1
|
Lambert MW. Critical role of alpha spectrin in DNA repair: the importance of μ-calpain and Fanconi anemia proteins. Exp Biol Med (Maywood) 2025; 250:10537. [PMID: 40375875 PMCID: PMC12078185 DOI: 10.3389/ebm.2025.10537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/17/2025] [Indexed: 05/18/2025] Open
Abstract
Nonerythroid spectrins are proteins important in maintaining the structural integrity and flexibility of the cell and nuclear membranes and are essential for a number of functionally important cellular processes. One of these proteins, nonerythroid α spectrin (αSpII), plays a critical role in DNA repair, specifically repair of DNA interstrand crosslinks (ICLs), where it acts as a scaffold, recruiting repair proteins to sites of damage. Loss or breakdown of αSpII is an important factor in a number of disorders. One of these is Fanconi anemia (FA), a genetic disorder characterized by bone marrow failure, chromosome instability, cancer predisposition, congenital abnormalities and a defect in DNA ICL repair. Significantly, breakdown of αSpII occurs in cells from a number of FA complementation groups, due to excessive cleavage by the protease, μ-calpain, leading to defective repair of DNA ICLs in telomeric and non-telomeric DNA. Knockdown of μ-calpain in FA cells by μ-calpain siRNA results in restoration of αSpII levels to normal and repair of DNA ICLs in telomeric and non-telomeric DNA, demonstrating the importance of αSpII stability in the repair process. It is hypothesized that there is a mechanistic link between excessive cleavage of αSpII by μ-calpain and defective DNA ICL repair in FA and that FA proteins, which are deficient in FA, play a key role in maintaining the stability of αSpII and preventing its cleavage by μ-calpain. All of these events are proposed to be important key factors involved in the pathophysiology of FA and suggest new avenues for potential therapeutic intervention.
Collapse
|
2
|
Jyothy A, Hussain J, S SC, Chandraprabha VR, Nair MG, Vasudevan S, Sreedharan H, Abraham B, Maliekal TT, Natarajan K, Sengupta S. α-Fodrin-CENP-E interaction is critical for pancreatic cancer progression and drug response. Cell Cycle 2025:1-25. [PMID: 40211684 DOI: 10.1080/15384101.2025.2485837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 04/15/2025] Open
Abstract
α-Fodrin, a known scaffolding protein for cytoskeleton stabilization, performs various functions including cell adhesion, cell motility, DNA repair and apoptosis. Based on our previous results revealing its role in mitosis in glioblastoma, we have examined its effect in pancreatic cancer, which is often linked to mitotic aberrations including aneuploidy and chromosome instability. Here, we show that the expression of α-Fodrin increases in pancreatic adenocarcinoma tissues compared to its normal counterpart, suggesting its tumor promoting role. shRNA-mediated knock-down of α-Fodrin significantly reduces the xenograft growth in immunocompromised mice underscoring the importance of α-Fodrin in tumor progression. CENP-E (centromere-associated protein E) is a motor protein essential for chromosomal alignment and segregation during mitosis. We have found that α-Fodrin interacts with CENP-E to recruit it to the kinetochore and depletion of α-Fodrin has a crucial role in controlling aneuploidy. As these mitotic defects can lead to apoptosis, we have further evaluated the activation of possible upstream pathways. Paclitaxel, a chemotherapeutic agent that stabilizes microtubules, disrupts mitosis and induces apoptosis. We found that Paclitaxel triggered stronger activation of JNK, ERK, and P38 MAPKs, altered BCL2/BAX ratios, cytochrome C release causing increased apoptosis in α-Fodrin knockdown cells compared to cells with wild-type α-Fodrin. This enhanced sensitivity to paclitaxel is consistent with improved survival in pancreatic cancer patients with low α-Fodrin (SPTAN1) and low CENP-E expression compared to poor prognosis with high expressions of both the genes. Taken together, this study provides the molecular mechanism by which α-Fodrin - CENP-E axis regulates pancreatic cancer progression and drug response.
Collapse
Affiliation(s)
- Athira Jyothy
- Department of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, India
| | - Julfequar Hussain
- Department of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sharanya C S
- Transdisciplinary Biology Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | - Madhumathy G Nair
- Division of Molecular Medicine, St. John's Research Institute, St John's Medical College, Bangalore, India
| | - Smreti Vasudevan
- Research Department, Rajiv Gandhi Cancer Institute and Research, Delhi, India
| | - Hariharan Sreedharan
- Laboratory of Cytogenetics and Molecular Diagnostics, Regional Cancer Centre, Thiruvananthapuram, India
| | - Betty Abraham
- Department of Pathology, DDRC SRL Diagnostics private limited, Thiruvananthapuram, India
| | - Tessy Thomas Maliekal
- Department of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Kathiresan Natarajan
- Transdisciplinary Biology Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Suparna Sengupta
- Department of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
3
|
Rui T, Zhu K, Mao Z, Wu J, Pan Y, Ye Q, Chen C, Xiang A, Guo J, Tang N, Zhang J, Zheng S, Liu J, Xu X. A Novel tRF, HCETSR, Derived From tRNA-Glu/TTC, Inhibits HCC Malignancy by Regulating the SPBTN1-catenin Complex Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415229. [PMID: 39921434 PMCID: PMC11967833 DOI: 10.1002/advs.202415229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/20/2025] [Indexed: 02/10/2025]
Abstract
tRNA-derived fragments (tRFs), a novel class of small non-coding RNAs cleaved from transfer RNAs, have been implicated in tumor regulation. In this study, the role of a specific tRF, HCETSR is investigated, which is significantly downregulated in hepatocellular carcinoma (HCC) and correlates with advanced tumor burden and higher HCC mortality. Functional analyses revealed that HCETSR inhibits HCC malignancy and serves as an independent predictor of poor prognosis. Mechanistically, a novel SPTBN1/catenin complex axis regulated by HCETSR is identified. HCETSR binds to a critical domain of SPTBN1, disrupting its interaction with the catenin complex (comprising β-catenin, α-catenin, and P120-catenin), and facilitates the transfer of the catenin complex from the cell membrane to the nucleus. Specifically, HCETSR decreases the proteasomal degradation of β-catenin and inhibits the synthesis of nascent β-catenin. Furthermore, HCETSR suppresses the transcriptional activity of LEF1 through P120-catenin rather than α-catenin, thereby reducing β-catenin's influence on LEF1 activity. It is demonstrated that HCETSR is spliced from tRNA-Glu/TTC. The biogenesis of HCETSR and tRNA-Glu/TTC is regulated by the spliceosome and Dicer1. In conclusion, These findings suggest that HCETSR, derived from tRNA-Glu/TTC, inhibits HCC malignancy via modulation of the SPTBN1/catenin axis and may represent a promising prognostic marker and therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Tao Rui
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
- The Center for Integrated Oncology and Precision MedicineAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310003China
| | - Kangbei Zhu
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
| | - Zonglei Mao
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
| | - Jiaping Wu
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
- The Center for Integrated Oncology and Precision MedicineAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310003China
| | - Yi Pan
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
- The Center for Integrated Oncology and Precision MedicineAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310003China
| | - Qianwei Ye
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
- The Center for Integrated Oncology and Precision MedicineAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310003China
| | - Cong Chen
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
- The Center for Integrated Oncology and Precision MedicineAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310003China
| | - Aizhai Xiang
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
| | - Jufeng Guo
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
| | - Ning Tang
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
| | - Jing Zhang
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
| | - Shusen Zheng
- Department of SurgeryCollaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseasesthe First Affiliated HospitalZhejiang University School of MedicineZhejiang University HangzhouHangzhou310003China
| | - Jian Liu
- Department of SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhou310003China
| | - Xiao Xu
- School of Clinical MedicineHangzhou Medical CollegeHangzhou310059China
- Institute of Translational MedicineZhejiang UniversityHangzhou310000China
- NHC Key Laboratory of Combined Multi‐Organ TransplantationInstitute of Organ TransplantationZhejiang UniversityHangzhou310003China
| |
Collapse
|
4
|
Jin L, Qin Y, Zhao Y, Zhou X, Zeng Y. Endothelial cytoskeleton in mechanotransduction and vascular diseases. J Biomech 2025; 182:112579. [PMID: 39938443 DOI: 10.1016/j.jbiomech.2025.112579] [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: 11/27/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The cytoskeleton is an important structural component that regulates various aspects of cell morphology, movement, and intracellular signaling. It plays a pivotal role in the cellular response to biomechanical stimuli, particularly in endothelial cells, which are critical for vascular homeostasis and the pathogenesis of cardiovascular diseases. Mechanical forces, such as shear and tension, activate intracellular signaling cascades that regulate transcription, translation, and cellular behaviors. Despite extensive research into cytoskeletal functions, the precise mechanisms by which the cytoskeleton transduces mechanical signals remain incompletely understood. This review focuses on the role of cytoskeletal components in membrane, cytoplasm, and nucleus in mechanotransduction, with an emphasis on their structure, mechanical and biological behaviors, dynamic interactions, and response to mechanical forces. The collaboration between membrane cytoskeleton, cytoplasmic cytoskeleton, and nucleoskeleton is indispensable for endothelial cells to respond to mechanical stimuli. Understanding their mechanoresponsive mechanisms is essential for advancing therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yunran Zhao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China.
| |
Collapse
|
5
|
De Winter J, Van de Vondel L, Ermanoska B, Monticelli A, Isapof A, Cohen E, Stojkovic T, Hackman P, Johari M, Palmio J, Waldrop MA, Meyer AP, Nicolau S, Flanigan KM, Töpf A, Diaz-Manera J, Straub V, Longman C, McWilliam CA, Orbach R, Verma S, Laine R, Donkervoort S, Bonnemann CG, Rebelo A, Züchner S, Grider T, Shy ME, Maystadt I, Demurger F, Cairns A, Beecroft S, Folland C, De Ridder W, Ravenscroft G, Bonne G, Udd B, Baets J. Heterozygous loss-of-function variants in SPTAN1 cause an early childhood onset distal myopathy. Genet Med 2025; 27:101399. [PMID: 40023774 DOI: 10.1016/j.gim.2025.101399] [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: 10/15/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
PURPOSE Heterozygous pathogenic variants in SPTAN1 cause a diverse spectrum of neurogenetic disorders ranging from peripheral and central nervous system involvement to complex syndromic presentations. We set out to investigate the role of SPTAN1 in genetically unsolved hereditary myopathies. METHODS Through international collaboration we identified 14 families with distal weakness and heterozygous SPTAN1 loss-of-function variants. Clinical data, electrophysiology, muscle computed tomography or magnetic resonance imaging, and muscle biopsy findings were collected and standardized. SPTAN1 protein, messenger RNA expression analysis and copy DNA sequencing was performed on muscle tissue from 2 participants. RESULTS Five families showed autosomal dominant mode of inheritance, whereas in 9 patients the variant was shown to be de novo, including 2 pairs of monozygotic twins. In 2 families, further segregation analysis was not possible. All affected participants presented with early childhood-onset distal weakness and foot abnormalities. Muscle magnetic resonance imaging or computed tomography in 10 patients showed fatty infiltration of the distal lower limb anterior compartment and/or selective involvement of the extensor hallucis longus muscle. Muscle biopsy revealed myopathic changes in 7 patients. Finally, we provide proof for nonsense-mediated decay in muscle tissue derived from 2 patients. CONCLUSION We present evidence linking heterozygous SPTAN1 loss-of-function variants to childhood-onset distal myopathy in 14 unrelated families.
Collapse
Affiliation(s)
- Jonathan De Winter
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Liedewei Van de Vondel
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Biljana Ermanoska
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Alice Monticelli
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Arnaud Isapof
- Centre de Référence des Maladies Neuromusculaires Nord-Est-Ile de France, Hôpital Armand Trousseau, APHP, Paris, France
| | - Enzo Cohen
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Tanya Stojkovic
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France; Centre de Référence des Maladies Neuromusculaires Nord-Est-Ile de France, Hôpital Pitié-Salpêtrière, Institut de Myologie, APHP, Paris, France
| | - Peter Hackman
- Folkhälsan Research Center, Helsinki, Finland and Medicum, University of Helsinki, Helsinki, Finland
| | - Mridul Johari
- Folkhälsan Research Center, Helsinki, Finland and Medicum, University of Helsinki, Helsinki, Finland; Centre for Medical Research, University of Western Australia, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Johanna Palmio
- Tampere Neuromuscular Center, Tampere University and Tampere University Hospital Tampere, Finland
| | - Megan A Waldrop
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology, Wexner Medical Center, Ohio State University, Columbus OH
| | - Alayne P Meyer
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH; Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH
| | - Stefan Nicolau
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH; Department of Pediatrics, The Ohio State University, Columbus, OH
| | - Kevin M Flanigan
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology, Wexner Medical Center, Ohio State University, Columbus OH
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Jordi Diaz-Manera
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Cheryl Longman
- West Scotland Regional Genetics Service, Queen Elizabeth University Hospital, Glasgow, Scotland
| | - Catherine A McWilliam
- West Scotland Regional Genetics Service, Queen Elizabeth University Hospital, Glasgow, Scotland
| | - Rotem Orbach
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Sumit Verma
- Department of Pediatrics and Neurology, Emory University School of Medicine, Atlanta, GA
| | - Regina Laine
- Department of Neurology, Boston Children's Hospital, Boston, MA
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Carsten G Bonnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Adriana Rebelo
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Tiffany Grider
- Neurology, The University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, IA
| | - Michael E Shy
- Neurology, The University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, IA
| | - Isabelle Maystadt
- Centre de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium; URPHYM, Department of Medicine, UNamur, Namur, Belgium
| | | | - Anita Cairns
- Neurosciences Department, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Sarah Beecroft
- Pawsey Supercomputing Research Centre, Kensington, WA, Australia
| | - Chiara Folland
- Centre for Medical Research, University of Western Australia, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Willem De Ridder
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Gina Ravenscroft
- Centre for Medical Research, University of Western Australia, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Gisèle Bonne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland and Medicum, University of Helsinki, Helsinki, Finland; Tampere Neuromuscular Center, Tampere University and Tampere University Hospital Tampere, Finland
| | - Jonathan Baets
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium.
| |
Collapse
|
6
|
Lu PN, Melton C, Dupont B, Jones JR, Abidi F, Rose A, Patterson WG, Lyons MJ, Flanagan-Steet H. Loss of Function SPTAN1 Variants Result in Ataxia and Intellectual Disability. Clin Genet 2025. [PMID: 39988451 DOI: 10.1111/cge.14732] [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: 11/25/2024] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025]
Abstract
SPTAN1 mutations have been reported in association with autosomal dominant early infantile epileptic encephalopathy 5. Individuals present with early-onset seizures and profound intellectual disability. Recent reports suggest a wider spectrum with later-onset seizures and milder developmental delay. Here we describe two patients with loss-of-function variants in SPTAN1. One patient has ataxia and mild intellectual disability stemming from a de novo homozygous p.(Gln1448Pro) variant associated with uniparental disomy 9. The second patient, carrying a heterozygous p.(Asn1839del) allele, exhibits more substantial motor issues, developmental delay, and seizures. Ectopically expressed wild-type or variant-containing forms of sptan1 in zebrafish indicate both variants create loss-of-function alleles, with the p.(Gln1448Pro) likely being hypomorphic. This conclusion is supported by reduced protein abundance and localization of Sptan1 variants in axons of developing embryos. Further, unlike wild-type sptan1, analysis of the p.(Gln1448Pro) variant showed it failed to restore voltage-gated sodium channel localization in sptan1-null axons. Additional behavioral analyses show supplementation with the amino acid D-aspartate improved motility in sptan1-null zebrafish, supporting its use for α-II spectrin-associated motor dysfunction.
Collapse
Affiliation(s)
- Po-Nien Lu
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Chandler Melton
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
- UNC Chapel Hill-Department of Psychiatry, Chapel Hill, North Carolina, USA
| | - Barbara Dupont
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Julie R Jones
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Fatima Abidi
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Aubrey Rose
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | | | | | | |
Collapse
|
7
|
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
|
8
|
Ghisleni A, Bonilla-Quintana M, Crestani M, Lavagnino Z, Galli C, Rangamani P, Gauthier NC. Mechanically induced topological transition of spectrin regulates its distribution in the mammalian cell cortex. Nat Commun 2024; 15:5711. [PMID: 38977673 PMCID: PMC11231315 DOI: 10.1038/s41467-024-49906-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
The cell cortex is a dynamic assembly formed by the plasma membrane and underlying cytoskeleton. As the main determinant of cell shape, the cortex ensures its integrity during passive and active deformations by adapting cytoskeleton topologies through yet poorly understood mechanisms. The spectrin meshwork ensures such adaptation in erythrocytes and neurons by adopting different organizations. Erythrocytes rely on triangular-like lattices of spectrin tetramers, whereas in neurons they are organized in parallel, periodic arrays. Since spectrin is ubiquitously expressed, we exploited Expansion Microscopy to discover that, in fibroblasts, distinct meshwork densities co-exist. Through biophysical measurements and computational modeling, we show that the non-polarized spectrin meshwork, with the intervention of actomyosin, can dynamically transition into polarized clusters fenced by actin stress fibers that resemble periodic arrays as found in neurons. Clusters experience lower mechanical stress and turnover, despite displaying an extension close to the tetramer contour length. Our study sheds light on the adaptive properties of spectrin, which participates in the protection of the cell cortex by varying its densities in response to key mechanical features.
Collapse
Affiliation(s)
- Andrea Ghisleni
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Mayte Bonilla-Quintana
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, USA
| | - Michele Crestani
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Laboratory of Applied Mechanobiology, Department for Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Zeno Lavagnino
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Camilla Galli
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Humanitas Cardio Center, IRCCS Humanitas Research Hospital, Rozzano (Milan, Italy
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, USA.
| | - Nils C Gauthier
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
9
|
Jin J, Guo Q, Yan Z. The Role of Lutheran/Basal Cell Adhesion Molecule in Hematological Diseases and Tumors. Int J Mol Sci 2024; 25:7268. [PMID: 39000374 PMCID: PMC11242806 DOI: 10.3390/ijms25137268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Cell adhesion is a dynamic process that plays a fundamental role in cell proliferation, maintenance, differentiation, and migration. Basal cell adhesion molecule (BCAM), also known as Lutheran (Lu), belongs to the immunoglobulin superfamily of cell adhesion molecules. Lu/BCAM, which is widely expressed in red blood cells, endothelial cells, smooth muscle cells and epithelial cells across various tissues, playing a crucial role in many cellular processes, including cell adhesion, cell motility and cell migration. Moreover, Lu/BCAM, dysregulated in many diseases, such as blood diseases and various types of cancer, may act as a biomarker and target for the treatment of these diseases. This review explores the significance of Lu/BCAM in cell adhesion and its potential as a novel target for treating hematological diseases and tumors.
Collapse
Affiliation(s)
| | | | - Zhibin Yan
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (J.J.); (Q.G.)
| |
Collapse
|
10
|
Guo R, Yang L, Pan Y, Shen J, Zhao F. Association between threat-related adverse childhood experiences and chronic lung diseases in a middle and older aged population: A cross-sectional and longitudinal study in China. J Psychosom Res 2024; 182:111692. [PMID: 38735102 DOI: 10.1016/j.jpsychores.2024.111692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
OBJECTIVES We investigated the association between threat-related adverse childhood experiences (ACEs) and the risk of chronic lung diseases (CLDs). METHODS The data used for this study were extracted from the China Health and Retirement Longitudinal Study (CHARLS), a nationally representative survey of respondents recruited from 450 villages/urban communities in 28 provinces. Threat-related ACEs were constructed using five adverse factors: household substance abuse, physical abuse, domestic violence, unsafe neighbourhood, and bullying). Participants were divided into three groups according to their number of threat-related ACEs at baseline and at follow-up. The association between threat-related ACEs and CLD prevalence in the cross-sectional study was calculated using logistic regression models. The association between threat-related ACEs and CLD onset was evaluated using Cox proportional regression models in the cohort study. Potential confounders were considered in both the cross-sectional and cohort studies. RESULTS The CLD prevalence in the total population, no exposure group, exposure to one threat-related ACE, and exposure to at least two threat-related ACEs were 10.07% (1320/13104), 9.20% (665/7232), 10.89% (421/3865), and 11.66% (234/2007), respectively. Exposure to one threat-related ACE (OR: 1.23, 95% CI: 1.07-1.41) and exposure to at least two threat-related ACEs (OR: 1.31, 95% CI: 1.11-1.55) were significantly associated with higher CLD prevalence rates. The cohort study included 11,645 participants. During the 7-year follow-up, 738 CLD incidents were identified. Similarly, exposure to one threat-related ACE (HR: 1.20, 95% CI: 1.01-1.43) and at least two threat-related ACEs (HR: 1.64, 95% CI: 1.35-2.00) were significantly associated with a higher CLD incidence risk. CONCLUSIONS Exposure to threat-related ACEs was significantly associated with a higher CLD prevalence risk and onset. It is crucial to identify individuals who have encountered childhood threats and prioritise the monitoring of their pulmonary function.
Collapse
Affiliation(s)
- Run Guo
- Department of General Medicine, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, China
| | - Lihong Yang
- Department of General Medicine, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, China
| | - Yunfei Pan
- Department of General Medicine, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, China
| | - Jiaying Shen
- Department of General Medicine, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, China
| | - Feimin Zhao
- Department of General Medicine, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, China.
| |
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: 3] [Impact Index Per Article: 3.0] [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
|
Benz PM, Frömel T, Laban H, Zink J, Ulrich L, Groneberg D, Boon RA, Poley P, Renne T, de Wit C, Fleming I. Cardiovascular Functions of Ena/VASP Proteins: Past, Present and Beyond. Cells 2023; 12:1740. [PMID: 37443774 PMCID: PMC10340426 DOI: 10.3390/cells12131740] [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: 04/26/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Actin binding proteins are of crucial importance for the spatiotemporal regulation of actin cytoskeletal dynamics, thereby mediating a tremendous range of cellular processes. Since their initial discovery more than 30 years ago, the enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) family has evolved as one of the most fascinating and versatile family of actin regulating proteins. The proteins directly enhance actin filament assembly, but they also organize higher order actin networks and link kinase signaling pathways to actin filament assembly. Thereby, Ena/VASP proteins regulate dynamic cellular processes ranging from membrane protrusions and trafficking, and cell-cell and cell-matrix adhesions, to the generation of mechanical tension and contractile force. Important insights have been gained into the physiological functions of Ena/VASP proteins in platelets, leukocytes, endothelial cells, smooth muscle cells and cardiomyocytes. In this review, we summarize the unique and redundant functions of Ena/VASP proteins in cardiovascular cells and discuss the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Peter M. Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
| | - Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Lea Ulrich
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Dieter Groneberg
- Institute of Physiology I, University of Würzburg, 97070 Würzburg, Germany
| | - Reinier A. Boon
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
- Centre of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe-University, 60596 Frankfurt am Main, Germany
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Centre, 1081 HZ Amsterdam, The Netherlands
| | - Philip Poley
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 VN51 Dublin, Ireland
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
| |
Collapse
|
13
|
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: 22] [Impact Index Per Article: 11.0] [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
|
14
|
Wu D, Pandupuspitasari NS, Zhang K, Tang Y, Khan FA, Li H, Huang C, Sun F. Cytoskeletal orchestration of glucose uptake in Sertoli cell to support efferocytosis of apoptotic germ cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119434. [PMID: 36716822 DOI: 10.1016/j.bbamcr.2023.119434] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/28/2023]
Abstract
Efferocytosis of non-viable germ cells by Sertoli cells (SCs) constitutes a sentinel for testis homeostasis, yet how SCs signal for the metabolic and cytoskeletal adaption to this energetically costly process remains unexplored. Spectrin is membrane-associated periodic skeleton assembled into an actin-spectrin-based cytoskeletal structure with an interaction with glucose transporter Glut1. The contribution of spectrin to glucose uptake and efferocytosis is unknown. In this study, we identified a cross-regulation between glucose metabolism and efferocytosis in SCs. Pharmacological or genetic inhibition of glucose uptake or glycolysis compromises efferocytosis activity. We further found that βII-spectrin is a hitherto unappreciated regulator of glucose metabolism and cytoskeletal architecture. βII-spectrin deficiency impairs glucose uptake and lactate production in SCs. Moreover, a defective assembly of cytoskeleton and a loss of blood-testis barrier integrity are also featured by SCs deficient in βII-spectrin. The disruption in glucose metabolism and cytoskeletal organization synergistically lead to a defective efferocytosis. In vivo siRNA-mediated targeting of βII-spectrin in testis causes an obvious morphological aberration in seminiferous epithelium with the presence of exfoliated germ cells and multinucleated giant cells. Importantly, a decrease in expression of αII/βII-spectrin was observed in testes of Adjudin-induced infertility model. By exploring the functional relevance of βII-spectrin to the metabolic and cytoskeletal regulation of efferocytosis, our study proposes a potential link between βII-spectrin deregulation and male infertility.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Nuruliarizki Shinta Pandupuspitasari
- Faculty of Animal and Agricultural Sciences, Universitas Diponegoro, Semarang 1269, Indonesia; Department of Biological Engineering, Massachusetts Institute of Technology, MA 02139, USA
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Yuan Tang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Faheem Ahmed Khan
- Department of Zoology, Faculty of Science and Technology, University of Central Punjab, Lahore 54782, Pakistan; Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat 10340, Indonesia
| | - Haitao Li
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
15
|
Progressive Ataxia, Memory Impairments, and Seizure Episodes in Spna2 R1098Q Mouse Variant Affecting Alpha II Spectrin's Scaffold Stability. Brain Sci 2023; 13:brainsci13020261. [PMID: 36831804 PMCID: PMC9953789 DOI: 10.3390/brainsci13020261] [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: 01/03/2023] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
SPTAN1 spectrinopathies refer to a group of rare, inherited diseases associated with damage to non-erythrocytic α-II spectrin (α-II). They are linked to a range of mild to severe neuropathologies of the central and peripheral nervous systems, such as early infantile epileptic encephalopathy type 5, cerebellar ataxia, inherited peripheral neuropathy, and spastic paraplegia. Modeling human SPTAN1 encephalopathies in laboratory animals has been challenging partially because no haploinsufficiency-related phenotypes unfold in heterozygous Spna2 deficient mice nor stable transgenic lines of mice mimicking missense human SPTAN1 mutations have been created to date. Here, we assess the motor and memory performance of a dominant-negative murine Spna2 (SPTAN1) variant carrying a spontaneous point mutation replacing an arginine 1098 in the repeat 10th of α-II with the glutamine (R1098Q). By comparing groups of heterozygous R1098Q mice at different ages, we find evidence for progressive ataxia, and age-related deterioration of motor performance and muscle strength. We also document stress-induced, long-lasting seizure episodes of R1098Q mice and their poor performance in novel object recognition memory tests. Overall, we propose that the complexity of neuropathology-related phenotypes presented by the R1098Q mice recapitulates a number of symptoms observed in human patients carrying SPTAN1 mutations affecting α-II scaffold stability. This makes the R1098Q mice a valuable animal model for preclinical research.
Collapse
|
16
|
Sreeja JS, Jyothy A, Nellikka RK, Ghorai S, Riya PA, James J, Sengupta S. The centrosomal recruitment of γ-tubulin and its microtubule nucleation activity is α-fodrin guided. Cell Cycle 2023; 22:361-378. [PMID: 36082994 PMCID: PMC9851242 DOI: 10.1080/15384101.2022.2119516] [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: 03/23/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 01/22/2023] Open
Abstract
The regulation and recruitment of γ-TuRCs, the prime nucleator of microtubules, to the centrosome are still thrust areas of research. The interaction of fodrin, a sub-plasmalemmal cytoskeletal protein, with γ-tubulin is a new area of interest. To understand the cellular significance of this interaction, we show that depletion of α-fodrin brings in a significant reduction of γ-tubulin in neural cell centrosomes making it functionally under-efficient. This causes a loss of nucleation ability that cannot efficiently form microtubules in interphase cells and astral microtubules in mitosis. Fluorescence Recovery after Photobleaching (FRAP) experiment implies that α-fodrin is important in the recruitment of γ-tubulin to the centrosome resulting in the aforementioned effects. Further, our experiments indicate that the interaction of α-fodrin with certain pericentriolar matrix proteins such as Pericentrin and CDK5RAP2 are crucial for the recruitment of γ-tubulin to the centrosome. Earlier we reported that α-fodrin limits the nucleation potential of γ-TuRC. In that context, this study suggests that α-fodrin is a γ-tubulin recruiting protein to the centrosome thus preventing cytoplasmic microtubule nucleation and thereby compartmentalizing the process to the centrosome for maximum efficiency. Summary statementα-fodrin is a γ-tubulin interacting protein that controls the process of γ-tubulin recruitment to the centrosome and thereby regulates the microtubule nucleation capacity spatially and temporally.
Collapse
Affiliation(s)
- Jamuna S. Sreeja
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Athira Jyothy
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, India
| | - Rohith Kumar Nellikka
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sayan Ghorai
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Paul Ann Riya
- Regenerative Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Jackson James
- Regenerative Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Suparna Sengupta
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
17
|
Ghisleni A, Bonilla-Quintana M, Crestani M, Fukuzawa A, Rangamani P, Gauthier N. Mechanically induced topological transition of spectrin regulates its distribution in the mammalian cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.02.522381. [PMID: 36712133 PMCID: PMC9881866 DOI: 10.1101/2023.01.02.522381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The cell cortex is a dynamic assembly that ensures cell integrity during passive deformation or active response by adapting cytoskeleton topologies with poorly understood mechanisms. The spectrin meshwork ensures such adaptation in erythrocytes and neurons. Erythrocytes rely on triangular-like lattices of spectrin tetramers, which in neurons are organized in periodic arrays. We exploited Expansion Microscopy to discover that these two distinct topologies can co-exist in other mammalian cells such as fibroblasts. We show through biophysical measurements and computational modeling that spectrin provides coverage of the cortex and, with the intervention of actomyosin, erythroid-like lattices can dynamically transition into condensates resembling neuron-like periodic arrays fenced by actin stress fibers. Spectrin condensates experience lower mechanical stress and turnover despite displaying an extension close to the contour length of the tetramer. Our study sheds light on the adaptive properties of spectrin, which ensures protection of the cortex by undergoing mechanically induced topological transitions.
Collapse
|
18
|
Alterations in Cerebellar Microtubule Cytoskeletal Network in a ValproicAcid-Induced Rat Model of Autism Spectrum Disorders. Biomedicines 2022; 10:biomedicines10123031. [PMID: 36551785 PMCID: PMC9776106 DOI: 10.3390/biomedicines10123031] [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: 10/13/2022] [Revised: 11/15/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental diseases characterised by deficits in social communication, restricted interests, and repetitive behaviours. The growing body of evidence points to a role for cerebellar changes in ASD pathology. Some of the findings suggest that not only motor problems but also social deficits, repetitive behaviours, and mental inflexibility associated with ASD are connected with damage to the cerebellum. However, the understanding of this brain structure's functions in ASD pathology needs future investigations. Therefore, in this study, we generated a rodent model of ASD through a single prenatal administration of valproic acid (VPA) into pregnant rats, followed by cerebellar morphological studies of the offspring, focusing on the alterations of key cytoskeletal elements. The expression (Western blot) of α/β-tubulin and the major neuronal MT-associated proteins (MAP) such as MAP-Tau and MAP1B, MAP2, MAP6 (STOP) along with actin-crosslinking αII-spectrin and neurofilament light polypeptide (NF-L) was investigated. We found that maternal exposure to VPA induces a significant decrease in the protein levels of α/β-tubulin, MAP-Tau, MAP1B, MAP2, and αII-spectrin. Moreover, excessive MAP-Tau phosphorylation at (Ser396) along with key Tau-kinases activation was indicated. Immunohistochemical staining showed chromatolysis in the cerebellum of autistic-like rats and loss of Purkinje cells shedding light on one of the possible molecular mechanisms underpinning neuroplasticity alterations in the ASD brain.
Collapse
|
19
|
The spectrin cytoskeleton integrates endothelial mechanoresponses. Nat Cell Biol 2022; 24:1226-1238. [PMID: 35817960 DOI: 10.1038/s41556-022-00953-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Physiological blood flow induces the secretion of vasoactive compounds, notably nitric oxide, and promotes endothelial cell elongation and reorientation parallel to the direction of applied shear. How shear is sensed and relayed to intracellular effectors is incompletely understood. Here, we demonstrate that an apical spectrin network is essential to convey the force imposed by shear to endothelial mechanosensors. By anchoring CD44, spectrins modulate the cell surface density of hyaluronan and sense and translate shear into changes in plasma membrane tension. Spectrins also regulate the stability of apical caveolae, where the mechanosensitive PIEZO1 channels are thought to reside. Accordingly, shear-induced PIEZO1 activation and the associated calcium influx were absent in spectrin-deficient cells. As a result, cell realignment and flow-induced endothelial nitric oxide synthase stimulation were similarly dependent on spectrin. We conclude that the apical spectrin network is not only required for shear sensing but also transmits and distributes the resulting tensile forces to mechanosensors that elicit protective and vasoactive responses.
Collapse
|
20
|
Outer Hair Cell Function is Normal in βV Spectrin Knockout Mice. Hear Res 2022; 423:108564. [DOI: 10.1016/j.heares.2022.108564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 05/31/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022]
|
21
|
Leitner DF, Kanshin E, Askenazi M, Siu Y, Friedman D, Devore S, Jones D, Ueberheide B, Wisniewski T, Devinsky O. Pilot study evaluating everolimus molecular mechanisms in tuberous sclerosis complex and focal cortical dysplasia. PLoS One 2022; 17:e0268597. [PMID: 35587487 PMCID: PMC9119437 DOI: 10.1371/journal.pone.0268597] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/02/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) and some focal cortical dysplasias (FCDs) are associated with dysfunctional mTOR signaling, resulting in increased cell growth and ribosomal S6 protein phosphorylation (phospho-S6). mTOR inhibitors can reduce TSC tumor growth and seizure frequency, and preclinical FCD studies indicate seizure suppression. This pilot study evaluated safety of mTOR inhibitor everolimus in treatment resistant (failure of >2 anti-seizure medications) TSC and FCD patients undergoing surgical resection and to assess mTOR signaling and molecular pathways. METHODS AND FINDINGS We evaluated everolimus in 14 treatment resistant epilepsy patients undergoing surgical resection (4.5 mg/m2 daily for 7 days; n = 4 Active, mean age 18.3 years, range 4-26; n = 10, Control, mean age 13.1, range 3-45). Everolimus was well tolerated. Mean plasma everolimus in Active participants were in target range (12.4 ng/ml). Brain phospho-S6 was similar in Active and Control participants with a lower trend in Active participants, with Ser235/236 1.19-fold (p = 0.67) and Ser240/244 1.15-fold lower (p = 0.66). Histologically, Ser235/236 was 1.56-fold (p = 0.37) and Ser240/244 was 5.55-fold lower (p = 0.22). Brain proteomics identified 11 proteins at <15% false discovery rate associated with coagulation system (p = 1.45x10-9) and acute phase response (p = 1.23x10-6) activation. A weighted gene correlation network analysis (WGCNA) of brain proteomics and phospho-S6 identified 5 significant modules. Higher phospho-S6 correlated negatively with cellular respiration and synaptic transmission and positively with organophosphate metabolic process, nuclear mRNA catabolic process, and neuron ensheathment. Brain metabolomics identified 14 increased features in Active participants, including N-acetylaspartylglutamic acid. Plasma proteomics and cytokine analyses revealed no differences. CONCLUSIONS Short-term everolimus before epilepsy surgery in TSC and FCD resulted in no adverse events and trending lower mTOR signaling (phospho-S6). Future studies should evaluate implications of our findings, including coagulation system activation and everolimus efficacy in FCD, in larger studies with long-term treatment to better understand molecular and clinical effects. CLINICAL TRIALS REGISTRATION ClinicalTrials.gov NCT02451696.
Collapse
Affiliation(s)
- Dominique F. Leitner
- Comprehensive Epilepsy Center, New York University School of Medicine, New York, New York, United States of America
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies, NYU School of Medicine, New York, New York, United States of America
| | - Manor Askenazi
- Biomedical Hosting LLC, Arlington, Massachusetts, United States of America
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Yik Siu
- Metabolomics Core Resource Laboratory, New York University School of Medicine, New York, New York, United States of America
| | - Daniel Friedman
- Comprehensive Epilepsy Center, New York University School of Medicine, New York, New York, United States of America
| | - Sasha Devore
- Comprehensive Epilepsy Center, New York University School of Medicine, New York, New York, United States of America
| | - Drew Jones
- Metabolomics Core Resource Laboratory, New York University School of Medicine, New York, New York, United States of America
| | - Beatrix Ueberheide
- Proteomics Laboratory, Division of Advanced Research Technologies, NYU School of Medicine, New York, New York, United States of America
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, New York, United States of America
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, New York, United States of America
- Department of Psychiatry, New York University School of Medicine, New York, New York, United States of America
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, New York University School of Medicine, New York, New York, United States of America
| |
Collapse
|
22
|
Li S, Liu T, Li K, Bai X, Xi K, Chai X, Mi L, Li J. Spectrins and human diseases. Transl Res 2022; 243:78-88. [PMID: 34979321 DOI: 10.1016/j.trsl.2021.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 11/18/2022]
Abstract
Spectrin, as one of the major components of a plasma membrane-associated cytoskeleton, is a cytoskeletal protein composed of the modular structure of α and β subunits. The spectrin-based skeleton is essential for preserving the integrity and mechanical characteristics of the cell membrane. Moreover, spectrin regulates a variety of cell processes including cell apoptosis, cell adhesion, cell spreading, and cell cycle. Dysfunction of spectrins is implicated in various human diseases including hemolytic anemia, neurodegenerative diseases, ataxia, heart diseases, and cancers. Here, we briefly discuss spectrins function as well as the clinical manifestations and currently known molecular mechanisms of human diseases related to spectrins, highlighting that strategies for targeting regulation of spectrins function may provide new avenues for therapeutic intervention for these diseases.
Collapse
Affiliation(s)
- Shan Li
- The First School of Clinical Medicine, Lanzhou University, Gansu, China
| | - Ting Liu
- The First School of Clinical Medicine, Lanzhou University, Gansu, China
| | - Kejing Li
- The First School of Clinical Medicine, Lanzhou University, Gansu, China
| | - Xinyi Bai
- The First School of Clinical Medicine, Lanzhou University, Gansu, China
| | - Kewang Xi
- The First School of Clinical Medicine, Lanzhou University, Gansu, China
| | - Xiaojing Chai
- Central Laboratory, The First Hospital of Lanzhou University, Gansu, China
| | - Leyuan Mi
- The First School of Clinical Medicine, Lanzhou University, Gansu, China; Clinical Laboratory Center, Gansu Provincial Maternity and Child Care Hospital, Gansu, China
| | - Juan Li
- Gansu Key Laboratory of Genetic Study of Hematopathy, The First Hospital of Lanzhou University, Gansu, China; Central Laboratory, The First Hospital of Lanzhou University, Gansu, China.
| |
Collapse
|
23
|
Skrzymowska J, Zalas M, Goszczyński TM, Miazek A. An alpha II spectrin mutant peptide with unstable scaffold structure and increased sensitivity to calpain cleavage. Biochem Biophys Res Commun 2021; 581:68-73. [PMID: 34656850 DOI: 10.1016/j.bbrc.2021.10.021] [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: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022]
Abstract
A spontaneous missense mutation in the alpha II spectrin (αII) gene, replacing a highly conserved arginine 1098 with the glutamine (R1098Q), causes progressive neurodegeneration in heterozygous mutant mice. The molecular mechanism underlying this phenotype is unknown but the accumulation of 150kD αII breakdown products in brains of homozygous mutant embryos suggests an imbalance in the substrate level control of αII cleavage by calpains. This is further supported by in silico simulation predicting unmasked calpain target site and increased spectrin scaffold bending and flexibility of R1098Q mutant peptide. Here, using spectroscopic and in situ enzymatic techniques, we aimed at obtaining direct experimental support for the impact of R1098Q mutation on the αII stability and its propensity for calpain-mediated degradation. Thermal circular dichroism analyses performed on recombinant wildtype and R1098Q mutant αII peptides, composed of spectrin repeat 9-10 revealed that although both had very similar secondary structure contents, thermal stability curve profiles varied and the observed midpoint of the unfolding transition (Tm) was 5.5 °C lower for the R1098Q peptide. Yet, the dynamic light scattering profiles of both peptides closely overlapped, implying the same thermal propensity to aggregate. Calpain digestion of plate-bound αII peptides with and without added calmodulin revealed an enhancement of the R1098Q peptide digestion rate relative to WT control. In summary, these results support the unstable scaffold structure of the R1098Q peptide as contributing to its enhanced intrinsic sensitivity to calpain and suggest physiologic relevance of a proper calpain/spectrin balance in preventing neurodegeneration.
Collapse
Affiliation(s)
- Joanna Skrzymowska
- Department of Tumor Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Michał Zalas
- Department of Tumor Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Tomasz M Goszczyński
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Arkadiusz Miazek
- Department of Tumor Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland.
| |
Collapse
|
24
|
Essential Role of Sptan1 in Cochlear Hair Cell Morphology and Function Via Focal Adhesion Signaling. Mol Neurobiol 2021; 59:386-404. [PMID: 34708331 PMCID: PMC8786805 DOI: 10.1007/s12035-021-02551-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/31/2021] [Indexed: 12/05/2022]
Abstract
Hearing loss is the most common human sensory deficit. Hearing relies on stereocilia, inserted into the cuticular plate of hair cells (HCs), where they play an important role in the perception of sound and its transmission. Although numerous genes have been associated with hearing loss, the function of many hair cell genes has yet to be elucidated. Herein, we focused on nonerythroid spectrin αII (SPTAN1), abundant in the cuticular plate, surrounding the rootlets of stereocilia and along the plasma membrane. Interestingly, mice with HC-specific Sptan1 knockout exhibited rapid deafness, abnormal formation of stereocilia and cuticular plates, and loss of HCs from middle and apical turns of the cochlea during early postnatal stages. Additionally, Sptan1 deficiency led to the decreased spreading of House Ear Institute-Organ of Corti 1 cells, and induced abnormal formation of focal adhesions and integrin signaling in mouse HCs. Altogether, our findings highlight SPTAN1 as a critical molecule for HC stereocilia morphology and auditory function via regulation of focal adhesion signaling.
Collapse
|
25
|
Fujitani M, Otani Y, Miyajima H. Pathophysiological Roles of Abnormal Axon Initial Segments in Neurodevelopmental Disorders. Cells 2021; 10:2110. [PMID: 34440880 PMCID: PMC8392614 DOI: 10.3390/cells10082110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 11/17/2022] Open
Abstract
The 20-60 μm axon initial segment (AIS) is proximally located at the interface between the axon and cell body. AIS has characteristic molecular and structural properties regulated by the crucial protein, ankyrin-G. The AIS contains a high density of Na+ channels relative to the cell body, which allows low thresholds for the initiation of action potential (AP). Molecular and physiological studies have shown that the AIS is also a key domain for the control of neuronal excitability by homeostatic mechanisms. The AIS has high plasticity in normal developmental processes and pathological activities, such as injury, neurodegeneration, and neurodevelopmental disorders (NDDs). In the first half of this review, we provide an overview of the molecular, structural, and ion-channel characteristics of AIS, AIS regulation through axo-axonic synapses, and axo-glial interactions. In the second half, to understand the relationship between NDDs and AIS, we discuss the activity-dependent plasticity of AIS, the human mutation of AIS regulatory genes, and the pathophysiological role of an abnormal AIS in NDD model animals and patients. We propose that the AIS may provide a potentially valuable structural biomarker in response to abnormal network activity in vivo as well as a new treatment concept at the neural circuit level.
Collapse
Affiliation(s)
- Masashi Fujitani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi 693-8501, Shimane, Japan; (Y.O.); (H.M.)
| | | | | |
Collapse
|
26
|
α-Fodrin in Cytoskeletal Organization and the Activity of Certain Key Microtubule Kinesins. Genes (Basel) 2021; 12:genes12050750. [PMID: 34067543 PMCID: PMC8156673 DOI: 10.3390/genes12050750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 11/17/2022] Open
Abstract
Cortical cytoskeletal proteins are significant in controlling various cellular mechanisms such as migration, cell adhesion, intercellular attachment, cellular signaling, exo- and endocytosis and plasma membrane integrity, stability and flexibility. Our earlier studies involving in vitro and ex vivo approaches led us to identify certain undiscovered characteristics of α-fodrin, a prominent cortical protein. The conventional functions attributed to this protein mainly support the plasma membrane. In the present study, we utilized a global protein expression analysis approach to detect underexplored functions of this protein. We report that downregulation of α-fodrin in glioblastoma cells, U-251 MG, results in upregulation of genes affecting the regulation of the cytoskeleton, cell cycle and apoptosis. Interestingly, certain key microtubule kinesins such as KIF23, KIF2B and KIF3C are downregulated upon α-fodrin depletion, as validated by real-time PCR studies.
Collapse
|
27
|
Regulation of Cardiac Conduction and Arrhythmias by Ankyrin/Spectrin-Based Macromolecular Complexes. J Cardiovasc Dev Dis 2021; 8:jcdd8050048. [PMID: 33946725 PMCID: PMC8146975 DOI: 10.3390/jcdd8050048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
The cardiac conduction system is an extended network of excitable tissue tasked with generation and propagation of electrical impulses to signal coordinated contraction of the heart. The fidelity of this system depends on the proper spatio-temporal regulation of ion channels in myocytes throughout the conduction system. Importantly, inherited or acquired defects in a wide class of ion channels has been linked to dysfunction at various stages of the conduction system resulting in life-threatening cardiac arrhythmia. There is growing appreciation of the role that adapter and cytoskeletal proteins play in organizing ion channel macromolecular complexes critical for proper function of the cardiac conduction system. In particular, members of the ankyrin and spectrin families have emerged as important nodes for normal expression and regulation of ion channels in myocytes throughout the conduction system. Human variants impacting ankyrin/spectrin function give rise to a broad constellation of cardiac arrhythmias. Furthermore, chronic neurohumoral and biomechanical stress promotes ankyrin/spectrin loss of function that likely contributes to conduction disturbances in the setting of acquired cardiac disease. Collectively, this review seeks to bring attention to the significance of these cytoskeletal players and emphasize the potential therapeutic role they represent in a myriad of cardiac disease states.
Collapse
|
28
|
Miazek A, Zalas M, Skrzymowska J, Bogin BA, Grzymajło K, Goszczynski TM, Levine ZA, Morrow JS, Stankewich MC. Age-dependent ataxia and neurodegeneration caused by an αII spectrin mutation with impaired regulation of its calpain sensitivity. Sci Rep 2021; 11:7312. [PMID: 33790315 PMCID: PMC8012654 DOI: 10.1038/s41598-021-86470-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
The neuronal membrane-associated periodic spectrin skeleton (MPS) contributes to neuronal development, remodeling, and organization. Post-translational modifications impinge on spectrin, the major component of the MPS, but their role remains poorly understood. One modification targeting spectrin is cleavage by calpains, a family of calcium-activated proteases. Spectrin cleavage is regulated by activated calpain, but also by the calcium-dependent binding of calmodulin (CaM) to spectrin. The physiologic significance of this balance between calpain activation and substrate-level regulation of spectrin cleavage is unknown. We report a strain of C57BL/6J mice harboring a single αII spectrin point mutation (Sptan1 c.3293G > A:p.R1098Q) with reduced CaM affinity and intrinsically enhanced sensitivity to calpain proteolysis. Homozygotes are embryonic lethal. Newborn heterozygotes of either gender appear normal, but soon develop a progressive ataxia characterized biochemically by accelerated calpain-mediated spectrin cleavage and morphologically by disruption of axonal and dendritic integrity and global neurodegeneration. Molecular modeling predicts unconstrained exposure of the mutant spectrin's calpain-cleavage site. These results reveal the critical importance of substrate-level regulation of spectrin cleavage for the maintenance of neuronal integrity. Given that excessive activation of calpain proteases is a common feature of neurodegenerative disease and traumatic encephalopathy, we propose that damage to the spectrin MPS may contribute to the neuropathology of many disorders.
Collapse
Affiliation(s)
- Arkadiusz Miazek
- Department of Tumor Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
- Department of Biochemistry and Molecular Biology, Wroclaw University of Environmental and Life Sciences, Norwida 31, 50-375, Wrocław, Poland
| | - Michał Zalas
- Department of Tumor Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Joanna Skrzymowska
- Department of Tumor Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Bryan A Bogin
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Krzysztof Grzymajło
- Department of Biochemistry and Molecular Biology, Wroclaw University of Environmental and Life Sciences, Norwida 31, 50-375, Wrocław, Poland
| | - Tomasz M Goszczynski
- Department of Tumor Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Zachary A Levine
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, 310 Cedar Street, LH108, New Haven, CT, 06520, USA
| | - Jon S Morrow
- Department of Pathology, Yale University School of Medicine, 310 Cedar Street, LH108, New Haven, CT, 06520, USA.
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA.
| | - Michael C Stankewich
- Department of Pathology, Yale University School of Medicine, 310 Cedar Street, LH108, New Haven, CT, 06520, USA.
| |
Collapse
|
29
|
Morrow JS, Stankewich MC. The Spread of Spectrin in Ataxia and Neurodegenerative Disease. JOURNAL OF EXPERIMENTAL NEUROLOGY 2021; 2:131-139. [PMID: 34528024 PMCID: PMC8439443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Experimental and hereditary defects in the ubiquitous scaffolding proteins of the spectrin gene family cause an array of neuropathologies. Most recognized are ataxias caused by missense, deletions, or truncations in the SPTBN2 gene that encodes beta III spectrin. Such mutations disrupt the organization of post-synaptic receptors, their active transport through the secretory pathway, and the organization and dynamics of the actin-based neuronal skeleton. Similar mutations in SPTAN1 that encodes alpha II spectrin cause severe and usually lethal neurodevelopmental defects including one form of early infantile epileptic encephalopathy type 5 (West syndrome). Defects in these and other spectrins are implicated in degenerative and psychiatric conditions. In recent published work, we describe in mice a novel variant of alpha II spectrin that results in a progressive ataxia with widespread neurodegenerative change. The action of this variant is distinct, in that rather than disrupting a constitutive ligand-binding function of spectrin, the mutation alters its response to calcium and calmodulin-regulated signaling pathways including its response to calpain activation. As such, it represents a novel spectrinopathy that targets a key regulatory pathway where calcium and tyrosine kinase signals converge. Here we briefly discuss the various roles of spectrin in neuronal processes and calcium activated regulatory inputs that control its participation in neuronal growth, organization, and remodeling. We hypothesize that damage to the neuronal spectrin scaffold may be a common final pathway in many neurodegenerative disorders. Targeting the pathways that regulate spectrin function may thus offer novel avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Jon S. Morrow
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA,Molecular & Cellular Developmental Biology, Yale University, New Haven, CT 06520, USA,Correspondence should be addressed to Jon S. Morrow; , Michael Stankewich;
| | - Michael C. Stankewich
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA,Correspondence should be addressed to Jon S. Morrow; , Michael Stankewich;
| |
Collapse
|
30
|
Fujishima K, Kurisu J, Yamada M, Kengaku M. βIII spectrin controls the planarity of Purkinje cell dendrites by modulating perpendicular axon-dendrite interactions. Development 2020; 147:226102. [PMID: 33234719 DOI: 10.1242/dev.194530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/04/2020] [Indexed: 01/14/2023]
Abstract
The mechanism underlying the geometrical patterning of axon and dendrite wiring remains elusive, despite its crucial importance in the formation of functional neural circuits. The cerebellar Purkinje cell (PC) arborizes a typical planar dendrite, which forms an orthogonal network with granule cell (GC) axons. By using electrospun nanofiber substrates, we reproduce the perpendicular contacts between PC dendrites and GC axons in culture. In the model system, PC dendrites show a preference to grow perpendicularly to aligned GC axons, which presumably contribute to the planar dendrite arborization in vivo We show that βIII spectrin, a causal protein for spinocerebellar ataxia type 5, is required for the biased growth of dendrites. βIII spectrin deficiency causes actin mislocalization and excessive microtubule invasion in dendritic protrusions, resulting in abnormally oriented branch formation. Furthermore, disease-associated mutations affect the ability of βIII spectrin to control dendrite orientation. These data indicate that βIII spectrin organizes the mouse dendritic cytoskeleton and thereby regulates the oriented growth of dendrites with respect to the afferent axons.
Collapse
Affiliation(s)
- Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Junko Kurisu
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Midori Yamada
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Mineko Kengaku
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
31
|
Complementary mesoscale dynamics of spectrin and acto-myosin shape membrane territories during mechanoresponse. Nat Commun 2020; 11:5108. [PMID: 33037189 PMCID: PMC7547731 DOI: 10.1038/s41467-020-18825-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 09/11/2020] [Indexed: 11/08/2022] Open
Abstract
The spectrin-based membrane skeleton is a major component of the cell cortex. While expressed by all metazoans, its dynamic interactions with the other cortex components, including the plasma membrane or the acto-myosin cytoskeleton, are poorly understood. Here, we investigate how spectrin re-organizes spatially and dynamically under the membrane during changes in cell mechanics. We find spectrin and acto-myosin to be spatially distinct but cooperating during mechanical challenges, such as cell adhesion and contraction, or compression, stretch and osmolarity fluctuations, creating a cohesive cortex supporting the plasma membrane. Actin territories control protrusions and contractile structures while spectrin territories concentrate in retractile zones and low-actin density/inter-contractile regions, acting as a fence that organize membrane trafficking events. We unveil here the existence of a dynamic interplay between acto-myosin and spectrin necessary to support a mesoscale organization of the lipid bilayer into spatially-confined cortical territories during cell mechanoresponse.
Collapse
|
32
|
Abstract
Fodrin and its erythroid cell-specific isoform spectrin are actin-associated fibrous proteins that play crucial roles in the maintenance of structural integrity in mammalian cells, which is necessary for proper cell function. Normal cell morphology is altered in diseases such as various cancers and certain neuronal disorders. Fodrin and spectrin are two-chain (αβ) molecules that are encoded by paralogous genes and share many features but also demonstrate certain differences. Fodrin (in humans, typically a heterodimer of the products of the SPTAN1 and SPTBN1 genes) is expressed in nearly all cell types and is especially abundant in neuronal tissues, whereas spectrin (in humans, a heterodimer of the products of the SPTA1 and SPTB1 genes) is expressed almost exclusively in erythrocytes. To fulfill a role in such a variety of different cell types, it was anticipated that fodrin would need to be a more versatile scaffold than spectrin. Indeed, as summarized here, domains unique to fodrin and its regulation by Ca2+, calmodulin, and a variety of posttranslational modifications (PTMs) endow fodrin with additional specific functions. However, how fodrin structural variations and misregulated PTMs may contribute to the etiology of various cancers and neurodegenerative diseases needs to be further investigated.
Collapse
|
33
|
Beijer D, Deconinck T, De Bleecker JL, Dotti MT, Malandrini A, Urtizberea JA, Zulaica M, López de Munain A, Asselbergh B, De Jonghe P, Baets J. Nonsense mutations in alpha-II spectrin in three families with juvenile onset hereditary motor neuropathy. Brain 2020; 142:2605-2616. [PMID: 31332438 DOI: 10.1093/brain/awz216] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/25/2019] [Accepted: 05/28/2019] [Indexed: 01/09/2023] Open
Abstract
Distal hereditary motor neuropathies are a rare subgroup of inherited peripheral neuropathies hallmarked by a length-dependent axonal degeneration of lower motor neurons without significant involvement of sensory neurons. We identified patients with heterozygous nonsense mutations in the αII-spectrin gene, SPTAN1, in three separate dominant hereditary motor neuropathy families via next-generation sequencing. Variable penetrance was noted for these mutations in two of three families, and phenotype severity differs greatly between patients. The mutant mRNA containing nonsense mutations is broken down by nonsense-mediated decay and leads to reduced protein levels in patient cells. Previously, dominant-negative αII-spectrin gene mutations were described as causal in a spectrum of epilepsy phenotypes.
Collapse
Affiliation(s)
- Danique Beijer
- Neurogenetics Group, Center for Molecular Neurology, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium
| | - Tine Deconinck
- Neurogenetics Group, Center for Molecular Neurology, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium
| | | | - Maria Teresa Dotti
- Department of Medicine, Surgery and Neuroscience, University of Siena, Italy
| | | | | | - Miren Zulaica
- Neuroscience Area, Institute Biodonostia, Hospital Universitario Donostia, San Sebastian, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, Madrid, Spain
| | - Adolfo López de Munain
- Neuroscience Area, Institute Biodonostia, Hospital Universitario Donostia, San Sebastian, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, Madrid, Spain
| | - Bob Asselbergh
- VIB-UAntwerp Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | - Peter De Jonghe
- Neurogenetics Group, Center for Molecular Neurology, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium.,Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Belgium
| | - Jonathan Baets
- Neurogenetics Group, Center for Molecular Neurology, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium.,Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Belgium
| |
Collapse
|
34
|
Machnicka B, Ponceau A, Picot J, Colin Y, Lecomte MC. Deficiency of αII-spectrin affects endothelial cell-matrix contact and migration leading to impairment of angiogenesis in vitro. Cell Mol Biol Lett 2020; 25:3. [PMID: 32042281 PMCID: PMC6998227 DOI: 10.1186/s11658-020-0200-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 01/20/2020] [Indexed: 12/15/2022] Open
Abstract
Background Precise coordination of cytoskeletal components and dynamic control of cell adhesion and migration are required for crucial cell processes such as differentiation and morphogenesis. We investigated the potential involvement of αII-spectrin, a ubiquitous scaffolding element of the membrane skeleton, in the adhesion and angiogenesis mechanism. Methods The cell models were primary human umbilical vein endothelial cells (HUVECs) and a human dermal microvascular endothelial cell line (HMEC-1). After siRNA- and shRNA-mediated knockdown of αII-spectrin, we assessed its expression and that of its partners and adhesion proteins using western blotting. The phenotypes of the control and spectrin-depleted cells were examined using immunofluorescence and video microscopy. Capillary tube formation was assessed using the thick gel Matrigel matrix-based method and a microscope equipped with a thermostatic chamber and a Nikon Biostation System camera. Results Knockdown of αII-spectrin leads to: modified cell shape; actin cytoskeleton organization with the presence of peripheral actin patches; and decreased formation of stress fibers. Spectrin deficiency affects cell adhesion on laminin and fibronectin and cell motility. This included modification of the localization of adhesion molecules, such as αVβ3- and α5-integrins, and organization of adhesion structures, such as focal points. Deficiency of αII-spectrin can also affect the complex mechanism of in vitro capillary tube formation, as demonstrated in a model of angiogenesis. Live imaging revealed that impairment of capillary tube assembly was mainly associated with a significant decrease in cell projection length and stability. αII-spectrin depletion is also associated with significantly decreased expression of three proteins involved in capillary tube formation and assembly: VE-cadherin, MCAM and β3-integrin. Conclusion Our data confirm the role of αII-spectrin in the control of cell adhesion and spreading. Moreover, our findings further support the participation of αII-spectrin in capillary tube formation in vitro through control of adhesion molecules, such as integrins. This indicates a new function of αII-spectrin in angiogenesis.
Collapse
Affiliation(s)
- Beata Machnicka
- 1University of Zielona Góra, Institute of Biological Sciences, Zielona Góra, Poland
| | - Aurélie Ponceau
- 2Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.,3Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Julien Picot
- 2Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.,3Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Yves Colin
- 2Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.,3Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Marie-Christine Lecomte
- 2Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.,3Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| |
Collapse
|
35
|
Li JH, Shi ZJ, Li Y, Pan B, Yuan SY, Shi LL, Hao Y, Cao FJ, Feng SQ. Bioinformatic identification of key candidate genes and pathways in axon regeneration after spinal cord injury in zebrafish. Neural Regen Res 2020; 15:103-111. [PMID: 31535658 PMCID: PMC6862403 DOI: 10.4103/1673-5374.264460] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Zebrafish and human genomes are highly homologous; however, despite this genomic similarity, adult zebrafish can achieve neuronal proliferation, regeneration and functional restoration within 6–8 weeks after spinal cord injury, whereas humans cannot. To analyze differentially expressed zebrafish genes between axon-regenerated neurons and axon-non-regenerated neurons after spinal cord injury, and to explore the key genes and pathways of axonal regeneration after spinal cord injury, microarray GSE56842 was analyzed using the online tool, GEO2R, in the Gene Expression Omnibus database. Gene ontology and protein-protein interaction networks were used to analyze the identified differentially expressed genes. Finally, we screened for genes and pathways that may play a role in spinal cord injury repair in zebrafish and mammals. A total of 636 differentially expressed genes were obtained, including 255 up-regulated and 381 down-regulated differentially expressed genes in axon-regenerated neurons. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment results were also obtained. A protein-protein interaction network contained 480 node genes and 1976 node connections. We also obtained the 10 hub genes with the highest correlation and the two modules with the highest score. The results showed that spectrin may promote axonal regeneration after spinal cord injury in zebrafish. Transforming growth factor beta signaling may inhibit repair after spinal cord injury in zebrafish. Focal adhesion or tight junctions may play an important role in the migration and proliferation of some cells, such as Schwann cells or neural progenitor cells, after spinal cord injury in zebrafish. Bioinformatic analysis identified key candidate genes and pathways in axonal regeneration after spinal cord injury in zebrafish, providing targets for treatment of spinal cord injury in mammals.
Collapse
Affiliation(s)
- Jia-He Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhong-Ju Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bin Pan
- Department of Orthopedics, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Shi-Yang Yuan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Lin-Lin Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Hao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fu-Jiang Cao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shi-Qing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
36
|
Lambert MW. The functional importance of lamins, actin, myosin, spectrin and the LINC complex in DNA repair. Exp Biol Med (Maywood) 2019; 244:1382-1406. [PMID: 31581813 PMCID: PMC6880146 DOI: 10.1177/1535370219876651] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Three major proteins in the nucleoskeleton, lamins, actin, and spectrin, play essential roles in maintenance of nuclear architecture and the integrity of the nuclear envelope, in mechanotransduction and mechanical coupling between the nucleoskeleton and cytoskeleton, and in nuclear functions such as regulation of gene expression, transcription and DNA replication. Less well known, but critically important, are the role these proteins play in DNA repair. The A-type and B-type lamins, nuclear actin and myosin, spectrin and the LINC (linker of nucleoskeleton and cytoskeleton) complex each function in repair of DNA damage utilizing various repair pathways. The lamins play a role in repair of DNA double-strand breaks (DSBs) by nonhomologous end joining (NHEJ) or homologous recombination (HR). Actin is involved in repair of DNA DSBs and interacts with myosin in facilitating relocalization of these DSBs in heterochromatin for HR repair. Nonerythroid alpha spectrin (αSpII) plays a critical role in repair of DNA interstrand cross-links (ICLs) where it acts as a scaffold in recruitment of repair proteins to sites of damage and is important in the initial damage recognition and incision steps of the repair process. The LINC complex contributes to the repair of DNA DSBs and ICLs. This review will address the important functions of these proteins in the DNA repair process, their mechanism of action, and the profound impact a defect or deficiency in these proteins has on cellular function. The critical roles of these proteins in DNA repair will be further emphasized by discussing the human disorders and the pathophysiological changes that result from or are related to deficiencies in these proteins. The demonstrated function for each of these proteins in the DNA repair process clearly indicates that there is another level of complexity that must be considered when mechanistically examining factors crucial for DNA repair.
Collapse
Affiliation(s)
- Muriel W Lambert
- Department of Pathology, Immunology and Laboratory
Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
37
|
Cheng C, Hu Z, Cao L, Peng C, He Y. The scavenger receptor SCARA1 (CD204) recognizes dead cells through spectrin. J Biol Chem 2019; 294:18881-18897. [PMID: 31653705 DOI: 10.1074/jbc.ra119.010110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/22/2019] [Indexed: 01/05/2023] Open
Abstract
Scavenger receptor class A member 1 (SCARA1 or CD204) is an immune receptor highly expressed on macrophages. It forms homotrimers on the cell surface and plays important roles in regulating immune responses via its involvement in multiple pathways. However, both the structure and the functional roles of SCARA1 are not fully understood. Here, we determined the crystal structure of the C-terminal SRCR domain of SCARA1 at 1.8 Å resolution, revealing its Ca2+-binding site. Results from cell-based assays revealed that SCARA1 can recognize dead cells, rather than live cells, specifically through its SRCR domain and in a Ca2+-dependent manner. Furthermore, by combining MS and biochemical assays, we found that cellular spectrin is the binding target of SCARA1 on dead cells and that the SRCR domain of SCARA1 recognizes the SPEC repeats of spectrin in the presence of Ca2+ We also found that macrophages can internalize dead cells or debris from both erythrocytes and other cells through the interaction between SCARA1 and spectrin, suggesting that SCARA1 could function as a scavenging receptor that recognizes dead cells. These results suggest that spectrin, which is one of the major components of the cytoskeleton, acts as a cellular marker that enables the recognition of dead cells by the immune system.
Collapse
Affiliation(s)
- Chen Cheng
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhenzheng Hu
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Longxing Cao
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chao Peng
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yongning He
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
38
|
Nellikka RK, Sreeja JS, Dharmapal D, John R, Monteiro A, Macedo JC, Conde C, Logarinho E, Sunkel CE, Sengupta S. α-Fodrin is required for the organization of functional microtubules during mitosis. Cell Cycle 2019; 18:2713-2726. [PMID: 31455186 DOI: 10.1080/15384101.2019.1656476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The cytoskeleton protein α-fodrin plays a major role in maintaining structural stability of membranes. It was also identified as part of the brain γ-tubulin ring complex, the major microtubule nucleator. Here, we investigated the requirement of α-fodrin for microtubule spindle assembly during mitotic progression. We found that α-fodrin depletion results in abnormal mitosis with uncongressed chromosomes, leading to prolonged activation of the spindle assembly checkpoint and a severe mitotic delay. Further, α-fodrin repression led to the formation of shortened spindles with unstable kinetochore-microtubule attachments. We also found that the mitotic kinesin CENP-E had reduced levels at kinetochores to likely account for the chromosome misalignment defects in α-fodrin-depleted cells. Importantly, we showed these cells to exhibit reduced levels of detyrosinated α-tubulin, which primarily drives CENP-E localization. Since proper microtubule dynamics and chromosome alignment are required for completion of normal mitosis, this study reveals an unforeseen role of α-fodrin in regulating mitotic progression. Future studies on these lines of observations should reveal important mechanistic insight for fodrin's involvement in cancer.
Collapse
Affiliation(s)
- Rohith Kumar Nellikka
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| | - Jamuna S Sreeja
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| | - Dhrishya Dharmapal
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| | - Rince John
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| | | | | | - Carlos Conde
- i3S-IBMC, Universidade do Porto , Porto , Portugal
| | | | - Claudio E Sunkel
- i3S-IBMC, Universidade do Porto , Porto , Portugal.,ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto , Porto , Portugal
| | - Suparna Sengupta
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| |
Collapse
|
39
|
Haaland ØA, Romanowska J, Gjerdevik M, Lie RT, Gjessing HK, Jugessur A. A genome-wide scan of cleft lip triads identifies parent-of-origin interaction effects between ANK3 and maternal smoking, and between ARHGEF10 and alcohol consumption. F1000Res 2019; 8:960. [PMID: 31372216 PMCID: PMC6662680 DOI: 10.12688/f1000research.19571.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2019] [Indexed: 01/08/2023] Open
Abstract
Background: Although both genetic and environmental factors have been reported to influence the risk of isolated cleft lip with or without cleft palate (CL/P), the exact mechanisms behind CL/P are still largely unaccounted for. We recently developed new methods to identify parent-of-origin (PoO) interactions with environmental exposures (PoOxE) and now apply them to data from a genome-wide association study (GWAS) of families with children born with isolated CL/P. Methods: Genotypes from 1594 complete triads and 314 dyads (1908 nuclear families in total) with CL/P were available for the current analyses. Of these families, 1024 were Asian, 825 were European and 59 had other ancestries. After quality control, 341,191 SNPs remained from the original 569,244. The exposures were maternal cigarette smoking, use of alcohol, and use of vitamin supplements in the periconceptional period. Our new methodology detects if PoO effects are different across environmental strata and is implemented in the R-package Haplin. Results: Among Europeans, there was evidence of a PoOxSmoke effect for ANK3 with three SNPs (rs3793861, q=0.20, p=2.6e-6; rs7087489, q=0.20, p=3.1e-6; rs4310561, q=0.67, p=4.0e-5) and a PoOxAlcohol effect for ARHGEF10 with two SNPs (rs2294035, q=0.32, p=2.9e-6; rs4876274, q=0.76, p=1.3e-5). Conclusion: Our results indicate that the detected PoOxE effects have a plausible biological basis, and thus warrant replication in other independent cleft samples. Our demonstration of the feasibility of identifying complex interactions between relevant environmental exposures and PoO effects offers new avenues for future research aimed at unravelling the complex etiology of cleft lip defects.
Collapse
Affiliation(s)
- Øystein Ariansen Haaland
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
| | - Julia Romanowska
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Computational Biology Unit, University of Bergen, Bergen, N-5020, Norway
| | - Miriam Gjerdevik
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Skøyen, Oslo, Skøyen, N-0213, Norway
| | - Rolv Terje Lie
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, Skøyen, Oslo, N-0213, Norway
| | - Håkon Kristian Gjessing
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, Skøyen, Oslo, N-0213, Norway
| | - Astanand Jugessur
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Skøyen, Oslo, Skøyen, N-0213, Norway
- Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, Skøyen, Oslo, N-0213, Norway
| |
Collapse
|
40
|
Haaland ØA, Romanowska J, Gjerdevik M, Lie RT, Gjessing HK, Jugessur A. A genome-wide scan of cleft lip triads identifies parent-of-origin interaction effects between ANK3 and maternal smoking, and between ARHGEF10 and alcohol consumption. F1000Res 2019; 8:960. [PMID: 31372216 PMCID: PMC6662680 DOI: 10.12688/f1000research.19571.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Although both genetic and environmental factors have been reported to influence the risk of isolated cleft lip with or without cleft palate (CL/P), the exact mechanisms behind CL/P are still largely unaccounted for. We recently developed new methods to identify parent-of-origin (PoO) interactions with environmental exposures (PoOxE) and applied them to families with children born with isolated cleft palate only. Here, we used the same genome-wide association study (GWAS) dataset and methodology to screen for PoOxE effects in the larger sample of CL/P triads. Methods: Genotypes from 1594 complete triads and 314 dyads (1908 nuclear families in total) with CL/P were available for the current analyses. Of these families, 1024 were Asian, 825 were European and 59 had other ancestries. After quality control, 341,191 SNPs remained from the original 569,244. The exposures were maternal cigarette smoking, use of alcohol, and use of vitamin supplements in the periconceptional period. The methodology applied in the analyses is implemented in the R-package Haplin. Results: Among Europeans, there was evidence of a PoOxSmoke effect for ANK3 with three SNPs (rs3793861, q=0.20, p=2.6e-6; rs7087489, q=0.20, p=3.1e-6; rs4310561, q=0.67, p=4.0e-5) and a PoOxAlcohol effect for ARHGEF10 with two SNPs (rs2294035, q=0.32, p=2.9e-6; rs4876274, q=0.76, p=1.3e-5). Conclusion: Our results indicate that the detected PoOxE effects have a plausible biological basis, and thus warrant replication in other independent cleft samples. Our demonstration of the feasibility of identifying complex interactions between relevant environmental exposures and PoO effects offers new avenues for future research aimed at unravelling the complex etiology of cleft lip defects.
Collapse
Affiliation(s)
- Øystein Ariansen Haaland
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
| | - Julia Romanowska
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Computational Biology Unit, University of Bergen, Bergen, N-5020, Norway
| | - Miriam Gjerdevik
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Skøyen, Oslo, Skøyen, N-0213, Norway
| | - Rolv Terje Lie
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, Skøyen, Oslo, N-0213, Norway
| | - Håkon Kristian Gjessing
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, Skøyen, Oslo, N-0213, Norway
| | - Astanand Jugessur
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, N-5020, Norway
- Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Skøyen, Oslo, Skøyen, N-0213, Norway
- Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, Skøyen, Oslo, N-0213, Norway
| |
Collapse
|
41
|
Machnicka B, Grochowalska R, Bogusławska DM, Sikorski AF. The role of spectrin in cell adhesion and cell-cell contact. Exp Biol Med (Maywood) 2019; 244:1303-1312. [PMID: 31226892 DOI: 10.1177/1535370219859003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Spectrins are proteins that are responsible for many aspects of cell function and adaptation to changing environments. Primarily the spectrin-based membrane skeleton maintains cell membrane integrity and its mechanical properties, together with the cytoskeletal network a support cell shape. The occurrence of a variety of spectrin isoforms in diverse cellular environments indicates that it is a multifunctional protein involved in numerous physiological pathways. Participation of spectrin in cell–cell and cell–extracellular matrix adhesion and formation of dynamic plasma membrane protrusions and associated signaling events is a subject of interest for researchers in the fields of cell biology and molecular medicine. In this mini-review, we focus on data concerning the role of spectrins in cell surface activities such as adhesion, cell–cell contact, and invadosome formation. We discuss data on different adhesion proteins that directly or indirectly interact with spectrin repeats. New findings support the involvement of spectrin in cell adhesion and spreading, formation of lamellipodia, and also the participation in morphogenetic processes, such as eye development, oogenesis, and angiogenesis. Here, we review the role of spectrin in cell adhesion and cell–cell contact.Impact statementThis article reviews properties of spectrins as a group of proteins involved in cell surface activities such as, adhesion and cell–cell contact, and their contribution to morphogenesis. We show a new area of research and discuss the involvement of spectrin in regulation of cell–cell contact leading to immunological synapse formation and in shaping synapse architecture during myoblast fusion. Data indicate involvement of spectrins in adhesion and cell–cell or cell–extracellular matrix interactions and therefore in signaling pathways. There is evidence of spectrin’s contribution to the processes of morphogenesis which are connected to its interactions with adhesion molecules, membrane proteins (and perhaps lipids), and actin. Our aim was to highlight the essential role of spectrin in cell–cell contact and cell adhesion.
Collapse
Affiliation(s)
- Beata Machnicka
- Department of Biochemistry and Bioinformatics, Faculty of Biological Sciences, University of Zielona Góra, Zielona Góra 65-516, Poland
| | - Renata Grochowalska
- Department of Biochemistry and Bioinformatics, Faculty of Biological Sciences, University of Zielona Góra, Zielona Góra 65-516, Poland
| | - Dżamila M Bogusławska
- Department of Biochemistry and Bioinformatics, Faculty of Biological Sciences, University of Zielona Góra, Zielona Góra 65-516, Poland
| | - Aleksander F Sikorski
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, Wrocław 50-383, Poland
| |
Collapse
|
42
|
Lubbers ER, Murphy NP, Musa H, Huang CYM, Gupta R, Price MV, Han M, Daoud G, Gratz D, El Refaey M, Xu X, Hoeflinger NK, Friel EL, Lancione P, Wallace MJ, Cavus O, Simmons SL, Williams JL, Skaf M, Koenig SN, Janssen PML, Rasband MN, Hund TJ, Mohler PJ. Defining new mechanistic roles for αII spectrin in cardiac function. J Biol Chem 2019; 294:9576-9591. [PMID: 31064843 PMCID: PMC6579463 DOI: 10.1074/jbc.ra119.007714] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/22/2019] [Indexed: 01/04/2023] Open
Abstract
Spectrins are cytoskeletal proteins essential for membrane biogenesis and regulation and serve critical roles in protein targeting and cellular signaling. αII spectrin (SPTAN1) is one of two α spectrin genes and αII spectrin dysfunction is linked to alterations in axon initial segment formation, cortical lamination, and neuronal excitability. Furthermore, human αII spectrin loss-of-function variants cause neurological disease. As global αII spectrin knockout mice are embryonic lethal, the in vivo roles of αII spectrin in adult heart are unknown and untested. Here, based on pronounced alterations in αII spectrin regulation in human heart failure we tested the in vivo roles of αII spectrin in the vertebrate heart. We created a mouse model of cardiomyocyte-selective αII spectrin-deficiency (cKO) and used this model to define the roles of αII spectrin in cardiac function. αII spectrin cKO mice displayed significant structural, cellular, and electrical phenotypes that resulted in accelerated structural remodeling, fibrosis, arrhythmia, and mortality in response to stress. At the molecular level, we demonstrate that αII spectrin plays a nodal role for global cardiac spectrin regulation, as αII spectrin cKO hearts exhibited remodeling of αI spectrin and altered β-spectrin expression and localization. At the cellular level, αII spectrin deficiency resulted in altered expression, targeting, and regulation of cardiac ion channels NaV1.5 and KV4.3. In summary, our findings define critical and unexpected roles for the multifunctional αII spectrin protein in the heart. Furthermore, our work provides a new in vivo animal model to study the roles of αII spectrin in the cardiomyocyte.
Collapse
Affiliation(s)
- Ellen R Lubbers
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
- Medical Scientist Training Program
- the Departments of Physiology and Cell Biology and
| | - Nathaniel P Murphy
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
- Medical Scientist Training Program
- the Departments of Physiology and Cell Biology and
| | - Hassan Musa
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Claire Yu-Mei Huang
- the Department of Neuroscience and Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas 77030, and
| | - Rohan Gupta
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Morgan V Price
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Mei Han
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Georges Daoud
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Daniel Gratz
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
- the Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 432310
| | - Mona El Refaey
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Xianyao Xu
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Nicole K Hoeflinger
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Emma L Friel
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Peter Lancione
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Michael J Wallace
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Omer Cavus
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Samantha L Simmons
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Jordan L Williams
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Michel Skaf
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Sara N Koenig
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
| | - Paul M L Janssen
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
- the Departments of Physiology and Cell Biology and
- Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio 432310
| | - Matthew N Rasband
- the Department of Neuroscience and Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas 77030, and
| | - Thomas J Hund
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia
- the Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 432310
- Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio 432310
| | - Peter J Mohler
- From the Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia,
- the Departments of Physiology and Cell Biology and
- Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio 432310
| |
Collapse
|
43
|
Grimes KM, Prasad V, McNamara JW. Supporting the heart: Functions of the cardiomyocyte's non-sarcomeric cytoskeleton. J Mol Cell Cardiol 2019; 131:187-196. [PMID: 30978342 DOI: 10.1016/j.yjmcc.2019.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
The non-contractile cytoskeleton in cardiomyocytes is comprised of cytoplasmic actin, microtubules, and intermediate filaments. In addition to providing mechanical support to these cells, these structures are important effectors of tension-sensing and signal transduction and also provide networks for the transport of proteins and organelles. The majority of our knowledge on the function and structure of these cytoskeletal networks comes from research on proliferative cell types. However, in recent years, researchers have begun to show that there are important cardiomyocyte-specific functions of the cytoskeleton. Here we will discuss the current state of cytoskeletal biology in cardiomyocytes, as well as research from other cell types, that together suggest there is a wealth of knowledge on cardiac health and disease waiting to be uncovered through exploration of the complex signaling networks of cardiomyocyte non-sarcomeric cytoskeletal proteins.
Collapse
Affiliation(s)
- Kelly M Grimes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Vikram Prasad
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - James W McNamara
- Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
44
|
Hu S, Jue D, Albanese J, Wang Y, Liu Q. Utilization of spectrins βI and βIII in diagnosis of hepatocellular carcinoma. Ann Diagn Pathol 2019; 39:86-91. [PMID: 30798076 DOI: 10.1016/j.anndiagpath.2019.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/08/2019] [Indexed: 12/24/2022]
Abstract
Spectrins are a group of cytoskeletal proteins which participate in many important cellular functions. It has been suggested that loss of spectrin isoforms may be associated with tumorigenesis of lymphoma, leukemia, gastric cancer and hepatocellular carcinoma (HCC). We recently reported that βI spectrin expression was present in normal hepatocytes but lost in HCC cells, which suggested that spectrins may be helpful markers in diagnosis of HCC. In this study, using immunohistochemical staining, we further investigated the expression pattern of four spectrin isoforms (αII, βI-III) on different benign and malignant liver tumors including focal nodular hyperplasia (FNH), hepatic adenoma (HA), HCC, and cholangiocarcinoma (CC). The results revealed that βI spectrin was moderately to strongly positive in FNH and HA tissues, but was only weakly positive or lost in HCC cases and was weakly positive in all CC cases. In addition, the βIII spectrin, majority of which was moderately positive in both FNH and HA tissues, was mostly lost in poorly differentiated HCC but remained at least moderately positive in most CC cases. These results suggest that spectrins βI and βIII may be used to differentiate well differentiated HCC from FNH or HA, and poorly differentiated HCC from CC, respectively.
Collapse
Affiliation(s)
- Shaomin Hu
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, United States of America
| | - Deborah Jue
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, United States of America
| | - Joseph Albanese
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, United States of America
| | - Yanhua Wang
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, United States of America.
| | - Qiang Liu
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, United States of America.
| |
Collapse
|
45
|
Lambert MW. Spectrin and its interacting partners in nuclear structure and function. Exp Biol Med (Maywood) 2019; 243:507-524. [PMID: 29557213 DOI: 10.1177/1535370218763563] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nonerythroid αII-spectrin is a structural protein whose roles in the nucleus have just begun to be explored. αII-spectrin is an important component of the nucleoskelelton and has both structural and non-structural functions. Its best known role is in repair of DNA ICLs both in genomic and telomeric DNA. αII-spectrin aids in the recruitment of repair proteins to sites of damage and a proposed mechanism of action is presented. It interacts with a number of different groups of proteins in the nucleus, indicating it has roles in additional cellular functions. αII-spectrin, in its structural role, associates/co-purifies with proteins important in maintaining the architecture and mechanical properties of the nucleus such as lamin, emerin, actin, protein 4.1, nuclear myosin, and SUN proteins. It is important for the resilience and elasticity of the nucleus. Thus, αII-spectrin's role in cellular functions is complex due to its structural as well as non-structural roles and understanding the consequences of a loss or deficiency of αII-spectrin in the nucleus is a significant challenge. In the bone marrow failure disorder, Fanconi anemia, there is a deficiency in αII-spectrin and, among other characteristics, there is defective DNA repair, chromosome instability, and congenital abnormalities. One may speculate that a deficiency in αII-spectrin plays an important role not only in the DNA repair defect but also in the congenital anomalies observed in Fanconi anemia , particularly since αII-spectrin has been shown to be important in embryonic development in a mouse model. The dual roles of αII-spectrin in the nucleus in both structural and non-structural functions make this an extremely important protein which needs to be investigated further. Such investigations should help unravel the complexities of αII-spectrin's interactions with other nuclear proteins and enhance our understanding of the pathogenesis of disorders, such as Fanconi anemia , in which there is a deficiency in αII-spectrin. Impact statement The nucleoskeleton is critical for maintaining the architecture and functional integrity of the nucleus. Nonerythroid α-spectrin (αIISp) is an essential nucleoskeletal protein; however, its interactions with other structural and non-structural nuclear proteins and its functional importance in the nucleus have only begun to be explored. This review addresses these issues. It describes αIISp's association with DNA repair proteins and at least one proposed mechanism of action for its role in DNA repair. Specific interactions of αIISp with other nucleoskeletal proteins as well as its important role in the biomechanical properties of the nucleus are reviewed. The consequences of loss of αIISp, in disorders such as Fanconi anemia, are examined, providing insights into the profound impact of this loss on critical processes known to be abnormal in FA, such as development, carcinogenesis, cancer progression and cellular functions dependent upon αIISp's interactions with other nucleoskeletal proteins.
Collapse
Affiliation(s)
- Muriel W Lambert
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
46
|
Gartner V, Markello TC, Macnamara E, De Biase A, Thurm A, Joseph L, Beggs A, Schmahmann JD, Berry GT, Anselm I, Boslet E, Tifft CJ, Gahl WA, Lee PR. Novel variants in SPTAN1 without epilepsy: An expansion of the phenotype. Am J Med Genet A 2018; 176:2768-2776. [PMID: 30548380 PMCID: PMC11157598 DOI: 10.1002/ajmg.a.40628] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/19/2018] [Accepted: 08/13/2018] [Indexed: 11/11/2022]
Abstract
We describe two unrelated children with de novo variants in the non-erythrocytic alpha-II-spectrin (SPTAN1) gene who have hypoplastic brain structures, intellectual disability, and both fine and gross motor impairments. Using agnostic exome sequencing, we identified a nonsense variant creating a premature stop codon in exon 21 of SPTAN1, and in a second patient we identified an intronic substitution in SPTAN1 prior to exon 50 creating a new donor acceptor site. Neither of these variants has been described previously. Although some of these patients' features are consistent with the known SPTAN1 encephalopathy phenotype, these two children do not have epilepsy, in contrast to reports about nearly every other patient with heterozygous SPTAN1 variants and in all patients with a variant near the C-terminal coding region. Moreover, both children have abnormal thyroid function, which has not been previously reported in association with SPTAN1 variant. We present a detailed discussion of the clinical manifestations of these two unique SPTAN1 variants and provide evidence that both variants result in reduced mRNA expression despite different locations within the gene and clinical phenotypes. These findings expand the motor, cognitive, and behavioral spectrum of the SPTAN1-associated phenotype and invite speculation about underlying pathophysiologies.
Collapse
Affiliation(s)
- Valerie Gartner
- Office of the Clinical Director, NHGRI, and NIH Undiagnosed Diseases Program, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - Thomas C. Markello
- Office of the Clinical Director, NHGRI, and NIH Undiagnosed Diseases Program, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - Ellen Macnamara
- Office of the Clinical Director, NHGRI, and NIH Undiagnosed Diseases Program, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | | | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Lisa Joseph
- Neurodevelopmental and Behavioral Phenotyping Service, Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Alan Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeremy D. Schmahmann
- Ataxia Unit, Cognitive Behavioral Neurology Unit, Laboratory for Neuroanatomy and Cerebellar Neurobiology, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Gerard T. Berry
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Irina Anselm
- Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Emma Boslet
- Office of the Clinical Director, NHGRI, and NIH Undiagnosed Diseases Program, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - Cynthia J. Tifft
- Office of the Clinical Director, NHGRI, and NIH Undiagnosed Diseases Program, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - William A. Gahl
- Office of the Clinical Director, NHGRI, and NIH Undiagnosed Diseases Program, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - Paul R. Lee
- Office of the Clinical Director, NHGRI, and NIH Undiagnosed Diseases Program, Office of the Director, National Institutes of Health, Bethesda, Maryland
- Division of Neurology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
47
|
Wirshing ACE, Cram EJ. Spectrin regulates cell contractility through production and maintenance of actin bundles in the Caenorhabditis elegans spermatheca. Mol Biol Cell 2018; 29:2433-2449. [PMID: 30091661 PMCID: PMC6233056 DOI: 10.1091/mbc.e18-06-0347] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Disruption to the contractility of cells, including smooth muscle cells of the cardiovascular system and myoepithelial cells of the glandular epithelium, contributes to the pathophysiology of contractile tissue diseases, including asthma, hypertension, and primary Sjögren's syndrome. Cell contractility is determined by myosin activity and actomyosin network organization and is mediated by hundreds of protein-protein interactions, many directly involving actin. Here we use a candidate RNA interference screen of more than 100 Caenorhabditis elegans genes with predicted actin-binding and regulatory domains to identify genes that contribute to the contractility of the somatic gonad. We identify the spectrin cytoskeleton composed of SPC-1/α-spectrin, UNC-70/β-spectrin, and SMA-1/β heavy-spectrin as required for contractility and actin organization in the myoepithelial cells of the C. elegans spermatheca. We use imaging of fixed and live animals as well as tissue- and developmental-stage-specific disruption of the spectrin cytoskeleton to show that spectrin regulates the production of prominent central actin bundles and is required for maintenance of central actin bundles throughout successive rounds of stretch and contraction. We conclude that the spectrin cytoskeleton contributes to spermathecal contractility by promoting maintenance of the robust actomyosin bundles that drive contraction.
Collapse
Affiliation(s)
| | - Erin J Cram
- Department of Biology, Northeastern University, Boston, MA 02115
| |
Collapse
|
48
|
Huang CYM, Rasband MN. Axon initial segments: structure, function, and disease. Ann N Y Acad Sci 2018; 1420:46-61. [PMID: 29749636 DOI: 10.1111/nyas.13718] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/13/2018] [Accepted: 03/17/2018] [Indexed: 11/28/2022]
Abstract
The axon initial segment (AIS) is located at the proximal axon and is the site of action potential initiation. This reflects the high density of ion channels found at the AIS. Adaptive changes to the location and length of the AIS can fine-tune the excitability of neurons and modulate plasticity in response to activity. The AIS plays an important role in maintaining neuronal polarity by regulating the trafficking and distribution of proteins that function in somatodendritic or axonal compartments of the neuron. In this review, we provide an overview of the AIS cytoarchitecture, mechanism of assembly, and recent studies revealing mechanisms of differential transport at the AIS that maintain axon and dendrite identities. We further discuss how genetic mutations in AIS components (i.e., ankyrins, ion channels, and spectrins) and injuries may cause neurological disorders.
Collapse
Affiliation(s)
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
49
|
αII-spectrin and βII-spectrin do not affect TGFβ1-induced myofibroblast differentiation. Cell Tissue Res 2018; 374:165-175. [PMID: 29725768 PMCID: PMC6132645 DOI: 10.1007/s00441-018-2842-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 04/10/2018] [Indexed: 12/13/2022]
Abstract
Mechanosensing of fibroblasts plays a key role in the development of fibrosis. So far, no effective treatments are available to treat this devastating disorder. Spectrins regulate cell morphology and are potential mechanosensors in a variety of non-erythroid cells, but little is known about the role of spectrins in fibroblasts. We investigate whether αII- and βII-spectrin are required for the phenotypic properties of adult human dermal (myo)fibroblasts. Knockdown of αII- or βII-spectrin in fibroblasts did not affect cell adhesion, cell size and YAP nuclear/cytosolic localization. We further investigated whether αII- and βII-spectrin play a role in the phenotypical switch from fibroblasts to myofibroblasts under the influence of the pro-fibrotic cytokine TGFβ1. Knockdown of spectrins did not affect myofibroblast formation, nor did we observe changes in the organization of αSMA stress fibers. Focal adhesion assembly was unaffected by spectrin deficiency, as was collagen type I mRNA expression and protein deposition. Wound closure was unaffected as well, showing that important functional properties of myofibroblasts are unchanged without αII- or βII-spectrin. In fact, fibroblasts stimulated with TGFβ1 demonstrated significantly lower endogenous mRNA levels of αII- and βII-spectrin. Taken together, despite the diverse roles of spectrins in a variety of other cells, αII- and βII-spectrin do not regulate cell adhesion, cell size and YAP localization in human dermal fibroblasts and are not required for the dermal myofibroblast phenotypical switch.
Collapse
|
50
|
Wang Y, Ji T, Nelson AD, Glanowska K, Murphy GG, Jenkins PM, Parent JM. Critical roles of αII spectrin in brain development and epileptic encephalopathy. J Clin Invest 2018; 128:760-773. [PMID: 29337302 DOI: 10.1172/jci95743] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/28/2017] [Indexed: 12/26/2022] Open
Abstract
The nonerythrocytic α-spectrin-1 (SPTAN1) gene encodes the cytoskeletal protein αII spectrin. Mutations in SPTAN1 cause early infantile epileptic encephalopathy type 5 (EIEE5); however, the role of αII spectrin in neurodevelopment and EIEE5 pathogenesis is unknown. Prior work suggests that αII spectrin is absent in the axon initial segment (AIS) and contributes to a diffusion barrier in the distal axon. Here, we have shown that αII spectrin is expressed ubiquitously in rodent and human somatodendritic and axonal domains. CRISPR-mediated deletion of Sptan1 in embryonic rat forebrain by in utero electroporation caused altered dendritic and axonal development, loss of the AIS, and decreased inhibitory innervation. Overexpression of human EIEE5 mutant SPTAN1 in embryonic rat forebrain and mouse hippocampal neurons led to similar developmental defects that were also observed in EIEE5 patient-derived neurons. Additionally, patient-derived neurons displayed aggregation of spectrin complexes. Taken together, these findings implicate αII spectrin in critical aspects of dendritic and axonal development and synaptogenesis, and support a dominant-negative mechanism of SPTAN1 mutations in EIEE5.
Collapse
Affiliation(s)
| | | | | | | | - Geoffrey G Murphy
- Molecular and Behavioral Neuroscience Institute.,Department of Molecular and Integrative Physiology, and
| | - Paul M Jenkins
- Department of Pharmacology.,Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jack M Parent
- Department of Neurology.,Ann Arbor VA Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|