1
|
Lima JT, Ferreira JG. Mechanobiology of the nucleus during the G2-M transition. Nucleus 2024; 15:2330947. [PMID: 38533923 DOI: 10.1080/19491034.2024.2330947] [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: 11/30/2023] [Accepted: 03/09/2024] [Indexed: 03/28/2024] Open
Abstract
Cellular behavior is continuously influenced by mechanical forces. These forces span the cytoskeleton and reach the nucleus, where they trigger mechanotransduction pathways that regulate downstream biochemical events. Therefore, the nucleus has emerged as a regulator of cellular response to mechanical stimuli. Cell cycle progression is regulated by cyclin-CDK complexes. Recent studies demonstrated these biochemical pathways are influenced by mechanical signals, highlighting the interdependence of cellular mechanics and cell cycle regulation. In particular, the transition from G2 to mitosis (G2-M) shows significant changes in nuclear structure and organization, ranging from nuclear pore complex (NPC) and nuclear lamina disassembly to chromosome condensation. The remodeling of these mechanically active nuclear components indicates that mitotic entry is particularly sensitive to forces. Here, we address how mechanical forces crosstalk with the nucleus to determine the timing and efficiency of the G2-M transition. Finally, we discuss how the deregulation of nuclear mechanics has consequences for mitosis.
Collapse
Affiliation(s)
- Joana T Lima
- Epithelial Polarity and Cell Division Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina do Porto, Porto, Portugal
- Programa Doutoral em Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Jorge G Ferreira
- Epithelial Polarity and Cell Division Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina do Porto, Porto, Portugal
| |
Collapse
|
2
|
Qin Q, Zhou ZY, Liu Y, Zhou F, Cao C, Teng L. Unraveling the nexus of nesprin in dilated cardiomyopathy: From molecular insights to therapeutic prospects. Life Sci 2024; 358:123126. [PMID: 39396640 DOI: 10.1016/j.lfs.2024.123126] [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: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Dilated cardiomyopathy is a complex and debilitating heart disorder characterized by the enlargement and weakening of the cardiac chambers, leading to impaired contractility and heart failure. Nesprins, a family of nuclear envelope spectrin repeat proteins that include isoforms Nesprin-1/-2, are integral components of the LInker of Nucleoskeleton and Cytoskeleton complex. They facilitate the connection between the nuclear envelope and the cytoskeleton, crucial for maintaining nuclear architecture, migration and positioning, and mechanical transduction and signaling. Nesprin-1/-2 are abundantly expressed in cardiac and skeletal muscles.They have emerged as key players in the pathogenesis of dilated cardiomyopathy. Mutations in synaptic nuclear envelope-1/-2 genes encoding Nesprin-1/-2 are associated with dilated cardiomyopathy, underscoring their significance in cardiac health. This review highlights the all known cases of Nesprin-1/-2 related dilated cardiomyopathy, focusing on their interactions with the nuclear envelope, their role in mechanical transduction, and their influence on gene expression. Moreover, it delves into the underlying mechanisms through which Nesprin dysfunction disrupts nuclear-cytoskeletal coupling, leading to abnormal nuclear morphology, impaired mechanotransduction, and altered gene regulation. The exploration of Nesprin's impact on dilated cardiomyopathy offers a promising avenue for therapeutic interventions aimed at ameliorating the disease. This review provides a comprehensive overview of recent advancements in understanding the pivotal role of Nesprins in dilated cardiomyopathy research.
Collapse
Affiliation(s)
- Qin Qin
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Zi-Yi Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Yangyuanzhi Liu
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Fei Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China
| | - Chunyu Cao
- School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China; College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Lin Teng
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, UK.
| |
Collapse
|
3
|
Leconte M, Bonne G, Bertrand AT. Recent insights in striated muscle laminopathies. Curr Opin Neurol 2024; 37:509-514. [PMID: 38989655 DOI: 10.1097/wco.0000000000001297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
PURPOSE OF REVIEW To highlight recent insights in different aspects of striated muscle laminopathies (SMLs) related to LMNA mutations. RECENT FINDINGS Clinical and genetic studies allow better patient management and diagnosis, with confirmation of ventricular tachyarrhythmias (VTA) risk prediction score to help with ICD implantation and development of models to help with classification of LMNA variants of uncertain significance. From a pathophysiology perspective, characterization of lamin interactomes in different contexts revealed new lamin A/C partners. Expression or function modulation of these partners evidenced them as potential therapeutic targets. After a positive phase 2, the first phase 3 clinical trial, testing a p38 inhibitor targeting the life-threatening cardiac disease of SML, has been recently stopped, thus highlighting the need for new therapeutic approaches together with new animal and cell models. SUMMARY Since the first LMNA mutation report in 1999, lamin A/C structure and functions have been actively explored to understand the SML pathophysiology. The latest discoveries of partners and altered pathways, highlight the importance of lamin A/C at the nuclear periphery and in the nucleoplasm. Modulation of altered pathways allowed some benefits, especially for cardiac involvement. However, additional studies are still needed to fully assess treatment efficacy and safety.
Collapse
Affiliation(s)
- Marine Leconte
- Sorbonne Université, Inserm, Institut de Myologie, Centre de recherche en Myologie, Paris, France
| | | | | |
Collapse
|
4
|
Liu N, Hsu J, Mahajan G, Sun H, Laurita KR, Naga Prasad SV, Barnard J, Van Wagoner DR, Kothapalli CR, Chung MK, Smith JD. Common SYNE2 Genetic Variant Associated With Atrial Fibrillation Lowers Expression of Nesprin-2α1 With Downstream Effects on Nuclear and Electrophysiological Traits. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004750. [PMID: 39355904 PMCID: PMC11522946 DOI: 10.1161/circgen.124.004750] [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: 05/23/2024] [Accepted: 08/08/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND Atrial fibrillation GWAS (genome-wide association studies) identified significant associations for rs1152591 and linked variants in the SYNE2 gene encoding Nesprin-2, which connects the nuclear membrane with the cytoskeleton. METHODS Reporter gene vector transfection and CRISPR-Cas9 editing were used to identify the causal variant regulating the expression of SYNE2α1. After SYNE2 knockdown or SYNE2α1 overexpression in human stem cell-derived cardiomyocytes, nuclear phenotypes were assessed by imaging and atomic force microscopy. Gene expression was assessed by RNAseq and gene set enrichment analysis. Fura-2 AM staining assessed calcium transients. Optical mapping assessed action potential duration and conduction velocity. RESULTS The risk allele of rs1152591 had lower promoter and enhancer activity and was significantly associated with lower expression of the short SYNE2α1 isoform in human stem cell-derived cardiomyocytes, without an effect on the expression of the full-length SYNE2 mRNA. SYNE2α1 overexpression had dominant negative effects on the nucleus with its overexpression or SYNE2 knockdown leading to increased nuclear area and decreased nuclear stiffness. Gene expression results from SYNE2α1 overexpression demonstrated both concordant and nonconcordant effects with SYNE2 knockdown. SYNE2α1 overexpression had a gain of function on electrophysiology, leading to significantly faster calcium reuptake and decreased assessed action potential duration, while SYNE2 knockdown showed both shortened assessed action potential duration and decreased conduction velocity. CONCLUSIONS rs1152591 was identified as a causal atrial fibrillation variant, with the risk allele decreasing SYNE2α1 expression. Downstream effects of SYNE2α1 overexpression include changes in nuclear stiffness and electrophysiology, which may contribute to the mechanism for the risk allele's association with AF.
Collapse
Affiliation(s)
- Nana Liu
- Depts of Cardiovascular & Metabolic Sciences, Cardiovascular Medicine
| | - Jeffrey Hsu
- Depts of Cardiovascular & Metabolic Sciences, Cardiovascular Medicine
| | - Gautam Mahajan
- Dept of Chemical and Biomedical Engineering, Cleveland State University
| | - Han Sun
- Dept of Quantitative Health Sciences
| | - Kenneth R. Laurita
- Dept of Medicine and Biomedical Engineering, Metrohealth Campus, Cleveland, OH
| | | | | | | | | | - Mina K. Chung
- Depts of Cardiovascular & Metabolic Sciences, Cardiovascular Medicine
- Dept of Cardiovascular Medicine, Cleveland Clinic
| | - Jonathan D. Smith
- Depts of Cardiovascular & Metabolic Sciences, Cardiovascular Medicine
- Dept of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| |
Collapse
|
5
|
Hashimoto K, Ohira M, Kodama A, Kimoto M, Inoue M, Toné S, Usui Y, Hanashima A, Goto T, Ogura Y, Ujihara Y, Mohri S. Loss of connectin novex-3 leads to heart dysfunction associated with impaired cardiomyocyte proliferation and abnormal nuclear mechanics. Sci Rep 2024; 14:13727. [PMID: 38877142 PMCID: PMC11178842 DOI: 10.1038/s41598-024-64608-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 06/11/2024] [Indexed: 06/16/2024] Open
Abstract
Connectin (also known as titin) is a giant striated muscle protein that functions as a molecular spring by providing elasticity to the sarcomere. Novex-3 is a short splice variant of connectin whose physiological function remains unknown. We have recently demonstrated using in vitro analyses that in addition to sarcomere expression, novex-3 was also expressed in cardiomyocyte nuclei exclusively during fetal life, where it provides elasticity/compliance to cardiomyocyte nuclei and promotes cardiomyocyte proliferation in the fetus, suggesting a non-sarcomeric function. Here, we analyzed novex-3 knockout mice to assess the involvement of this function in cardiac pathophysiology in vivo. Deficiency of novex-3 compromised fetal cardiomyocyte proliferation and induced the enlargement of individual cardiomyocytes in neonates. In adults, novex-3 deficiency resulted in chamber dilation and systolic dysfunction, associated with Ca2+ dysregulation, resulting in a reduced life span. Mechanistic analyses revealed a possible association between impaired proliferation and abnormal nuclear mechanics, including stiffer nuclei positioned peripherally with stabilized circumnuclear microtubules in knockout cardiomyocytes. Although the underlying causal relationships were not fully elucidated, these data show that novex-3 has a vital non-sarcomeric function in cardiac pathophysiology and serves as an early contributor to cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Ken Hashimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki, 701-0192, Japan.
| | - Momoko Ohira
- First Department of Physiology, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Aya Kodama
- First Department of Physiology, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Misaki Kimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Mariko Inoue
- Central Research Institute, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Shigenobu Toné
- Laboratory of Molecular Developmental Biology, Graduate School of Science and Engineering, Tokyo Denki University, Hatoyama, Saitama, 350-0394, Japan
| | - Yuu Usui
- First Department of Physiology, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Akira Hanashima
- First Department of Physiology, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Takato Goto
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Nagoya, 466-8555, Japan
| | - Yuhei Ogura
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Nagoya, 466-8555, Japan
| | - Yoshihiro Ujihara
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Nagoya, 466-8555, Japan
| | - Satoshi Mohri
- First Department of Physiology, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| |
Collapse
|
6
|
Zuela-Sopilniak N, Morival J, Lammerding J. Multi-level transcriptomic analysis of LMNA -related dilated cardiomyopathy identifies disease-driving processes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598511. [PMID: 38915720 PMCID: PMC11195185 DOI: 10.1101/2024.06.11.598511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
LMNA- related dilated cardiomyopathy ( LMNA -DCM) is one of the most severe forms of DCM. The incomplete understanding of the molecular disease mechanisms results in lacking treatment options, leading to high mortality amongst patients. Here, using an inducible, cardiomyocyte-specific lamin A/C depletion mouse model, we conducted a comprehensive transcriptomic study, combining both bulk and single nucleus RNA sequencing, and spanning LMNA -DCM disease progression, to identify potential disease drivers. Our refined analysis pipeline identified 496 genes already misregulated early in disease. The expression of these genes was largely driven by disease specific cardiomyocyte sub-populations and involved biological processes mediating cellular response to DNA damage, cytosolic pattern recognition, and innate immunity. Indeed, DNA damage in LMNA -DCM hearts was significantly increased early in disease and correlated with reduced cardiomyocyte lamin A levels. Activation of cytosolic pattern recognition in cardiomyocytes was independent of cGAS, which is rarely expressed in cardiomyocytes, but likely occurred downstream of other pattern recognition sensors such as IFI16. Altered gene expression in cardiac fibroblasts and immune cell infiltration further contributed to tissue-wide changes in gene expression. Our transcriptomic analysis further predicted significant alterations in cell-cell communication between cardiomyocytes, fibroblasts, and immune cells, mediated through early changes in the extracellular matrix (ECM) in the LMNA -DCM hearts. Taken together, our work suggests a model in which nuclear damage in cardiomyocytes leads to activation of DNA damage responses, cytosolic pattern recognition pathway, and other signaling pathways that activate inflammation, immune cell recruitment, and transcriptional changes in cardiac fibroblasts, which collectively drive LMNA -DCM pathogenesis.
Collapse
|
7
|
Kirby TJ, Zahr HC, Fong EHH, Lammerding J. Eliminating elevated p53 signaling fails to rescue skeletal muscle defects or extend survival in lamin A/C-deficient mice. Cell Death Discov 2024; 10:245. [PMID: 38778055 PMCID: PMC11111808 DOI: 10.1038/s41420-024-01998-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Lamins A and C, encoded by the LMNA gene, are nuclear intermediate filaments that provide structural support to the nucleus and contribute to chromatin organization and transcriptional regulation. LMNA mutations cause muscular dystrophies, dilated cardiomyopathy, and other diseases. The mechanisms by which many LMNA mutations result in muscle-specific diseases have remained elusive, presenting a major hurdle in the development of effective treatments. Previous studies using striated muscle laminopathy mouse models found that cytoskeletal forces acting on mechanically fragile Lmna-mutant nuclei led to transient nuclear envelope rupture, extensive DNA damage, and activation of DNA damage response (DDR) pathways in skeletal muscle cells in vitro and in vivo. Furthermore, hearts of Lmna mutant mice have elevated activation of the tumor suppressor protein p53, a central regulator of DDR signaling. We hypothesized that elevated p53 activation could present a pathogenic mechanism in striated muscle laminopathies, and that eliminating p53 activation could improve muscle function and survival in laminopathy mouse models. Supporting a pathogenic function of p53 activation in muscle, stabilization of p53 was sufficient to reduce contractility and viability in wild-type muscle cells in vitro. Using three laminopathy models, we found that increased p53 activity in Lmna-mutant muscle cells primarily resulted from mechanically induced damage to the myonuclei, and not from altered transcriptional regulation due to loss of lamin A/C expression. However, global deletion of p53 in a severe muscle laminopathy model did not reduce the disease phenotype or increase survival, indicating that additional drivers of disease must contribute to the disease pathogenesis.
Collapse
Affiliation(s)
- Tyler J Kirby
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam Movement Sciences, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, Netherlands.
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA.
| | - Hind C Zahr
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Ern Hwei Hannah Fong
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
8
|
Sikder K, Phillips E, Zhong Z, Wang N, Saunders J, Mothy D, Kossenkov A, Schneider T, Nichtova Z, Csordas G, Margulies KB, Choi JC. Perinuclear damage from nuclear envelope deterioration elicits stress responses that contribute to LMNA cardiomyopathy. SCIENCE ADVANCES 2024; 10:eadh0798. [PMID: 38718107 PMCID: PMC11078192 DOI: 10.1126/sciadv.adh0798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/03/2024] [Indexed: 05/12/2024]
Abstract
Mutations in the LMNA gene encoding lamins A/C cause an array of tissue-selective diseases, with the heart being the most commonly affected organ. Despite progress in understanding the perturbations emanating from LMNA mutations, an integrative understanding of the pathogenesis underlying cardiac dysfunction remains elusive. Using a novel conditional deletion model capable of translatome profiling, we observed that cardiomyocyte-specific Lmna deletion in adult mice led to rapid cardiomyopathy with pathological remodeling. Before cardiac dysfunction, Lmna-deleted cardiomyocytes displayed nuclear abnormalities, Golgi dilation/fragmentation, and CREB3-mediated stress activation. Translatome profiling identified MED25 activation, a transcriptional cofactor that regulates Golgi stress. Autophagy is disrupted in the hearts of these mice, which can be recapitulated by disrupting the Golgi. Systemic administration of modulators of autophagy or ER stress significantly delayed cardiac dysfunction and prolonged survival. These studies support a hypothesis wherein stress responses emanating from the perinuclear space contribute to the LMNA cardiomyopathy development.
Collapse
Affiliation(s)
- Kunal Sikder
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Elizabeth Phillips
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Zhijiu Zhong
- Translational Research and Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nadan Wang
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Jasmine Saunders
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - David Mothy
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Andrew Kossenkov
- Bioinformatics Facility, The Wistar Institute Cancer Center, Philadelphia, PA, USA
| | - Timothy Schneider
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zuzana Nichtova
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gyorgy Csordas
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kenneth B. Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| |
Collapse
|
9
|
Zi-Yi Z, Qin Q, Fei Z, Cun-Yu C, Lin T. Nesprin proteins: bridging nuclear envelope dynamics to muscular dysfunction. Cell Commun Signal 2024; 22:208. [PMID: 38566066 PMCID: PMC10986154 DOI: 10.1186/s12964-024-01593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
This review presents a comprehensive exploration of the pivotal role played by the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex, with a particular focus on Nesprin proteins, in cellular mechanics and the pathogenesis of muscular diseases. Distinguishing itself from prior works, the analysis delves deeply into the intricate interplay of the LINC complex, emphasizing its indispensable contribution to maintaining cellular structural integrity, especially in mechanically sensitive tissues such as cardiac and striated muscles. Additionally, the significant association between mutations in Nesprin proteins and the onset of Dilated Cardiomyopathy (DCM) and Emery-Dreifuss Muscular Dystrophy (EDMD) is highlighted, underscoring their pivotal role in disease pathogenesis. Through a comprehensive examination of DCM and EDMD cases, the review elucidates the disruptions in the LINC complex, nuclear morphology alterations, and muscular developmental disorders, thus emphasizing the essential function of an intact LINC complex in preserving muscle physiological functions. Moreover, the review provides novel insights into the implications of Nesprin mutations for cellular dynamics in the pathogenesis of muscular diseases, particularly in maintaining cardiac structural and functional integrity. Furthermore, advanced therapeutic strategies, including rectifying Nesprin gene mutations, controlling Nesprin protein expression, enhancing LINC complex functionality, and augmenting cardiac muscle cell function are proposed. By shedding light on the intricate molecular mechanisms underlying nuclear-cytoskeletal interactions, the review lays the groundwork for future research and therapeutic interventions aimed at addressing genetic muscle disorders.
Collapse
Affiliation(s)
- Zhou Zi-Yi
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Qin Qin
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Zhou Fei
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
| | - Cao Cun-Yu
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
- College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and immunotherapy, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Teng Lin
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China.
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, SE5 9NU, UK.
| |
Collapse
|
10
|
van Heerden D, Klima S, van den Bout I. How nuclear envelope dynamics can direct laminopathy phenotypes. Curr Opin Cell Biol 2024; 86:102290. [PMID: 38048657 DOI: 10.1016/j.ceb.2023.102290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/25/2023] [Accepted: 11/09/2023] [Indexed: 12/06/2023]
Abstract
The nuclear envelope separates the genome from the cytoplasmic environment. However, the nuclear envelope is also physically associated with the genome and exerts influence on gene expression and genome modification. The nucleus is dynamic, changing shape and responding to cell movement, disassembling and assembling during cell division, and undergoing rupture and repair. These dynamics can be impacted by genetic disease, leading to a family of diseases called laminopathies. Their disparate phenotypes suggest that multiple processes are affected. We highlight three such processes here, which we believe can be used to classify most of the laminopathies. While much still needs to be learned, some commonalities between these processes, such as proteins involved in nuclear envelope formation and rupture repair, may drive a variety of laminopathies. Here we review the latest information regarding nuclear dynamics and its role in laminopathies related to mutations in the nuclear lamina and linker of nucleoskeleton and cytoskeleton complex (LINC) proteins.
Collapse
Affiliation(s)
- David van Heerden
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, South Africa; Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, South Africa
| | - Stefanie Klima
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, South Africa; Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, South Africa
| | - Iman van den Bout
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, South Africa; Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, South Africa.
| |
Collapse
|
11
|
West G, Sedighi S, Agnetti G, Taimen P. Intermediate filaments in the heart: The dynamic duo of desmin and lamins orchestrates mechanical force transmission. Curr Opin Cell Biol 2023; 85:102280. [PMID: 37972529 DOI: 10.1016/j.ceb.2023.102280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Abstract
The intermediate filament (IF) cytoskeleton supports cellular structural integrity, particularly in response to mechanical stress. The most abundant IF proteins in mature cardiomyocytes are desmin and lamins. The desmin network tethers the contractile apparatus and organelles to the nuclear envelope and the sarcolemma, while lamins, as components of the nuclear lamina, provide structural stability to the nucleus and the genome. Mutations in desmin or A-type lamins typically result in cardiomyopathies and recent studies emphasized the synergistic roles of desmin and lamins in the maintenance of nuclear integrity in cardiac myocytes. Here we explore the emerging roles of the interdependent relationship between desmin and lamins in providing resilience to nuclear structure while transducing extracellular mechanical cues into the nucleus.
Collapse
Affiliation(s)
- Gun West
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku, 20520, Turku, Finland
| | - Sogol Sedighi
- Johns Hopkins University School of Medicine, 21205, Baltimore, MD, USA
| | - Giulio Agnetti
- Johns Hopkins University School of Medicine, 21205, Baltimore, MD, USA; DIBINEM - University of Bologna, 40123, Bologna, Italy.
| | - Pekka Taimen
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku, 20520, Turku, Finland; Department of Pathology, Turku University Hospital, 20520, Turku, Finland.
| |
Collapse
|
12
|
Jebane C, Varlet AA, Karnat M, Hernandez- Cedillo LM, Lecchi A, Bedu F, Desgrouas C, Vigouroux C, Vantyghem MC, Viallat A, Rupprecht JF, Helfer E, Badens C. Enhanced cell viscosity: A new phenotype associated with lamin A/C alterations. iScience 2023; 26:107714. [PMID: 37701573 PMCID: PMC10494210 DOI: 10.1016/j.isci.2023.107714] [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: 05/03/2023] [Revised: 07/13/2023] [Accepted: 08/22/2023] [Indexed: 09/14/2023] Open
Abstract
Lamin A/C is a well-established key contributor to nuclear stiffness and its role in nucleus mechanical properties has been extensively studied. However, its impact on whole-cell mechanics has been poorly addressed, particularly concerning measurable physical parameters. In this study, we combined microfluidic experiments with theoretical analyses to quantitatively estimate the whole-cell mechanical properties. This allowed us to characterize the mechanical changes induced in cells by lamin A/C alterations and prelamin A accumulation resulting from atazanavir treatment or lipodystrophy-associated LMNA R482W pathogenic variant. Our results reveal a distinctive increase in long-time viscosity as a signature of cells affected by lamin A/C alterations. Furthermore, they show that the whole-cell response to mechanical stress is driven not only by the nucleus but also by the nucleo-cytoskeleton links and the microtubule network. The enhanced cell viscosity assessed with our microfluidic assay could serve as a valuable diagnosis marker for lamin-related diseases.
Collapse
Affiliation(s)
- Cécile Jebane
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | | | - Marc Karnat
- Aix Marseille Univ, Université de Toulon, CNRS, CPT, Turing Centre for Living Systems, Marseille, France
| | | | | | | | | | - Corinne Vigouroux
- Assistance Publique–Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, National Reference Centre for Rares diseases of Insulin-Secretion and Insulin-Sensitivity (PRISIS), Department of Endocrinology, Paris, France
- Sorbonne University, Saint-Antoine Research Centre, Inserm UMR_S938, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Marie-Christine Vantyghem
- Endocrinology, Diabetology and Metabolism Department, Inserm U1190, EGID, Lille University Hospital, Lille, France
| | - Annie Viallat
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | - Jean-François Rupprecht
- Aix Marseille Univ, Université de Toulon, CNRS, CPT, Turing Centre for Living Systems, Marseille, France
| | - Emmanuèle Helfer
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | - Catherine Badens
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- AP-HM, Laboratoire de Biochimie, Marseille, France
| |
Collapse
|
13
|
Tan CY, Chan PS, Tan H, Tan SW, Lee CJM, Wang JW, Ye S, Werner H, Loh YJ, Lee YL, Ackers-Johnson M, Foo RSY, Jiang J. Systematic in vivo candidate evaluation uncovers therapeutic targets for LMNA dilated cardiomyopathy and risk of Lamin A toxicity. J Transl Med 2023; 21:690. [PMID: 37840136 PMCID: PMC10577912 DOI: 10.1186/s12967-023-04542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a severe, non-ischemic heart disease which ultimately results in heart failure (HF). Decades of research on DCM have revealed diverse aetiologies. Among them, familial DCM is the major form of DCM, with pathogenic variants in LMNA being the second most common form of autosomal dominant DCM. LMNA DCM is a multifactorial and complex disease with no specific treatment thus far. Many studies have demonstrated that perturbing candidates related to various dysregulated pathways ameliorate LMNA DCM. However, it is unknown whether these candidates could serve as potential therapeutic targets especially in long term efficacy. METHODS We evaluated 14 potential candidates including Lmna gene products (Lamin A and Lamin C), key signaling pathways (Tgfβ/Smad, mTor and Fgf/Mapk), calcium handling, proliferation regulators and modifiers of LINC complex function in a cardiac specific Lmna DCM model. Positive candidates for improved cardiac function were further assessed by survival analysis. Suppressive roles and mechanisms of these candidates in ameliorating Lmna DCM were dissected by comparing marker gene expression, Tgfβ signaling pathway activation, fibrosis, inflammation, proliferation and DNA damage. Furthermore, transcriptome profiling compared the differences between Lamin A and Lamin C treatment. RESULTS Cardiac function was restored by several positive candidates (Smad3, Yy1, Bmp7, Ctgf, aYAP1, Sun1, Lamin A, and Lamin C), which significantly correlated with suppression of HF/fibrosis marker expression and cardiac fibrosis in Lmna DCM. Lamin C or Sun1 shRNA administration achieved consistent, prolonged survival which highly correlated with reduced heart inflammation and DNA damage. Importantly, Lamin A treatment improved but could not reproduce long term survival, and Lamin A administration to healthy hearts itself induced DCM. Mechanistically, we identified this lapse as caused by a dose-dependent toxicity of Lamin A, which was independent from its maturation. CONCLUSIONS In vivo candidate evaluation revealed that supplementation of Lamin C or knockdown of Sun1 significantly suppressed Lmna DCM and achieve prolonged survival. Conversely, Lamin A supplementation did not rescue long term survival and may impart detrimental cardiotoxicity risk. This study highlights a potential of advancing Lamin C and Sun1 as therapeutic targets for the treatment of LMNA DCM.
Collapse
Affiliation(s)
- Chia Yee Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore
| | - Pui Shi Chan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore
| | - Hansen Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore
| | - Sung Wei Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore
| | - Chang Jie Mick Lee
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore
| | - Jiong-Wei Wang
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Centre for NanoMedicine, Nanomedicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117609, Singapore
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore
| | - Shu Ye
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore
| | - Hendrikje Werner
- Nuevocor Pte Ltd, 1 Biopolis Drive, Amnios, #05-01, Singapore, 138622, Singapore
| | - Ying Jie Loh
- Nuevocor Pte Ltd, 1 Biopolis Drive, Amnios, #05-01, Singapore, 138622, Singapore
| | - Yin Loon Lee
- Nuevocor Pte Ltd, 1 Biopolis Drive, Amnios, #05-01, Singapore, 138622, Singapore
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #06-06, Singapore, 138665, Singapore
| | - Matthew Ackers-Johnson
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore
| | - Roger S Y Foo
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore
| | - Jianming Jiang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Centre for Translational Medicine, Cardiovascular Research Institute (CVRI), National University Health System, 14 Medical Drive, Singapore, 117599, Singapore.
- Cardiovascular Disease Translational Research Programme, NUS Yong Loo Lin School of Medicine, 14 Medical Drive, Level 8, Singapore, 117599, Singapore.
| |
Collapse
|
14
|
Coscarella IL, Landim-Vieira M, Rastegarpouyani H, Chase PB, Irianto J, Pinto JR. Nucleus Mechanosensing in Cardiomyocytes. Int J Mol Sci 2023; 24:13341. [PMID: 37686151 PMCID: PMC10487505 DOI: 10.3390/ijms241713341] [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: 07/14/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Cardiac muscle contraction is distinct from the contraction of other muscle types. The heart continuously undergoes contraction-relaxation cycles throughout an animal's lifespan. It must respond to constantly varying physical and energetic burdens over the short term on a beat-to-beat basis and relies on different mechanisms over the long term. Muscle contractility is based on actin and myosin interactions that are regulated by cytoplasmic calcium ions. Genetic variants of sarcomeric proteins can lead to the pathophysiological development of cardiac dysfunction. The sarcomere is physically connected to other cytoskeletal components. Actin filaments, microtubules and desmin proteins are responsible for these interactions. Therefore, mechanical as well as biochemical signals from sarcomeric contractions are transmitted to and sensed by other parts of the cardiomyocyte, particularly the nucleus which can respond to these stimuli. Proteins anchored to the nuclear envelope display a broad response which remodels the structure of the nucleus. In this review, we examine the central aspects of mechanotransduction in the cardiomyocyte where the transmission of mechanical signals to the nucleus can result in changes in gene expression and nucleus morphology. The correlation of nucleus sensing and dysfunction of sarcomeric proteins may assist the understanding of a wide range of functional responses in the progress of cardiomyopathic diseases.
Collapse
Affiliation(s)
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Hosna Rastegarpouyani
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
- Institute for Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Prescott Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
15
|
Wallace M, Zahr H, Perati S, Morsink CD, Johnson LE, Gacita AM, Lai S, Wallrath LL, Benjamin IJ, McNally EM, Kirby TJ, Lammerding J. Nuclear damage in LMNA mutant iPSC-derived cardiomyocytes is associated with impaired lamin localization to the nuclear envelope. Mol Biol Cell 2023; 34:mbcE21100527. [PMID: 37585285 PMCID: PMC10846625 DOI: 10.1091/mbc.e21-10-0527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
The LMNA gene encodes the nuclear envelope proteins Lamins A and C, which comprise a major part of the nuclear lamina, provide mechanical support to the nucleus, and participate in diverse intracellular signaling. LMNA mutations give rise to a collection of diseases called laminopathies, including dilated cardiomyopathy (LMNA-DCM) and muscular dystrophies. Although nuclear deformities are a hallmark of LMNA-DCM, the role of nuclear abnormalities in the pathogenesis of LMNA-DCM remains incompletely understood. Using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from LMNA mutant patients and healthy controls, we show that LMNA mutant iPSC-CM nuclei have altered shape or increased size compared to healthy control iPSC-CM nuclei. The LMNA mutation exhibiting the most severe nuclear deformities, R249Q, additionally caused reduced nuclear stiffness and increased nuclear fragility. Importantly, for all cell lines, the degree of nuclear abnormalities corresponded to the degree of Lamin A/C and Lamin B1 mislocalization from the nuclear envelope. The mislocalization was likely due to altered assembly of Lamin A/C. Collectively, these results point to the importance of correct lamin assembly at the nuclear envelope in providing mechanical stability to the nucleus and suggest that defects in nuclear lamina organization may contribute to the nuclear and cellular dysfunction in LMNA-DCM.
Collapse
Affiliation(s)
- Melanie Wallace
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
| | - Hind Zahr
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
| | - Shriya Perati
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
| | - Chloé D. Morsink
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, VU Medical Center, 1081 HZ Amsterdam, The Netherlands
| | | | - Anthony M. Gacita
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern Medicine, Chicago, IL 60611
| | - Shuping Lai
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Lori L. Wallrath
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242
| | - Ivor J. Benjamin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern Medicine, Chicago, IL 60611
| | - Tyler J. Kirby
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, VU Medical Center, 1081 HZ Amsterdam, The Netherlands
| | - Jan Lammerding
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Ithaca, NY 14853
| |
Collapse
|
16
|
De Silva S, Fan Z, Kang B, Shanahan CM, Zhang Q. Nesprin-1: novel regulator of striated muscle nuclear positioning and mechanotransduction. Biochem Soc Trans 2023; 51:1331-1345. [PMID: 37171063 PMCID: PMC10317153 DOI: 10.1042/bst20221541] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023]
Abstract
Nesprins (nuclear envelope spectrin repeat proteins) are multi-isomeric scaffolding proteins. Giant nesprin-1 and -2 localise to the outer nuclear membrane, interact with SUN (Sad1p/UNC-84) domain-containing proteins at the inner nuclear membrane to form the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex, which, in association with lamin A/C and emerin, mechanically couples the nucleus to the cytoskeleton. Despite ubiquitous expression of nesprin giant isoforms, pathogenic mutations in nesprin-1 and -2 are associated with tissue-specific disorders, particularly related to striated muscle such as dilated cardiomyopathy and Emery-Dreifuss muscular dystrophy. Recent evidence suggests this muscle-specificity might be attributable in part, to the small muscle specific isoform, nesprin-1α2, which has a novel role in striated muscle function. Our current understanding of muscle-specific functions of nesprin-1 and its isoforms will be summarised in this review to provide insight into potential pathological mechanisms of nesprin-related muscle disease and may inform potential targets of therapeutic modulation.
Collapse
Affiliation(s)
- Shanelle De Silva
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
| | - Zhijuan Fan
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
- Clinical Laboratory, Tianjin Third Central Hospital, Tianjin 300170, China
| | - Baoqiang Kang
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
| | - Catherine M. Shanahan
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
| | - Qiuping Zhang
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
| |
Collapse
|
17
|
Wang H, Krause A, Escobar H, Müthel S, Metzler E, Spuler S. LMNA Co-Regulated Gene Expression as a Suitable Readout after Precise Gene Correction. Int J Mol Sci 2022; 23:ijms232415525. [PMID: 36555163 PMCID: PMC9778678 DOI: 10.3390/ijms232415525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
LMNA-related muscular dystrophy is an autosomal-dominant progressive disorder caused by mutations in LMNA. LMNA missense mutations are becoming correctable with CRISPR/Cas9-derived tools. Evaluating the functional recovery of LMNA after gene editing bears challenges as there is no reported direct loss of function of lamin A/C proteins in patient-derived cells. The proteins encoded by LMNA are lamins A/C, important ubiquitous nuclear envelope proteins but absent in pluripotent stem cells. We induced lamin A/C expression in induced pluripotent stem cells (iPSCs) of two patients with LMNA-related muscular dystrophy, NM_170707.4 (LMNA): c.1366A > G, p.(Asn456Asp) and c.1494G > T, p.(Trp498Cys), using a short three-day, serum-induced differentiation protocol and analyzed expression profiles of co-regulated genes, examples being COL1A2 and S100A6. We then performed precise gene editing of LMNA c.1366A > G using the near-PAMless (PAM: protospacer-adjacent motif) cytosine base editor. We show that the mutation can be repaired to 100% efficiency in individual iPSC clones. The fast differentiation protocol provided a functional readout and demonstrated increased lamin A/C expression as well as normalized expression of co-regulated genes. Collectively, our findings demonstrate the power of CRISPR/Cas9-mediated gene correction and effective outcome measures in a disease with, so far, little perspective on therapies.
Collapse
Affiliation(s)
- Haicui Wang
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Berlin Institute of Health, Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
- Correspondence: (H.W.); (S.S.); Tel.: +49-30-450-540-518 (H.W.); +49-30-450-540-501 (S.S.); Fax: +49-30-450-540-914 (H.W. & S.S.)
| | - Anne Krause
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Berlin Institute of Health, Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Helena Escobar
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Berlin Institute of Health, Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Stefanie Müthel
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Berlin Institute of Health, Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Eric Metzler
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Berlin Institute of Health, Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Berlin Institute of Health, Charité Universitätsmedizin Berlin, 13125 Berlin, Germany
- Correspondence: (H.W.); (S.S.); Tel.: +49-30-450-540-518 (H.W.); +49-30-450-540-501 (S.S.); Fax: +49-30-450-540-914 (H.W. & S.S.)
| |
Collapse
|