid.5640.70000 0001 2162 9922Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Room 463.10.30, Linköping, Sweden
| | - Lech Kaczmarczyk
- grid.5640.70000 0001 2162 9922Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Room 463.10.30, Linköping, Sweden ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Srivathsa Subramanya Magadi
- grid.5640.70000 0001 2162 9922Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Room 463.10.30, Linköping, Sweden
| | - Walker S. Jackson
- grid.5640.70000 0001 2162 9922Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Room 463.10.30, Linköping, Sweden ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases, Bonn, Germany
| |
Collapse
|
20
|
Abstract
The ankyrin proteins (Ankyrin-R, Ankyrin-B, and Ankyrin-G) are a family of scaffolding, or membrane adaptor proteins necessary for the regulation and targeting of several types of ion channels and membrane transporters throughout the body. These include voltage-gated sodium, potassium, and calcium channels in the nervous system, heart, lungs, and muscle. At these sites, ankyrins recruit ion channels, and other membrane proteins, to specific subcellular domains, which are then stabilized through ankyrin's interaction with the submembranous spectrin-based cytoskeleton. Several recent studies have expanded our understanding of both ankyrin expression and their ion channel binding partners. This review provides an updated overview of ankyrin proteins and their known channel and transporter interactions. We further discuss several potential avenues of future research that would expand our understanding of these important organizational proteins.
Collapse
Affiliation(s)
- Sharon R. Stevens
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA,CONTACT Matthew N. Rasband Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX77030, USA
| |
Collapse
|
21
|
Ali AB, Islam A, Constanti A. The fate of interneurons, GABA A receptor sub-types and perineuronal nets in Alzheimer's disease. Brain Pathol 2022; 33:e13129. [PMID: 36409151 PMCID: PMC9836378 DOI: 10.1111/bpa.13129] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurological disease, which is associated with gradual memory loss and correlated with synaptic hyperactivity and abnormal oscillatory rhythmic brain activity that precedes phenotypic alterations and is partly responsible for the spread of the disease pathology. Synaptic hyperactivity is thought to be because of alteration in the homeostasis of phasic and tonic synaptic inhibition, which is orchestrated by the GABAA inhibitory system, encompassing subclasses of interneurons and GABAA receptors, which play a vital role in cognitive functions, including learning and memory. Furthermore, the extracellular matrix, the perineuronal nets (PNNs) which often go unnoticed in considerations of AD pathology, encapsulate the inhibitory cells and neurites in critical brain regions and have recently come under the light for their crucial role in synaptic stabilisation and excitatory-inhibitory balance and when disrupted, serve as a potential trigger for AD-associated synaptic imbalance. Therefore, in this review, we summarise the current understanding of the selective vulnerability of distinct interneuron subtypes, their synaptic and extrasynaptic GABAA R subtypes as well as the changes in PNNs in AD, detailing their contribution to the mechanisms of disease development. We aim to highlight how seemingly unique malfunction in each component of the interneuronal GABA inhibitory system can be tied together to result in critical circuit dysfunction, leading to the irreversible symptomatic damage observed in AD.
Collapse
|
22
|
Fawcett JW, Fyhn M, Jendelova P, Kwok JCF, Ruzicka J, Sorg BA. The extracellular matrix and perineuronal nets in memory. Mol Psychiatry 2022; 27:3192-3203. [PMID: 35760878 PMCID: PMC9708575 DOI: 10.1038/s41380-022-01634-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 02/06/2023]
Abstract
All components of the CNS are surrounded by a diffuse extracellular matrix (ECM) containing chondroitin sulphate proteoglycans (CSPGs), heparan sulphate proteoglycans (HSPGs), hyaluronan, various glycoproteins including tenascins and thrombospondin, and many other molecules that are secreted into the ECM and bind to ECM components. In addition, some neurons, particularly inhibitory GABAergic parvalbumin-positive (PV) interneurons, are surrounded by a more condensed cartilage-like ECM called perineuronal nets (PNNs). PNNs surround the soma and proximal dendrites as net-like structures that surround the synapses. Attention has focused on the role of PNNs in the control of plasticity, but it is now clear that PNNs also play an important part in the modulation of memory. In this review we summarize the role of the ECM, particularly the PNNs, in the control of various types of memory and their participation in memory pathology. PNNs are now being considered as a target for the treatment of impaired memory. There are many potential treatment targets in PNNs, mainly through modulation of the sulphation, binding, and production of the various CSPGs that they contain or through digestion of their sulphated glycosaminoglycans.
Collapse
Affiliation(s)
- James W Fawcett
- John van Geest Centre for Brain Repair, Department Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK.
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic.
| | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Pavla Jendelova
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Jessica C F Kwok
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jiri Ruzicka
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Barbara A Sorg
- Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| |
Collapse
|
23
|
Vallese F, Kim K, Yen LY, Johnston JD, Noble AJ, Calì T, Clarke OB. Architecture of the human erythrocyte ankyrin-1 complex. Nat Struct Mol Biol 2022; 29:706-718. [PMID: 35835865 PMCID: PMC10373098 DOI: 10.1038/s41594-022-00792-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/24/2022] [Indexed: 12/28/2022]
Abstract
The stability and shape of the erythrocyte membrane is provided by the ankyrin-1 complex, but how it tethers the spectrin-actin cytoskeleton to the lipid bilayer and the nature of its association with the band 3 anion exchanger and the Rhesus glycoproteins remains unknown. Here we present structures of ankyrin-1 complexes purified from human erythrocytes. We reveal the architecture of a core complex of ankyrin-1, the Rhesus proteins RhAG and RhCE, the band 3 anion exchanger, protein 4.2, glycophorin A and glycophorin B. The distinct T-shaped conformation of membrane-bound ankyrin-1 facilitates recognition of RhCE and, unexpectedly, the water channel aquaporin-1. Together, our results uncover the molecular details of ankyrin-1 association with the erythrocyte membrane, and illustrate the mechanism of ankyrin-mediated membrane protein clustering.
Collapse
Affiliation(s)
- Francesca Vallese
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA.,Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.,Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| | - Kookjoo Kim
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA.,Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.,Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| | - Laura Y Yen
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Jake D Johnston
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.,Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, USA
| | - Alex J Noble
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, USA
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Padua Neuroscience Center (PNC), University of Padua, Padua, Italy.,Study Center for Neurodegeneration (CESNE), University of Padua, Padua, Italy
| | - Oliver Biggs Clarke
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA. .,Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA. .,Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA.
| |
Collapse
|
24
|
Liu L, Zhang Y, Ju J. Removal of perineuronal nets leads to altered neuronal excitability and synaptic transmission in the visual cortex with distinct time courses. Neurosci Lett 2022; 785:136763. [PMID: 35760385 DOI: 10.1016/j.neulet.2022.136763] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/30/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022]
Abstract
Parvalbumin-expressing (PV) interneurons fast inhibit excitatory neurons in various brain areas. Perineuronal nets (PNNs), accumulating around PV neurons, have been shown to play critical roles in neuronal function and plasticity. The cellular mechanisms underlying their functions are still in debate, for example, do PNNs contribute significantly to the excitability of inhibitory neurons especially those containing PV? On the other hand, whether PNNs have significant contributions to synaptic transmission of PV neurons is much less unknown. In this study, we designed experiments to address these questions and found that removing PNNs in vivo using chondroitinase ABC (ChABC) led to distinct changes in neuronal excitability and synaptic transmission, depending on the duration of ChABC treatment. The results showed 7 days after ChABC treatment reduced both intrinsic excitability of PV neurons and synaptic transmission to both PV neurons and excitatory neurons in the primary visual cortex. However, 1 day after ChABC treatment digested PNNs effectively but had no effects on intrinsic excitability and synaptic transmission. These results suggest the contribution of PNNs to neuronal excitability and synaptic transmission depends on different time courses of ChABC digestion.
Collapse
Affiliation(s)
- Luping Liu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Yujie Zhang
- The Pediatric Neurology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Jun Ju
- Brain Research Centre and Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
25
|
Richardson A, Ciampani V, Stancu M, Bondarenko K, Newton S, Steinert JR, Pilati N, Graham BP, Kopp-Scheinpflug C, Forsythe ID. Kv3.3 subunits control presynaptic action potential waveform and neurotransmitter release at a central excitatory synapse. eLife 2022; 11:75219. [PMID: 35510987 PMCID: PMC9110028 DOI: 10.7554/elife.75219] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/29/2022] [Indexed: 11/29/2022] Open
Abstract
Kv3 potassium currents mediate rapid repolarisation of action potentials (APs), supporting fast spikes and high repetition rates. Of the four Kv3 gene family members, Kv3.1 and Kv3.3 are highly expressed in the auditory brainstem and we exploited this to test for subunit-specific roles at the calyx of Held presynaptic terminal in the mouse. Deletion of Kv3.3 (but not Kv3.1) reduced presynaptic Kv3 channel immunolabelling, increased presynaptic AP duration and facilitated excitatory transmitter release; which in turn enhanced short-term depression during high-frequency transmission. The response to sound was delayed in the Kv3.3KO, with higher spontaneous and lower evoked firing, thereby reducing signal-to-noise ratio. Computational modelling showed that the enhanced EPSC and short-term depression in the Kv3.3KO reflected increased vesicle release probability and accelerated activity-dependent vesicle replenishment. We conclude that Kv3.3 mediates fast repolarisation for short precise APs, conserving transmission during sustained high-frequency activity at this glutamatergic excitatory synapse.
Collapse
Affiliation(s)
- Amy Richardson
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Victoria Ciampani
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Mihai Stancu
- Division of Neurobiology, Ludwig-Maximilians-Universität München, Munchen, Germany
| | - Kseniia Bondarenko
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Sherylanne Newton
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Joern R Steinert
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Nadia Pilati
- Istituto di Ricerca Pediatrica Citta'della Speranza, Padova, Italy
| | - Bruce P Graham
- Computing Science and Mathematics, University of Stirling, Stirling, United Kingdom
| | | | - Ian D Forsythe
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
26
|
Shi Y, Wu X, Zhou J, Cui W, Wang J, Hu Q, Zhang S, Han L, Zhou M, Luo J, Wang Q, Liu H, Feng D, Ge S, Qu Y. Single-Nucleus RNA Sequencing Reveals that Decorin Expression in the Amygdala Regulates Perineuronal Nets Expression and Fear Conditioning Response after Traumatic Brain Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104112. [PMID: 35038242 PMCID: PMC8895134 DOI: 10.1002/advs.202104112] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Indexed: 06/14/2023]
Abstract
Traumatic brain injury (TBI) is a risk factor for posttraumatic stress disorder (PTSD). Augmented fear is a defining characteristic of PTSD, and the amygdala is considered the main brain region to process fear. The mechanism by which the amygdala is involved in fear conditioning after TBI is still unclear. Using single-nucleus RNA sequencing (snRNA-seq), transcriptional changes in cells in the amygdala after TBI are investigated. In total, 72 328 nuclei are obtained from the sham and TBI groups. 7 cell types, and analysis of differentially expressed genes (DEGs) reveals widespread transcriptional changes in each cell type after TBI are identified. In in vivo experiments, it is demonstrated that Decorin (Dcn) expression in the excitatory neurons of the amygdala significantly increased after TBI, and Dcn knockout in the amygdala mitigates TBI-associated fear conditioning. Of note, this effect is caused by a Dcn-mediated decrease in the expression of perineuronal nets (PNNs), which affect the glutamate-γ-aminobutyric acid balance in the amygdala. Finally, the results suggest that Dcn functions by interacting with collagen VI α3 (Col6a3). Consequently, the findings reveal transcriptional changes in different cell types of the amygdala after TBI and provide direct evidence that Dcn relieves fear conditioning by regulating PNNs.
Collapse
Affiliation(s)
- Yingwu Shi
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Xun Wu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Jinpeng Zhou
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Wenxing Cui
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Jin Wang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Qing Hu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Shenghao Zhang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Liying Han
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Meixuan Zhou
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Jianing Luo
- Department of NeurosurgeryWest Theater General HospitalChengduSichuan610083China
| | - Qiang Wang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Haixiao Liu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Dayun Feng
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Shunnan Ge
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Yan Qu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| |
Collapse
|
27
|
Laham BJ, Gould E. How Stress Influences the Dynamic Plasticity of the Brain’s Extracellular Matrix. Front Cell Neurosci 2022; 15:814287. [PMID: 35145379 PMCID: PMC8821883 DOI: 10.3389/fncel.2021.814287] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Diffuse and structured extracellular matrix (ECM) comprise ∼20% of the brain’s volume and play important roles in development and adult plasticity. Perineuronal nets (PNNs), specialized ECM structures that surround certain types of neurons in the brain, emerge during the postnatal period, making their development and maintenance potentially sensitive to experience. Recent studies have shown that stress affects diffuse ECM as well as PNNs, and that such effects are dependent on life stage and brain region. Given that the ECM participates in synaptic plasticity, the generation of neuronal oscillations, and synchronous firing across brain regions, all of which have been linked to cognition and emotional regulation, ECM components may be candidate therapeutic targets for stress-induced neuropsychiatric disease. This review considers the influence of stress over diffuse and structured ECM during postnatal life with a focus on functional outcomes and the potential for translational relevance.
Collapse
|
28
|
Disruption of MeCP2-TCF20 complex underlies distinct neurodevelopmental disorders. Proc Natl Acad Sci U S A 2022; 119:2119078119. [PMID: 35074918 PMCID: PMC8794850 DOI: 10.1073/pnas.2119078119] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 12/16/2022] Open
Abstract
MeCP2 is associated with Rett syndrome (RTT), MECP2 duplication syndrome, and a number of conditions with isolated features of these diseases, including autism, intellectual disability, and motor dysfunction. MeCP2 is known to broadly bind methylated DNA, but the precise molecular mechanism driving disease pathogenesis remains to be determined. Using proximity-dependent biotinylation (BioID), we identified a transcription factor 20 (TCF20) complex that interacts with MeCP2 at the chromatin interface. Importantly, RTT-causing mutations in MECP2 disrupt this interaction. TCF20 and MeCP2 are highly coexpressed in neurons and coregulate the expression of key neuronal genes. Reducing Tcf20 partially rescued the behavioral deficits caused by MECP2 overexpression, demonstrating a functional relationship between MeCP2 and TCF20 in MECP2 duplication syndrome pathogenesis. We identified a patient exhibiting RTT-like neurological features with a missense mutation in the PHF14 subunit of the TCF20 complex that abolishes the MeCP2-PHF14-TCF20 interaction. Our data demonstrate the critical role of the MeCP2-TCF20 complex for brain function.
Collapse
|
29
|
Stevens SR, van der Heijden ME, Ogawa Y, Lin T, Sillitoe RV, Rasband MN. Ankyrin-R Links Kv3.3 to the Spectrin Cytoskeleton and Is Required for Purkinje Neuron Survival. J Neurosci 2022; 42:2-15. [PMID: 34785580 PMCID: PMC8741159 DOI: 10.1523/jneurosci.1132-21.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/26/2021] [Accepted: 10/31/2021] [Indexed: 11/21/2022] Open
Abstract
Ankyrin scaffolding proteins are critical for membrane domain organization and protein stabilization in many different cell types including neurons. In the cerebellum, Ankyrin-R (AnkR) is highly enriched in Purkinje neurons, granule cells, and in the cerebellar nuclei (CN). Using male and female mice with a floxed allele for Ank1 in combination with Nestin-Cre and Pcp2-Cre mice, we found that ablation of AnkR from Purkinje neurons caused ataxia, regional and progressive neurodegeneration, and altered cerebellar output. We show that AnkR interacts with the cytoskeletal protein β3 spectrin and the potassium channel Kv3.3. Loss of AnkR reduced somatic membrane levels of β3 spectrin and Kv3.3 in Purkinje neurons. Thus, AnkR links Kv3.3 channels to the β3 spectrin-based cytoskeleton. Our results may help explain why mutations in β3 spectrin and Kv3.3 both cause spinocerebellar ataxia.SIGNIFICANCE STATEMENT Ankyrin scaffolding proteins localize and stabilize ion channels in the membrane by linking them to the spectrin-based cytoskeleton. Here, we show that Ankyrin-R (AnkR) links Kv3.3 K+ channels to the β3 spectrin-based cytoskeleton in Purkinje neurons. Loss of AnkR causes Purkinje neuron degeneration, altered cerebellar physiology, and ataxia, which is consistent with mutations in Kv3.3 and β3 spectrin causing spinocerebellar ataxia.
Collapse
Affiliation(s)
- Sharon R Stevens
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | | | - Yuki Ogawa
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Tao Lin
- Department Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030
| | - Roy V Sillitoe
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Department Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
30
|
Zhang C, Joshi A, Liu Y, Sert O, Haddix SG, Teliska LH, Rasband A, Rodney GG, Rasband MN. Ankyrin-dependent Na + channel clustering prevents neuromuscular synapse fatigue. Curr Biol 2021; 31:3810-3819.e4. [PMID: 34289389 DOI: 10.1016/j.cub.2021.06.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 01/18/2023]
Abstract
Skeletal muscle contraction depends on activation of clustered acetylcholine receptors (AchRs) and muscle-specific Na+ channels (Nav1.4). Some Nav1.4 channels are highly enriched at the neuromuscular junction (NMJ), and their clustering is thought to be essential for effective muscle excitation. However, this has not been experimentally tested, and how NMJ Na+ channels are clustered is unknown. Here, using muscle-specific ankyrinR, ankyrinB, and ankyrinG single, double, and triple-conditional knockout mice, we show that Nav1.4 channels fail to cluster only after deletion of all three ankyrins. Remarkably, ankyrin-deficient muscles have normal NMJ morphology, AchR clustering, sarcolemmal levels of Nav1.4, and muscle force, and they show no indication of degeneration. However, mice lacking clustered NMJ Na+ channels have significantly reduced levels of motor activity and their NMJs rapidly fatigue after repeated nerve-dependent stimulation. Thus, the triple redundancy of ankyrins facilitates NMJ Na+ channel clustering to prevent neuromuscular synapse fatigue.
Collapse
Affiliation(s)
- Chuansheng Zhang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Abhijeet Joshi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yanhong Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ozlem Sert
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Seth G Haddix
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lindsay H Teliska
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anne Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - George G Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|