1
|
Lee SH, Lim G, Kim H, Suh D, Choi HK, Kim HP, Yoon HG, Park SW, Kang SM, Kwon C, Oh J, Lee SH. Generation of an induced pluripotent stem cell line from a patient with arrhythmogenic right ventricular cardiomyopathy harboring a TMEM43 splice-site variant. Stem Cell Res 2024; 78:103453. [PMID: 38824800 DOI: 10.1016/j.scr.2024.103453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/03/2024] [Accepted: 05/24/2024] [Indexed: 06/04/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a cardiomyopathy that is predominantly inherited and characterized by cardiac arrhythmias and structural abnormalities. TMEM43 (transmembrane protein 43) is one of the well-known genetic culprits behind ACM. In this study, we successfully generated an induced pluripotent stem cell (iPSC) line, YCMi010-A, derived from a male patient diagnosed with ACM. Although these iPSCs harbored a heterozygous intronic splice variant, TMEM43 c.443-2A > G, they still displayed normal cellular morphology and were confirmed to express pluripotency markers. YCMi010-A iPSC line is a promising model for investigating the pathomechanisms associated with ACM and exploring potential therapeutic strategies.
Collapse
Affiliation(s)
- Sun-Ho Lee
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Gibbeum Lim
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyoeun Kim
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - David Suh
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hyo-Kyoung Choi
- Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
| | - Hyoung-Pyo Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Brain Korea 21 Project, Yonsei Genome Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sahng Wook Park
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Institute of Genetic Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seok-Min Kang
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jaewon Oh
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Seung-Hyun Lee
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute of Genetic Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
2
|
Risato G, Brañas Casas R, Cason M, Bueno Marinas M, Pinci S, De Gaspari M, Visentin S, Rizzo S, Thiene G, Basso C, Pilichou K, Tiso N, Celeghin R. In Vivo Approaches to Understand Arrhythmogenic Cardiomyopathy: Perspectives on Animal Models. Cells 2024; 13:1264. [PMID: 39120296 PMCID: PMC11311808 DOI: 10.3390/cells13151264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a hereditary cardiac disorder characterized by the gradual replacement of cardiomyocytes with fibrous and adipose tissue, leading to ventricular wall thinning, chamber dilation, arrhythmias, and sudden cardiac death. Despite advances in treatment, disease management remains challenging. Animal models, particularly mice and zebrafish, have become invaluable tools for understanding AC's pathophysiology and testing potential therapies. Mice models, although useful for scientific research, cannot fully replicate the complexity of the human AC. However, they have provided valuable insights into gene involvement, signalling pathways, and disease progression. Zebrafish offer a promising alternative to mammalian models, despite the phylogenetic distance, due to their economic and genetic advantages. By combining animal models with in vitro studies, researchers can comprehensively understand AC, paving the way for more effective treatments and interventions for patients and improving their quality of life and prognosis.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
- Department of Biology, University of Padua, I-35131 Padua, Italy;
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | | | - Marco Cason
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Maria Bueno Marinas
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Serena Pinci
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Monica De Gaspari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Natascia Tiso
- Department of Biology, University of Padua, I-35131 Padua, Italy;
| | - Rudy Celeghin
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| |
Collapse
|
3
|
Bueno Marinas M, Cason M, Bariani R, Celeghin R, De Gaspari M, Pinci S, Cipriani A, Rigato I, Zorzi A, Rizzo S, Thiene G, Perazzolo Marra M, Corrado D, Basso C, Bauce B, Pilichou K. A Comprehensive Analysis of Non-Desmosomal Rare Genetic Variants in Arrhythmogenic Cardiomyopathy: Integrating in Padua Cohort Literature-Derived Data. Int J Mol Sci 2024; 25:6267. [PMID: 38892455 PMCID: PMC11173278 DOI: 10.3390/ijms25116267] [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: 05/10/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited myocardial disease at risk of sudden death. Genetic testing impacts greatly in ACM diagnosis, but gene-disease associations have yet to be determined for the increasing number of genes included in clinical panels. Genetic variants evaluation was undertaken for the most relevant non-desmosomal disease genes. We retrospectively studied 320 unrelated Italian ACM patients, including 243 cases with predominant right-ventricular (ARVC) and 77 cases with predominant left-ventricular (ALVC) involvement, who did not carry pathogenic/likely pathogenic (P/LP) variants in desmosome-coding genes. The aim was to assess rare genetic variants in transmembrane protein 43 (TMEM43), desmin (DES), phospholamban (PLN), filamin c (FLNC), cadherin 2 (CDH2), and tight junction protein 1 (TJP1), based on current adjudication guidelines and reappraisal on reported literature data. Thirty-five rare genetic variants, including 23 (64%) P/LP, were identified in 39 patients (16/243 ARVC; 23/77 ALVC): 22 FLNC, 9 DES, 2 TMEM43, and 2 CDH2. No P/LP variants were found in PLN and TJP1 genes. Gene-based burden analysis, including P/LP variants reported in literature, showed significant enrichment for TMEM43 (3.79-fold), DES (10.31-fold), PLN (117.8-fold) and FLNC (107-fold). A non-desmosomal rare genetic variant is found in a minority of ARVC patients but in about one third of ALVC patients; as such, clinical decision-making should be driven by genes with robust evidence. More than two thirds of non-desmosomal P/LP variants occur in FLNC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy; (M.B.M.); (M.C.); (R.B.); (R.C.); (M.D.G.); (S.P.); (A.C.); (I.R.); (A.Z.); (S.R.); (G.T.); (M.P.M.); (D.C.); (B.B.); (K.P.)
| | | | | |
Collapse
|
4
|
Desai YB, Parikh VN. Genetic Risk Stratification in Arrhythmogenic Left Ventricular Cardiomyopathy. Card Electrophysiol Clin 2023; 15:391-399. [PMID: 37558308 DOI: 10.1016/j.ccep.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Arrhythmogenic left ventricular cardiomyopathy is characterized by early malignant ventricular arrhythmia associated with varying degrees and times of onset of left ventricular dysfunction. Variants in numerous genes have been associated with this phenotype. Here, the authors review the literature on recent cohort studies of patients with variants in desmoplakin, lamin A/C, filamin-C, phospholamban, RBM20, TMEM43, and selected channelopathy genes also associated with structural disease. Unlike traditional sudden cardiac death risk assessment in nonischemic cardiomyopathy, left ventricular systolic function is an insensitive predictor of risk in patients with these genetic diagnoses.
Collapse
Affiliation(s)
- Yaanik B Desai
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Falk CRVC, 300 Pasteur Drive, Stanford, CA 94305, USA.
| | - Victoria N Parikh
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Falk CRVC, 300 Pasteur Drive, Stanford, CA 94305, USA.
| |
Collapse
|
5
|
Orgil BO, Munkhsaikhan U, Pierre JF, Li N, Xu F, Alberson NR, Johnson JN, Wetzel GT, Boukens BJD, Lu L, Towbin JA, Purevjav E. The TMEM43 S358L mutation affects cardiac, small intestine, and metabolic homeostasis in a knock-in mouse model. Am J Physiol Heart Circ Physiol 2023; 324:H866-H880. [PMID: 37083466 PMCID: PMC10190833 DOI: 10.1152/ajpheart.00712.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/22/2023]
Abstract
The transmembrane protein 43 (TMEM43/LUMA) p.S358L mutation causes arrhythmogenic cardiomyopathy named as ARVC5, a fully penetrant disease with high risk of ventricular arrhythmias, sudden death, and heart failure. Male gender and vigorous exercise independently predicted deleterious outcome. Our systems genetics analysis revealed the importance of Tmem43 for cardiac and metabolic pathways associated with elevated lipid absorption from small intestine. This study sought to delineate gender-specific cardiac, intestinal, and metabolic phenotypes in vivo and investigate underlying pathophysiological mechanisms of S358L mutation. Serial echocardiography, surface electrocardiography (ECG), treadmill running, and body EchoMRI have been used in knock-in heterozygous (Tmem43WT/S358L), homozygous (Tmem43S358L), and wildtype (Tmem43WT) littermate mice. Electron microscopy, histology, immunohistochemistry, transcriptome, and protein analysis have been performed in cardiac and intestinal tissues. Systolic dysfunction was apparent in 3-mo-old Tmem43S358L and 6-mo-old Tmem43WT/S358L mutants. Both mutant lines displayed intolerance to acute stress at 6 mo of age, arrhythmias, fibro-fatty infiltration, and subcellular abnormalities in the myocardium. Microarray analysis found significantly differentially expressed genes between left ventricular (LV) and right ventricular (RV) myocardium. Mutants displayed diminished PPARG activities and significantly reduced TMEM43 and β-catenin expression in the heart, whereas junctional plakoglobin (JUP) translocated into nuclei of mutant cardiomyocytes. Conversely, elongated villi, fatty infiltration, and overexpression of gut epithelial proliferation markers, β-catenin and Ki-67, were evident in small intestine of mutants. We defined Tmem43 S358L-induced pathological effects on cardiac and intestinal homeostasis via distinctly disturbed WNT-β-catenin and PPARG signaling thereby contributing to ARVC5 pathophysiology. Results suggest that cardiometabolic assessment in mutation carriers may be important for predictive and personalized care.NEW & NOTEWORTHY This manuscript describes the findings of our investigation of cardiac, small intestine, and metabolic features of Tmem43-S358L mouse model. By investigating interorgan pathologies, we uncovered multiple mechanisms of the S358L-induced disease, and these unique mechanisms likely appear to contribute to the disease pathogenesis. We hope our findings are important and novel and open new avenues in the hunting for additional diagnostic and therapeutic targets in subjects carrying TMEM43 mutation.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| | - Undral Munkhsaikhan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Joseph F Pierre
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ning Li
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
- Department of Cardiology, Second Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Fuyi Xu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- School of Pharmacy, Binzhou Medical University, Yantai, People's Republic of China
| | - Neely R Alberson
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| | - Jason N Johnson
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| | - Glenn T Wetzel
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| | - Bastiaan J D Boukens
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Jeffrey A Towbin
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
- Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| |
Collapse
|
6
|
Walker SG, Langland CJ, Viles J, Hecker LA, Wallrath LL. Drosophila Models Reveal Properties of Mutant Lamins That Give Rise to Distinct Diseases. Cells 2023; 12:cells12081142. [PMID: 37190051 DOI: 10.3390/cells12081142] [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: 01/26/2023] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Mutations in the LMNA gene cause a collection of diseases known as laminopathies, including muscular dystrophies, lipodystrophies, and early-onset aging syndromes. The LMNA gene encodes A-type lamins, lamins A/C, intermediate filaments that form a meshwork underlying the inner nuclear membrane. Lamins have a conserved domain structure consisting of a head, coiled-coil rod, and C-terminal tail domain possessing an Ig-like fold. This study identified differences between two mutant lamins that cause distinct clinical diseases. One of the LMNA mutations encodes lamin A/C p.R527P and the other codes lamin A/C p.R482W, which are typically associated with muscular dystrophy and lipodystrophy, respectively. To determine how these mutations differentially affect muscle, we generated the equivalent mutations in the Drosophila Lamin C (LamC) gene, an orthologue of human LMNA. The muscle-specific expression of the R527P equivalent showed cytoplasmic aggregation of LamC, a reduced larval muscle size, decreased larval motility, and cardiac defects resulting in a reduced adult lifespan. By contrast, the muscle-specific expression of the R482W equivalent caused an abnormal nuclear shape without a change in larval muscle size, larval motility, and adult lifespan compared to controls. Collectively, these studies identified fundamental differences in the properties of mutant lamins that cause clinically distinct phenotypes, providing insights into disease mechanisms.
Collapse
Affiliation(s)
- Sydney G Walker
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Christopher J Langland
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Jill Viles
- Independent Researcher, Gowrie, IA 50543, USA
| | - Laura A Hecker
- Department of Biology, Clarke University, Dubuque, IA 52001, USA
| | - Lori L Wallrath
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
7
|
Bianchi L, Damiani I, Castiglioni S, Carleo A, De Salvo R, Rossi C, Corsini A, Bellosta S. Smooth Muscle Cell Phenotypic Switch Induced by Traditional Cigarette Smoke Condensate: A Holistic Overview. Int J Mol Sci 2023; 24:ijms24076431. [PMID: 37047404 PMCID: PMC10094728 DOI: 10.3390/ijms24076431] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/19/2023] [Accepted: 03/25/2023] [Indexed: 04/01/2023] Open
Abstract
Cigarette smoke (CS) is a risk factor for inflammatory diseases, such as atherosclerosis. CS condensate (CSC) contains lipophilic components that may represent a systemic cardiac risk factor. To better understand CSC effects, we incubated mouse and human aortic smooth muscle cells (SMCs) with CSC. We evaluated specific markers for contractile [i.e., actin, aortic smooth muscle (ACTA2), calponin-1 (CNN1), the Kruppel-like factor 4 (KLF4), and myocardin (MYOCD) genes] and inflammatory [i.e., IL-1β, and IL-6, IL-8, and galectin-3 (LGALS-3) genes] phenotypes. CSC increased the expression of inflammatory markers and reduced the contractile ones in both cell types, with KLF4 modulating the SMC phenotypic switch. Next, we performed a mass spectrometry-based differential proteomic approach on human SMCs and could show 11 proteins were significantly affected by exposition to CSC (FC ≥ 2.7, p ≤ 0.05). These proteins are active in signaling pathways related to expression of pro-inflammatory cytokines and IFN, inflammasome assembly and activation, cytoskeleton regulation and SMC contraction, mitochondrial integrity and cellular response to oxidative stress, proteostasis control via ubiquitination, and cell proliferation and epithelial-to-mesenchymal transition. Through specific bioinformatics resources, we showed their tight functional correlation in a close interaction niche mainly orchestrated by the interferon-induced double-stranded RNA-activated protein kinase (alternative name: protein kinase RNA-activated; PKR) (EIF2AK2/PKR). Finally, by combining gene expression and protein abundance data we obtained a hybrid network showing reciprocal integration of the CSC-deregulated factors and indicating KLF4 and PKR as the most relevant factors.
Collapse
|
8
|
Using Zebrafish Animal Model to Study the Genetic Underpinning and Mechanism of Arrhythmogenic Cardiomyopathy. Int J Mol Sci 2023; 24:ijms24044106. [PMID: 36835518 PMCID: PMC9966228 DOI: 10.3390/ijms24044106] [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: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is largely an autosomal dominant genetic disorder manifesting fibrofatty infiltration and ventricular arrhythmia with predominantly right ventricular involvement. ACM is one of the major conditions associated with an increased risk of sudden cardiac death, most notably in young individuals and athletes. ACM has strong genetic determinants, and genetic variants in more than 25 genes have been identified to be associated with ACM, accounting for approximately 60% of ACM cases. Genetic studies of ACM in vertebrate animal models such as zebrafish (Danio rerio), which are highly amenable to large-scale genetic and drug screenings, offer unique opportunities to identify and functionally assess new genetic variants associated with ACM and to dissect the underlying molecular and cellular mechanisms at the whole-organism level. Here, we summarize key genes implicated in ACM. We discuss the use of zebrafish models, categorized according to gene manipulation approaches, such as gene knockdown, gene knock-out, transgenic overexpression, and CRISPR/Cas9-mediated knock-in, to study the genetic underpinning and mechanism of ACM. Information gained from genetic and pharmacogenomic studies in such animal models can not only increase our understanding of the pathophysiology of disease progression, but also guide disease diagnosis, prognosis, and the development of innovative therapeutic strategies.
Collapse
|
9
|
Zink M, Seewald A, Rohrbach M, Brodehl A, Liedtke D, Williams T, Childs SJ, Gerull B. Altered Expression of TMEM43 Causes Abnormal Cardiac Structure and Function in Zebrafish. Int J Mol Sci 2022; 23:9530. [PMID: 36076925 PMCID: PMC9455580 DOI: 10.3390/ijms23179530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disease caused by heterozygous missense mutations within the gene encoding for the nuclear envelope protein transmembrane protein 43 (TMEM43). The disease is characterized by myocyte loss and fibro-fatty replacement, leading to life-threatening ventricular arrhythmias and sudden cardiac death. However, the role of TMEM43 in the pathogenesis of ACM remains poorly understood. In this study, we generated cardiomyocyte-restricted transgenic zebrafish lines that overexpress eGFP-linked full-length human wild-type (WT) TMEM43 and two genetic variants (c.1073C>T, p.S358L; c.332C>T, p.P111L) using the Tol2-system. Overexpression of WT and p.P111L-mutant TMEM43 was associated with transcriptional activation of the mTOR pathway and ribosome biogenesis, and resulted in enlarged hearts with cardiomyocyte hypertrophy. Intriguingly, mutant p.S358L TMEM43 was found to be unstable and partially redistributed into the cytoplasm in embryonic and adult hearts. Moreover, both TMEM43 variants displayed cardiac morphological defects at juvenile stages and ultrastructural changes within the myocardium, accompanied by dysregulated gene expression profiles in adulthood. Finally, CRISPR/Cas9 mutants demonstrated an age-dependent cardiac phenotype characterized by heart enlargement in adulthood. In conclusion, our findings suggest ultrastructural remodeling and transcriptomic alterations underlying the development of structural and functional cardiac defects in TMEM43-associated cardiomyopathy.
Collapse
Affiliation(s)
- Miriam Zink
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Anne Seewald
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Mareike Rohrbach
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Daniel Liedtke
- Institute for Human Genetics, Biocenter, Julius-Maximilians-University Würzburg, 97074 Würzburg, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sarah J. Childs
- Department of Biochemistry and Molecular Biology, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Brenda Gerull
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
10
|
Cheng LC, Zhang X, Abhinav K, Nguyen JA, Baboo S, Martinez-Bartolomé S, Branon TC, Ting AY, Loose E, Yates JR, Gerace L. Shared and Distinctive Neighborhoods of Emerin and Lamin B Receptor Revealed by Proximity Labeling and Quantitative Proteomics. J Proteome Res 2022; 21:2197-2210. [PMID: 35972904 PMCID: PMC9442789 DOI: 10.1021/acs.jproteome.2c00281] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Emerin and lamin B receptor (LBR) are abundant transmembrane
proteins
of the nuclear envelope that are concentrated at the inner nuclear
membrane (INM). Although both proteins interact with chromatin and
nuclear lamins, they have distinctive biochemical and functional properties.
Here, we have deployed proximity labeling using the engineered biotin
ligase TurboID (TbID) and quantitative proteomics to compare the neighborhoods
of emerin and LBR in cultured mouse embryonic fibroblasts. Our analysis
revealed 232 high confidence proximity partners that interact selectively
with emerin and/or LBR, 49 of which are shared by both. These included
previously characterized NE-concentrated proteins, as well as a host
of additional proteins not previously linked to emerin or LBR functions.
Many of these are TM proteins of the ER, including two E3 ubiquitin
ligases. Supporting these results, we found that 11/12 representative
proximity relationships identified by TbID also were detected at the
NE with the proximity ligation assay. Overall, this work presents
methodology that may be used for large-scale mapping of the landscape
of the INM and reveals a group of new proteins with potential functional
connections to emerin and LBR.
Collapse
Affiliation(s)
- Li-Chun Cheng
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| | - Xi Zhang
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| | - Kanishk Abhinav
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| | - Julie A Nguyen
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| | - Sabyasachi Baboo
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| | - Salvador Martinez-Bartolomé
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| | - Tess C Branon
- Department of Genetics, Stanford University, Stanford, California 94305, United States
| | - Alice Y Ting
- Department of Genetics, Stanford University, Stanford, California 94305, United States
| | - Esther Loose
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| | - John R Yates
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| | - Larry Gerace
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
| |
Collapse
|
11
|
Klinke N, Meyer H, Ratnavadivel S, Reinhardt M, Heinisch JJ, Malmendal A, Milting H, Paululat A. A Drosophila melanogaster model for TMEM43-related arrhythmogenic right ventricular cardiomyopathy type 5. Cell Mol Life Sci 2022; 79:444. [PMID: 35869176 PMCID: PMC9307560 DOI: 10.1007/s00018-022-04458-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/01/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022]
Abstract
AbstractArrhythmogenic right ventricular cardiomyopathy (ARVC) is a severe cardiac disease that leads to heart failure or sudden cardiac death (SCD). For the pathogenesis of ARVC, various mutations in at least eight different genes have been identified. A rare form of ARVC is associated with the mutation TMEM43 p.S358L, which is a fully penetrant variant in male carriers. TMEM43 p.S358 is homologous to CG8111 p.S333 in Drosophila melanogaster. We established CRISPR/Cas9-mediated CG8111 knock-out mutants in Drosophila, as well as transgenic fly lines carrying an overexpression construct of the CG8111 p.S333L substitution. Knock-out flies developed normally, whereas the overexpression of CG8111 p.S333L caused growth defects, loss of body weight, cardiac arrhythmias, and premature death. An evaluation of a series of model mutants that replaced S333 by selected amino acids proved that the conserved serine is critical for the physiological function of CG8111. Metabolomic and proteomic analyses revealed that the S333 in CG8111 is essential to proper energy homeostasis and lipid metabolism in the fly. Of note, metabolic impairments were also found in the murine Tmem43 disease model, and fibrofatty replacement is a hallmark of human ARVC5. These findings contribute to a more comprehensive understanding of the molecular functions of CG8111 in Drosophila, and can represent a valuable basis to assess the aetiology of the human TMEM43 p.S358L variant in more detail.
Collapse
|
12
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
13
|
Abstract
The nuclear envelope is composed of the nuclear membranes, nuclear lamina, and nuclear pore complexes. Laminopathies are diseases caused by mutations in genes encoding protein components of the lamina and these other nuclear envelope substructures. Mutations in the single gene encoding lamin A and C, which are expressed in most differentiated somatic cells, cause diseases affecting striated muscle, adipose tissue, peripheral nerve, and multiple systems with features of accelerated aging. Mutations in genes encoding other nuclear envelope proteins also cause an array of diseases that selectively affect different tissues or organs. In some instances, the molecular and cellular consequences of laminopathy-causing mutations are known. However, even when these are understood, mechanisms explaining specific tissue or organ pathology remain enigmatic. Current mechanistic hypotheses focus on how alterations in the nuclear envelope may affect gene expression, including via the regulation of signaling pathways, or cellular mechanics, including responses to mechanical stress.
Collapse
Affiliation(s)
- Ji-Yeon Shin
- Department of Medicine and Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Howard J. Worman
- Department of Medicine and Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
14
|
Gu Q, Xu F, Orgil BO, Khuchua Z, Munkhsaikhan U, Johnson JN, Alberson NR, Pierre JF, Black DD, Dong D, Brennan JA, Cathey BM, Efimov IR, Towbin JA, Purevjav E, Lu L. Systems genetics analysis defines importance of TMEM43/ LUMA for cardiac- and metabolic-related pathways. Physiol Genomics 2022; 54:22-35. [PMID: 34766515 PMCID: PMC8721901 DOI: 10.1152/physiolgenomics.00066.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/07/2021] [Accepted: 11/08/2021] [Indexed: 12/31/2022] Open
Abstract
Broad cellular functions and diseases including muscular dystrophy, arrhythmogenic right ventricular cardiomyopathy (ARVC5) and cancer are associated with transmembrane protein43 (TMEM43/LUMA). The study aimed to investigate biological roles of TMEM43 through genetic regulation, gene pathways and gene networks, candidate interacting genes, and up- or downstream regulators. Cardiac transcriptomes from 40 strains of recombinant inbred BXD mice and two parental strains representing murine genetic reference population (GRP) were applied for genetic correlation, functional enrichment, and coexpression network analysis using systems genetics approach. The results were validated in a newly created knock-in Tmem43-S358L mutation mouse model (Tmem43S358L) that displayed signs of cardiac dysfunction, resembling ARVC5 phenotype seen in humans. We found high Tmem43 levels among BXDs with broad variability in expression. Expression of Tmem43 highly negatively correlated with heart mass and heart rate among BXDs, whereas levels of Tmem43 highly positively correlated with plasma high-density lipoproteins (HDL). Through finding differentially expressed genes (DEGs) between Tmem43S358L mutant and wild-type (Tmem43WT) lines, 18 pathways (out of 42 found in BXDs GRP) that are involved in ARVC, hypertrophic cardiomyopathy, dilated cardiomyopathy, nonalcoholic fatty liver disease, Alzheimer's disease, Parkinson's disease, and Huntington's disease were verified. We further constructed Tmem43-mediated gene network, in which Ctnna1, Adcy6, Gnas, Ndufs6, and Uqcrc2 were significantly altered in Tmem43S358L mice versus Tmem43WT controls. Our study defined the importance of Tmem43 for cardiac- and metabolism-related pathways, suggesting that cardiovascular disease-relevant risk factors may also increase risk of metabolic and neurodegenerative diseases via TMEM43-mediated pathways.
Collapse
Affiliation(s)
- Qingqing Gu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Cardiology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Fuyi Xu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Buyan-Ochir Orgil
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee
| | - Zaza Khuchua
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Biochemistry, Sechenov University, Moscow, Russia
| | - Undral Munkhsaikhan
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee
| | - Jason N Johnson
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee
| | - Neely R Alberson
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee
| | - Joseph F Pierre
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee
| | - Dennis D Black
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee
| | - Deli Dong
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jaclyn A Brennan
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Brianna M Cathey
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Jeffrey A Towbin
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee
- Department of Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
15
|
Padilla-Mejia NE, Makarov AA, Barlow LD, Butterfield ER, Field MC. Evolution and diversification of the nuclear envelope. Nucleus 2021; 12:21-41. [PMID: 33435791 PMCID: PMC7889174 DOI: 10.1080/19491034.2021.1874135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Eukaryotic cells arose ~1.5 billion years ago, with the endomembrane system a central feature, facilitating evolution of intracellular compartments. Endomembranes include the nuclear envelope (NE) dividing the cytoplasm and nucleoplasm. The NE possesses universal features: a double lipid bilayer membrane, nuclear pore complexes (NPCs), and continuity with the endoplasmic reticulum, indicating common evolutionary origin. However, levels of specialization between lineages remains unclear, despite distinct mechanisms underpinning various nuclear activities. Several distinct modes of molecular evolution facilitate organellar diversification and to understand which apply to the NE, we exploited proteomic datasets of purified nuclear envelopes from model systems for comparative analysis. We find enrichment of core nuclear functions amongst the widely conserved proteins to be less numerous than lineage-specific cohorts, but enriched in core nuclear functions. This, together with consideration of additional evidence, suggests that, despite a common origin, the NE has evolved as a highly diverse organelle with significant lineage-specific functionality.
Collapse
Affiliation(s)
- Norma E. Padilla-Mejia
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alexandr A. Makarov
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Lael D. Barlow
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Erin R. Butterfield
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Mark C. Field
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České, Czech Republic
| |
Collapse
|
16
|
Shinomiya H, Kato H, Kuramoto Y, Watanabe N, Tsuruda T, Arimura T, Miyashita Y, Miyasaka Y, Mashimo T, Takuwa A, Motooka D, Okuzaki D, Matsuoka K, Tsukamoto O, Hakui H, Yamada N, Lee JK, Kioka H, Kitakaze M, Takashima S, Sakata Y, Asano Y. Aberrant accumulation of TMEM43 accompanied by perturbed transmural gene expression in arrhythmogenic cardiomyopathy. FASEB J 2021; 35:e21994. [PMID: 34674311 DOI: 10.1096/fj.202100800r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 01/06/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) caused by TMEM43 p.S358L is a fully penetrant heart disease that results in impaired cardiac function or fatal arrhythmia. However, the molecular mechanism of ACM caused by the TMEM43 variant has not yet been fully elucidated. In this study, we generated knock-in (KI) rats harboring a Tmem43 p.S358L mutation and established induced pluripotent stem cells (iPSCs) from patients based on the identification of TMEM43 p.S358L variant from a family with ACM. The Tmem43-S358L KI rats exhibited ventricular arrhythmia and fibrotic myocardial replacement in the subepicardium, which recapitulated the human ACM phenotype. The four-transmembrane protein TMEM43 with the p.S358L variant (TMEM43S358L ) was found to be modified by N-linked glycosylation in both KI rat cardiomyocytes and patient-specific iPSC-derived cardiomyocytes. TMEM43S358L glycosylation increased under the conditions of enhanced endoplasmic reticulum (ER) stress caused by pharmacological stimulation or age-dependent decline of the ER function. Intriguingly, the specific glycosylation of TMEM43S358L resulted from the altered membrane topology of TMEM43. Moreover, unlike TMEM43WT , which is mainly localized to the ER, TMEM43S358L accumulated at the nuclear envelope of cardiomyocytes with the increase in glycosylation. Finally, our comprehensive transcriptomic analysis demonstrated that the regional differences in gene expression patterns between the inner and outer layers observed in the wild type myocardium were partially diminished in the KI myocardium prior to exhibiting histological changes indicative of ACM. Altogether, these findings suggest that the aberrant accumulation of TMEM43S358L underlies the pathogenesis of ACM caused by TMEM43 p.S358L variant by affecting the transmural gene expression within the myocardium.
Collapse
Affiliation(s)
- Haruki Shinomiya
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuki Kuramoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Nozomi Watanabe
- Division of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Toshihiro Tsuruda
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Tadaaki Arimura
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yohei Miyashita
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshiki Miyasaka
- Institute of Experimental Animal Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tomoji Mashimo
- Institute of Experimental Animal Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ayako Takuwa
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ken Matsuoka
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hideyuki Hakui
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Noriaki Yamada
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jong-Kook Lee
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hidetaka Kioka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
17
|
Takemoto Y, Kadota S, Minami I, Otsuka S, Okuda S, Abo M, Punzalan LL, Shen Y, Shiba Y, Uesugi M. Chemical Genetics Reveals a Role of Squalene Synthase in TGFβ Signaling and Cardiomyogenesis. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yasushi Takemoto
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Shin Kadota
- Institute for Biomedical Sciences Shinshu University Matsumoto, Nagano 390-8621 Japan
| | - Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS) Kyoto University Kyoto 606-8501 Japan
| | - Shinya Otsuka
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Satoshi Okuda
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Masahiro Abo
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Louvy Lynn Punzalan
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Yan Shen
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Yuji Shiba
- Institute for Biomedical Sciences Shinshu University Matsumoto, Nagano 390-8621 Japan
| | - Motonari Uesugi
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS) Kyoto University Kyoto 606-8501 Japan
- School of Pharmacy Fudan University Shanghai 201203 China
| |
Collapse
|
18
|
Takemoto Y, Kadota S, Minami I, Otsuka S, Okuda S, Abo M, Punzalan LL, Shen Y, Shiba Y, Uesugi M. Chemical Genetics Reveals a Role of Squalene Synthase in TGFβ Signaling and Cardiomyogenesis. Angew Chem Int Ed Engl 2021; 60:21824-21831. [PMID: 34374184 DOI: 10.1002/anie.202100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Indexed: 11/11/2022]
Abstract
KY02111 is a widely used small molecule that boosts cardiomyogenesis of the mesoderm cells derived from pluripotent stem cells, yet its molecular mechanism of action remains elusive. The present study resolves the initially perplexing effects of KY02111 on Wnt signaling and subsequently identifies squalene synthase (SQS) as a molecular target of KY02111 and its optimized version, KY-I. By disrupting the interaction of SQS with cardiac ER-membrane protein TMEM43, KY02111 impairs TGFβ signaling, but not Wnt signaling, and thereby recapitulates the clinical mutation of TMEM43 that causes arrhythmogenic right ventricular cardiomyopathy (ARVC), an inherited heart disease that involves a substitution of myocardium with fatty tissue. These findings reveal a heretofore undescribed role of SQS in TGFβ signaling and cardiomyogenesis. KY02111 may find its use in ARVC modeling as well as serve as a chemical tool for studying TGFβ/SMAD signaling.
Collapse
Affiliation(s)
- Yasushi Takemoto
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Shin Kadota
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, Nagano, 390-8621, Japan
| | - Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Shinya Otsuka
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Satoshi Okuda
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Masahiro Abo
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Louvy Lynn Punzalan
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yan Shen
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yuji Shiba
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, Nagano, 390-8621, Japan
| | - Motonari Uesugi
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan.,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan.,School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
19
|
A nonsense TMEM43 variant leads to disruption of connexin-linked function and autosomal dominant auditory neuropathy spectrum disorder. Proc Natl Acad Sci U S A 2021; 118:2019681118. [PMID: 34050020 PMCID: PMC8179140 DOI: 10.1073/pnas.2019681118] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Genes that are primarily expressed in cochlear glia-like supporting cells (GLSs) have not been clearly associated with progressive deafness. Herein, we present a deafness locus mapped to chromosome 3p25.1 and an auditory neuropathy spectrum disorder (ANSD) gene, TMEM43, mainly expressed in GLSs. We identify p.(Arg372Ter) of TMEM43 by linkage analysis and exome sequencing in two large Asian families segregating ANSD, which is characterized by inability to discriminate speech despite preserved sensitivity to sound. The knock-in mouse with the p.(Arg372Ter) variant recapitulates a progressive hearing loss with histological abnormalities in GLSs. Mechanistically, TMEM43 interacts with the Connexin26 and Connexin30 gap junction channels, disrupting the passive conductance current in GLSs in a dominant-negative fashion when the p.(Arg372Ter) variant is introduced. Based on these mechanistic insights, cochlear implant was performed on three subjects, and speech discrimination was successfully restored. Our study highlights a pathological role of cochlear GLSs by identifying a deafness gene and its causal relationship with ANSD.
Collapse
|
20
|
Fenech EJ, Lari F, Charles PD, Fischer R, Laétitia-Thézénas M, Bagola K, Paton AW, Paton JC, Gyrd-Hansen M, Kessler BM, Christianson JC. Interaction mapping of endoplasmic reticulum ubiquitin ligases identifies modulators of innate immune signalling. eLife 2020; 9:e57306. [PMID: 32614325 PMCID: PMC7332293 DOI: 10.7554/elife.57306] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/11/2020] [Indexed: 12/25/2022] Open
Abstract
Ubiquitin ligases (E3s) embedded in the endoplasmic reticulum (ER) membrane regulate essential cellular activities including protein quality control, calcium flux, and sterol homeostasis. At least 25 different, transmembrane domain (TMD)-containing E3s are predicted to be ER-localised, but for most their organisation and cellular roles remain poorly defined. Using a comparative proteomic workflow, we mapped over 450 protein-protein interactions for 21 stably expressed, full-length E3s. Bioinformatic analysis linked ER-E3s and their interactors to multiple homeostatic, regulatory, and metabolic pathways. Among these were four membrane-embedded interactors of RNF26, a polytopic E3 whose abundance is auto-regulated by ubiquitin-proteasome dependent degradation. RNF26 co-assembles with TMEM43, ENDOD1, TMEM33 and TMED1 to form a complex capable of modulating innate immune signalling through the cGAS-STING pathway. This RNF26 complex represents a new modulatory axis of STING and innate immune signalling at the ER membrane. Collectively, these data reveal the broad scope of regulation and differential functionalities mediated by ER-E3s for both membrane-tethered and cytoplasmic processes.
Collapse
Affiliation(s)
- Emma J Fenech
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Federica Lari
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Philip D Charles
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of OxfordOxfordUnited Kingdom
| | - Roman Fischer
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of OxfordOxfordUnited Kingdom
| | - Marie Laétitia-Thézénas
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of OxfordOxfordUnited Kingdom
| | - Katrin Bagola
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of AdelaideAdelaideAustralia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of AdelaideAdelaideAustralia
| | - Mads Gyrd-Hansen
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Benedikt M Kessler
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of OxfordOxfordUnited Kingdom
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - John C Christianson
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Botnar Research CentreOxfordUnited Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Botnar Research CentreOxfordUnited Kingdom
| |
Collapse
|
21
|
Gerull B, Brodehl A. Genetic Animal Models for Arrhythmogenic Cardiomyopathy. Front Physiol 2020; 11:624. [PMID: 32670084 PMCID: PMC7327121 DOI: 10.3389/fphys.2020.00624] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Arrhythmogenic cardiomyopathy has been clinically defined since the 1980s and causes right or biventricular cardiomyopathy associated with ventricular arrhythmia. Although it is a rare cardiac disease, it is responsible for a significant proportion of sudden cardiac deaths, especially in athletes. The majority of patients with arrhythmogenic cardiomyopathy carry one or more genetic variants in desmosomal genes. In the 1990s, several knockout mouse models of genes encoding for desmosomal proteins involved in cell-cell adhesion revealed for the first time embryonic lethality due to cardiac defects. Influenced by these initial discoveries in mice, arrhythmogenic cardiomyopathy received an increasing interest in human cardiovascular genetics, leading to the discovery of mutations initially in desmosomal genes and later on in more than 25 different genes. Of note, even in the clinic, routine genetic diagnostics are important for risk prediction of patients and their relatives with arrhythmogenic cardiomyopathy. Based on improvements in genetic animal engineering, different transgenic, knock-in, or cardiac-specific knockout animal models for desmosomal and nondesmosomal proteins have been generated, leading to important discoveries in this field. Here, we present an overview about the existing animal models of arrhythmogenic cardiomyopathy with a focus on the underlying pathomechanism and its importance for understanding of this disease. Prospectively, novel mechanistic insights gained from the whole animal, organ, tissue, cellular, and molecular levels will lead to the development of efficient personalized therapies for treatment of arrhythmogenic cardiomyopathy.
Collapse
Affiliation(s)
- Brenda Gerull
- Comprehensive Heart Failure Center Wuerzburg, Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany.,Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center NRW, University Hospitals of the Ruhr-University of Bochum, Bad Oeynhausen, Germany
| |
Collapse
|
22
|
Cheng LC, Baboo S, Lindsay C, Brusman L, Martinez-Bartolomé S, Tapia O, Zhang X, Yates JR, Gerace L. Identification of new transmembrane proteins concentrated at the nuclear envelope using organellar proteomics of mesenchymal cells. Nucleus 2020; 10:126-143. [PMID: 31142202 PMCID: PMC6550788 DOI: 10.1080/19491034.2019.1618175] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The double membrane nuclear envelope (NE), which is contiguous with the ER, contains nuclear pore complexes (NPCs) – the channels for nucleocytoplasmic transport, and the nuclear lamina (NL) – a scaffold for NE and chromatin organization. Since numerous human diseases linked to NE proteins occur in mesenchyme-derived cells, we used proteomics to characterize NE and other subcellular fractions isolated from mesenchymal stem cells and from adipocytes and myocytes. Based on spectral abundance, we calculated enrichment scores for proteins in the NE fractions. We demonstrated by quantitative immunofluorescence microscopy that five little-characterized proteins with high enrichment scores are substantially concentrated at the NE, with Itprip exposed at the outer nuclear membrane, Smpd4 enriched at the NPC, and Mfsd10, Tmx4, and Arl6ip6 likely residing in the inner nuclear membrane. These proteins provide new focal points for studying the functions of the NE. Moreover, our datasets provide a resource for evaluating additional potential NE proteins.
Collapse
Affiliation(s)
- Li-Chun Cheng
- a Department of Molecular Medicine , The Scripps Research Institute , La Jolla , CA , USA
| | - Sabyasachi Baboo
- a Department of Molecular Medicine , The Scripps Research Institute , La Jolla , CA , USA
| | - Cory Lindsay
- a Department of Molecular Medicine , The Scripps Research Institute , La Jolla , CA , USA
| | - Liza Brusman
- a Department of Molecular Medicine , The Scripps Research Institute , La Jolla , CA , USA
| | | | - Olga Tapia
- a Department of Molecular Medicine , The Scripps Research Institute , La Jolla , CA , USA
| | - Xi Zhang
- a Department of Molecular Medicine , The Scripps Research Institute , La Jolla , CA , USA
| | - John R Yates
- a Department of Molecular Medicine , The Scripps Research Institute , La Jolla , CA , USA
| | - Larry Gerace
- a Department of Molecular Medicine , The Scripps Research Institute , La Jolla , CA , USA
| |
Collapse
|
23
|
James C, Müller M, Goldberg MW, Lenz C, Urlaub H, Kehlenbach RH. Proteomic mapping by rapamycin-dependent targeting of APEX2 identifies binding partners of VAPB at the inner nuclear membrane. J Biol Chem 2019; 294:16241-16254. [PMID: 31519755 DOI: 10.1074/jbc.ra118.007283] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 08/05/2019] [Indexed: 11/06/2022] Open
Abstract
Vesicle-associated membrane protein-associated protein B (VAPB) is a tail-anchored protein that is present at several contact sites of the endoplasmic reticulum (ER). We now show by immunoelectron microscopy that VAPB also localizes to the inner nuclear membrane (INM). Using a modified enhanced ascorbate peroxidase 2 (APEX2) approach with rapamycin-dependent targeting of the peroxidase to a protein of interest, we searched for proteins that are in close proximity to VAPB, particularly at the INM. In combination with stable isotope labeling with amino acids in cell culture (SILAC), we confirmed many well-known interaction partners at the level of the ER with a clear distinction between specific and nonspecific hits. Furthermore, we identified emerin, TMEM43, and ELYS as potential interaction partners of VAPB at the INM and the nuclear pore complex, respectively.
Collapse
Affiliation(s)
- Christina James
- Department of Molecular Biology, Faculty of Medicine, Göttingen Center for Molecular Biosciences (GZMB), Georg August University Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Marret Müller
- Department of Molecular Biology, Faculty of Medicine, Göttingen Center for Molecular Biosciences (GZMB), Georg August University Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Martin W Goldberg
- School of Biological and Biomedical Sciences, Durham University, Durham DH1 3LE, United Kingdom
| | - Christof Lenz
- Bioanalytics Group, Institute of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany.,Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Henning Urlaub
- Bioanalytics Group, Institute of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany.,Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Ralph H Kehlenbach
- Department of Molecular Biology, Faculty of Medicine, Göttingen Center for Molecular Biosciences (GZMB), Georg August University Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| |
Collapse
|
24
|
Padrón-Barthe L, Villalba-Orero M, Gómez-Salinero JM, Domínguez F, Román M, Larrasa-Alonso J, Ortiz-Sánchez P, Martínez F, López-Olañeta M, Bonzón-Kulichenko E, Vázquez J, Martí-Gómez C, Santiago DJ, Prados B, Giovinazzo G, Gómez-Gaviro MV, Priori S, Garcia-Pavia P, Lara-Pezzi E. Severe Cardiac Dysfunction and Death Caused by Arrhythmogenic Right Ventricular Cardiomyopathy Type 5 Are Improved by Inhibition of Glycogen Synthase Kinase-3β. Circulation 2019; 140:1188-1204. [PMID: 31567019 PMCID: PMC6784777 DOI: 10.1161/circulationaha.119.040366] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy/arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited cardiac disease characterized by fibrofatty replacement of the myocardium, resulting in heart failure and sudden cardiac death. The most aggressive arrhythmogenic cardiomyopathy/ARVC subtype is ARVC type 5 (ARVC5), caused by a p.S358L mutation in TMEM43 (transmembrane protein 43). The function and localization of TMEM43 are unknown, as is the mechanism by which the p.S358L mutation causes the disease. Here, we report the characterization of the first transgenic mouse model of ARVC5. METHODS We generated transgenic mice overexpressing TMEM43 in either its wild-type or p.S358L mutant (TMEM43-S358L) form in postnatal cardiomyocytes under the control of the α-myosin heavy chain promoter. RESULTS We found that mice expressing TMEM43-S358L recapitulate the human disease and die at a young age. Mutant TMEM43 causes cardiomyocyte death and severe fibrofatty replacement. We also demonstrate that TMEM43 localizes at the nuclear membrane and interacts with emerin and β-actin. TMEM43-S358L shows partial delocalization to the cytoplasm, reduced interaction with emerin and β-actin, and activation of glycogen synthase kinase-3β (GSK3β). Furthermore, we show that targeting cardiac fibrosis has no beneficial effect, whereas overexpression of the calcineurin splice variant calcineurin Aβ1 results in GSK3β inhibition and improved cardiac function and survival. Similarly, treatment of TMEM43 mutant mice with a GSK3β inhibitor improves cardiac function. Finally, human induced pluripotent stem cells bearing the p.S358L mutation also showed contractile dysfunction that was partially restored after GSK3β inhibition. CONCLUSIONS Our data provide evidence that TMEM43-S358L leads to sustained cardiomyocyte death and fibrofatty replacement. Overexpression of calcineurin Aβ1 in TMEM43 mutant mice or chemical GSK3β inhibition improves cardiac function and increases mice life span. Our results pave the way toward new therapeutic approaches for ARVC5.
Collapse
Affiliation(s)
- Laura Padrón-Barthe
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - María Villalba-Orero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Jesús M Gómez-Salinero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Fernando Domínguez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.)
| | - Marta Román
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.)
| | - Javier Larrasa-Alonso
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Paula Ortiz-Sánchez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Fernando Martínez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Marina López-Olañeta
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Elena Bonzón-Kulichenko
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Carlos Martí-Gómez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Demetrio J Santiago
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Belén Prados
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Giovanna Giovinazzo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - María Victoria Gómez-Gaviro
- Departamento de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain (M.V.G.-G.).,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain (M.V.G.-G.)
| | - Silvia Priori
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.).,Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy (S.P.)
| | - Pablo Garcia-Pavia
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.).,Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain (P.G.-P.).,Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain (P.G.-P.)
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,Faculty of Medicine, National Heart & Lung Institute, Imperial College London, UK (E.L.-P.)
| |
Collapse
|
25
|
Yan H, Xin S, Ma J, Wang H, Zhang H, Liu J. A three microRNA-based prognostic signature for small cell lung cancer overall survival. J Cell Biochem 2019; 120:8723-8730. [PMID: 30536412 DOI: 10.1002/jcb.28159] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/08/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND Small-cell lung cancer (SCLC) is one of the most aggressive cancers with mechanisms far from understood. OBJECTIVE We proposed to identify valuable prognostic signature for SCLC prognosis prediction. METHODS microRNA (miRNA) expression profiles of 42 SCLC patients were acquired from the Gene Expression Omnibus. miRNAs that significantly associated with SCLC overall survival (OS-relevant) were identified through univariate Cox regression analysis followed by random survival forest analysis for identification of more reliable miRNA signature. RESULTS Eleven OS-relevant miRNAs were obtained, and hsa-miR-194, hsa-miR-608, and hsa-miR-9 were further refined through RFS. A formula composed of the three miRNAs' expression values weighted by their multivariate Cox regression coefficients was constructed, and based on which, SCLC patients with longer OS could be well distinguished from those with shorter OS. CONCLUSIONS This study should provide a valuable clue for SCLC prognosis evaluation.
Collapse
Affiliation(s)
- Hao Yan
- Department of Oncology, Institute of Integrative Oncology, Tianjin Union Medicine Center, Tianjin, China
| | - Shaobin Xin
- Intensive Care Unit, Tianjin Union Medicine Center, Tianjin, China
| | - Jing Ma
- Department of Integrated Chinese and Western Medicine, Tianjin Anding Hospital, Tianjin, China
| | - Hui Wang
- Department of Oncology, Institute of Integrative Oncology, Tianjin Union Medicine Center, Tianjin, China
| | - Heng Zhang
- Department of Oncology, Institute of Integrative Oncology, Tianjin Union Medicine Center, Tianjin, China
| | - Jindong Liu
- Department of Oncology, Institute of Integrative Oncology, Tianjin Union Medicine Center, Tianjin, China
| |
Collapse
|
26
|
Dubińska-Magiera M, Kozioł K, Machowska M, Piekarowicz K, Filipczak D, Rzepecki R. Emerin Is Required for Proper Nucleus Reassembly after Mitosis: Implications for New Pathogenetic Mechanisms for Laminopathies Detected in EDMD1 Patients. Cells 2019; 8:E240. [PMID: 30871242 PMCID: PMC6468536 DOI: 10.3390/cells8030240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/26/2019] [Accepted: 03/05/2019] [Indexed: 12/29/2022] Open
Abstract
Emerin is an essential LEM (LAP2, Emerin, MAN1) domain protein in metazoans and an integral membrane protein associated with inner and outer nuclear membranes. Mutations in the human EMD gene coding for emerin result in the rare genetic disorder: Emery⁻Dreifuss muscular dystrophy type 1 (EDMD1). This disease belongs to a broader group called laminopathies-a heterogeneous group of rare genetic disorders affecting tissues of mesodermal origin. EDMD1 phenotype is characterized by progressive muscle wasting, contractures of the elbow and Achilles tendons, and cardiac conduction defects. Emerin is involved in many cellular and intranuclear processes through interactions with several partners: lamins; barrier-to-autointegration factor (BAF), β-catenin, actin, and tubulin. Our study demonstrates the presence of the emerin fraction which associates with mitotic spindle microtubules and centrosomes during mitosis and colocalizes during early mitosis with lamin A/C, BAF, and membranes at the mitotic spindle. Transfection studies with cells expressing EGFP-emerin protein demonstrate that the emerin fusion protein fraction also localizes to centrosomes and mitotic spindle microtubules during mitosis. Transient expression of emerin deletion mutants revealed that the resulting phenotypes vary and are mutant dependent. The most frequent phenotypes include aberrant nuclear shape, tubulin network mislocalization, aberrant mitosis, and mislocalization of centrosomes. Emerin deletion mutants demonstrated different chromatin binding capacities in an in vitro nuclear assembly assay and chromatin-binding properties correlated with the strength of phenotypic alteration in transfected cells. Aberrant tubulin staining and microtubule network phenotype appearance depended on the presence of the tubulin binding region in the expressed deletion mutants. We believe that the association with tubulin might help to "deliver" emerin and associated membranes to decondensing chromatin. Preliminary analyses of cells from Polish patients with EDMD1 revealed that for several mutations thought to be null for emerin protein, a truncated emerin protein was present. We infer that the EDMD1 phenotype may be strengthened by the toxicity of truncated emerin expressed in patients with certain nonsense mutations in EMD.
Collapse
Affiliation(s)
- Magda Dubińska-Magiera
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland.
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335 Wroclaw, Poland.
| | - Katarzyna Kozioł
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Magdalena Machowska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Katarzyna Piekarowicz
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Daria Filipczak
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Ryszard Rzepecki
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
27
|
Brull A, Morales Rodriguez B, Bonne G, Muchir A, Bertrand AT. The Pathogenesis and Therapies of Striated Muscle Laminopathies. Front Physiol 2018; 9:1533. [PMID: 30425656 PMCID: PMC6218675 DOI: 10.3389/fphys.2018.01533] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/11/2018] [Indexed: 01/04/2023] Open
Abstract
Emery-Dreifuss muscular dystrophy (EDMD) is a genetic condition characterized by early contractures, skeletal muscle weakness, and cardiomyopathy. During the last 20 years, various genetic approaches led to the identification of causal genes of EDMD and related disorders, all encoding nuclear envelope proteins. By their respective localization either at the inner nuclear membrane or the outer nuclear membrane, these proteins interact with each other and establish a connection between the nucleus and the cytoskeleton. Beside this physical link, these proteins are also involved in mechanotransduction, responding to environmental cues, such as increased tension of the cytoskeleton, by the activation or repression of specific sets of genes. This ability of cells to adapt to environmental conditions is altered in EDMD. Increased knowledge on the pathophysiology of EDMD has led to the development of drug or gene therapies that have been tested on mouse models. This review proposed an overview of the functions played by the different proteins involved in EDMD and related disorders and the current therapeutic approaches tested so far.
Collapse
Affiliation(s)
- Astrid Brull
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| | - Blanca Morales Rodriguez
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France.,Sanofi R&D, Chilly Mazarin, France
| | - Gisèle Bonne
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| | - Antoine Muchir
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| | - Anne T Bertrand
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| |
Collapse
|
28
|
Atrial Structural Remodeling Gene Variants in Patients with Atrial Fibrillation. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4862480. [PMID: 30276209 PMCID: PMC6151856 DOI: 10.1155/2018/4862480] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/30/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022]
Abstract
Atrial fibrillation (AF) is a common arrhythmia for which the genetic studies mainly focused on the genes involved in electrical remodeling, rather than left atrial muscle remodeling. To identify rare variants involved in atrial myopathy using mutational screening, a high-throughput next-generation sequencing (NGS) workflow was developed based on a custom AmpliSeq™ panel of 55 genes potentially involved in atrial myopathy. This workflow was applied to a cohort of 94 patients with AF, 76 with atrial dilatation and 18 without. Bioinformatic analyses used NextGENe® software and in silico tools for variant interpretation. The AmpliSeq custom-made panel efficiently explored 96.58% of the targeted sequences. Based on in silico analysis, 11 potentially pathogenic missense variants were identified that were not previously associated with AF. These variants were located in genes involved in atrial tissue structural remodeling. Three patients were also carriers of potential variants in prevalent arrhythmia-causing genes, usually associated with AF. Most of the variants were found in patients with atrial dilatation (n=9, 82%). This NGS approach was a sensitive and specific method that identified 11 potentially pathogenic variants, which are likely to play roles in the predisposition to left atrial myopathy. Functional studies are needed to confirm their pathogenicity.
Collapse
|
29
|
TMEM43-S358L mutation enhances NF-κB-TGFβ signal cascade in arrhythmogenic right ventricular dysplasia/cardiomyopathy. Protein Cell 2018; 10:104-119. [PMID: 29980933 PMCID: PMC6340891 DOI: 10.1007/s13238-018-0563-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 06/12/2018] [Indexed: 01/29/2023] Open
Abstract
Arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C) is a genetic cardiac muscle disease that accounts for approximately 30% sudden cardiac death in young adults. The Ser358Leu mutation of transmembrane protein 43 (TMEM43) was commonly identified in the patients of highly lethal and fully penetrant ARVD subtype, ARVD5. Here, we generated TMEM43 S358L mouse to explore the underlying mechanism. This mouse strain showed the classic pathologies of ARVD patients, including structural abnormalities and cardiac fibrofatty. TMEM43 S358L mutation led to hyper-activated nuclear factor κB (NF-κB) activation in heart tissues and primary cardiomyocyte cells. Importantly, this hyper activation of NF-κB directly drove the expression of pro-fibrotic gene, transforming growth factor beta (TGFβ1), and enhanced downstream signal, indicating that TMEM43 S358L mutation up-regulates NF-κB-TGFβ signal cascade during ARVD cardiac fibrosis. Our study partially reveals the regulatory mechanism of ARVD development.
Collapse
|
30
|
Linker of nucleoskeleton and cytoskeleton complex proteins in cardiomyopathy. Biophys Rev 2018; 10:1033-1051. [PMID: 29869195 PMCID: PMC6082319 DOI: 10.1007/s12551-018-0431-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/24/2018] [Indexed: 12/21/2022] Open
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex couples the nuclear lamina to the cytoskeleton. The LINC complex and its associated proteins play diverse roles in cells, ranging from genome organization, nuclear morphology, gene expression, to mechanical stability. The importance of a functional LINC complex is highlighted by the large number of mutations in genes encoding LINC complex proteins that lead to skeletal and cardiac myopathies. In this review, the structure, function, and interactions between components of the LINC complex will be described. Mutations that are known to cause cardiomyopathy in patients will be discussed alongside their respective mouse models. Furthermore, future challenges for the field and emerging technologies to investigate LINC complex function will be discussed.
Collapse
|
31
|
Stroud MJ, Fang X, Zhang J, Guimarães-Camboa N, Veevers J, Dalton ND, Gu Y, Bradford WH, Peterson KL, Evans SM, Gerace L, Chen J. Luma is not essential for murine cardiac development and function. Cardiovasc Res 2018; 114:378-388. [PMID: 29040414 PMCID: PMC6019056 DOI: 10.1093/cvr/cvx205] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/11/2017] [Indexed: 12/13/2022] Open
Abstract
AIMS Luma is a recently discovered, evolutionarily conserved protein expressed in mammalian heart, which is associated with the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex. The LINC complex structurally integrates the nucleus and the cytoplasm and plays a critical role in mechanotransduction across the nuclear envelope. Mutations in several LINC components in both humans and mice result in various cardiomyopathies, implying they play essential, non-redundant roles. A single amino acid substitution of serine 358 to leucine (S358L) in Luma is the unequivocal cause of a distinct form of arrhythmogenic cardiomyopathy. However, the role of Luma in heart has remained obscure. In addition, it also remains to be determined how the S358L mutation in Luma leads to cardiomyopathy. METHODS AND RESULTS To determine the role of Luma in the heart, we first determined the expression pattern of Luma in mouse heart. Luma was sporadically expressed in cardiomyocytes throughout the heart, but was highly and uniformly expressed in cardiac fibroblasts and vascular smooth muscle cells. We also generated germline null Luma mice and discovered that germline null mutants were viable and exhibited normal cardiac function. Luma null mice also responded normally to pressure overload induced by transverse aortic constriction. In addition, localization and expression of other LINC complex components in both cardiac myocytes and fibroblasts was unaffected by global loss of Luma. Furthermore, we also generated and characterized Luma S358L knock-in mice, which displayed normal cardiac function and morphology. CONCLUSION Our data suggest that Luma is dispensable for murine cardiac development and function and that the Luma S358L mutation alone may not cause cardiomyopathy in mice.
Collapse
MESH Headings
- Animals
- Arrhythmogenic Right Ventricular Dysplasia/genetics
- Arrhythmogenic Right Ventricular Dysplasia/metabolism
- Cells, Cultured
- Cytoskeleton/metabolism
- Female
- Fibroblasts/metabolism
- Gene Expression Regulation, Developmental
- Genetic Predisposition to Disease
- Heart/embryology
- Heart/physiopathology
- Humans
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mechanotransduction, Cellular
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Morphogenesis
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Smooth Muscle/metabolism
- Nuclear Matrix/metabolism
Collapse
Affiliation(s)
- Matthew J Stroud
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Xi Fang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jianlin Zhang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Nuno Guimarães-Camboa
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer Veevers
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Nancy D Dalton
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yusu Gu
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - William H Bradford
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kirk L Peterson
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Sylvia M Evans
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Larry Gerace
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Corresponding author. Tel: 858 822 4276; fax: 858 822 3027, E-mail:
| |
Collapse
|
32
|
Vélez-Aguilera G, de Dios Gómez-López J, Jiménez-Gutiérrez GE, Vásquez-Limeta A, Laredo-Cisneros MS, Gómez P, Winder SJ, Cisneros B. Control of nuclear β-dystroglycan content is crucial for the maintenance of nuclear envelope integrity and function. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2018; 1865:406-420. [PMID: 29175376 DOI: 10.1016/j.bbamcr.2017.11.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/07/2017] [Accepted: 11/20/2017] [Indexed: 01/28/2023]
Abstract
β-Dystroglycan (β-DG) is a plasma membrane protein that has ability to target to the nuclear envelope (NE) to maintain nuclear architecture. Nevertheless, mechanisms controlling β-DG nuclear localization and the physiological consequences of a failure of trafficking are largely unknown. We show that β-DG has a nuclear export pathway in myoblasts that depends on the recognition of a nuclear export signal located in its transmembrane domain, by CRM1. Remarkably, NES mutations forced β-DG nuclear accumulation resulting in mislocalization and decreased levels of emerin and lamin B1 and disruption of various nuclear processes in which emerin (centrosome-nucleus linkage and β-catenin transcriptional activity) and lamin B1 (cell cycle progression and nucleoli structure) are critically involved. In addition to nuclear export, the lifespan of nuclear β-DG is restricted by its nuclear proteasomal degradation. Collectively our data show that control of nuclear β-DG content by the combination of CRM1 nuclear export and nuclear proteasome pathways is physiologically relevant to preserve proper NE structure and activity.
Collapse
Affiliation(s)
- Griselda Vélez-Aguilera
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan de Dios Gómez-López
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Guadalupe E Jiménez-Gutiérrez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Alejandra Vásquez-Limeta
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico; Laboratory of Protein Dynamics and Signaling, Center for Cancer Research-Frederick, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, United States
| | - Marco S Laredo-Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Pablo Gómez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Steve J Winder
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, United Kingdom
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico.
| |
Collapse
|
33
|
Cho S, Irianto J, Discher DE. Mechanosensing by the nucleus: From pathways to scaling relationships. J Cell Biol 2017; 216:305-315. [PMID: 28043971 PMCID: PMC5294790 DOI: 10.1083/jcb.201610042] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/05/2016] [Accepted: 12/14/2016] [Indexed: 01/01/2023] Open
Abstract
The nucleus is linked mechanically to the extracellular matrix via multiple polymers that transmit forces to the nuclear envelope and into the nuclear interior. Here, we review some of the emerging mechanisms of nuclear mechanosensing, which range from changes in protein conformation and transcription factor localization to chromosome reorganization and membrane dilation up to rupture. Nuclear mechanosensing encompasses biophysically complex pathways that often converge on the main structural proteins of the nucleus, the lamins. We also perform meta-analyses of public transcriptomics and proteomics data, which indicate that some of the mechanosensing pathways relaying signals from the collagen matrix to the nucleus apply to a broad range of species, tissues, and diseases.
Collapse
Affiliation(s)
- Sangkyun Cho
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Jerome Irianto
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Dennis E Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
34
|
Jiang C, Zhu Y, Zhou Z, Gumin J, Bengtsson L, Wu W, Songyang Z, Lang FF, Lin X. TMEM43/LUMA is a key signaling component mediating EGFR-induced NF-κB activation and tumor progression. Oncogene 2016; 36:2813-2823. [DOI: 10.1038/onc.2016.430] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023]
|
35
|
Incessant left ventricular tachycardia of unusual etiology. Indian Pacing Electrophysiol J 2016; 16:104-106. [PMID: 27788994 PMCID: PMC5067854 DOI: 10.1016/j.ipej.2016.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/18/2016] [Indexed: 12/04/2022] Open
Abstract
Apart from Coronary artery disease, left ventricular tachycardia may result from cardiac sarcoidosis, left ventricular tumor, chagas disease and idiopathic left ventricular tachycardia. We report a rare case of incessant left ventricular tachycardia resulting from left dominant arrhythmogenic cardiomyopathy.
Collapse
|
36
|
Meinke P, Schirmer EC. LINC'ing form and function at the nuclear envelope. FEBS Lett 2015; 589:2514-21. [PMID: 26096784 DOI: 10.1016/j.febslet.2015.06.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 06/03/2015] [Accepted: 06/06/2015] [Indexed: 11/15/2022]
Abstract
The nuclear envelope is an amazing piece of engineering. On one hand it is built like a mediaeval fortress with filament systems reinforcing its membrane walls and its double membrane structure forming a lumen like a castle moat. On the other hand its structure can adapt while maintaining its integrity like a reed bending in a river. Like a fortress it has guarded drawbridges in the nuclear pore complexes, but also has other mechanical means of communication. All this is enabled largely because of the LINC complex, a multi-protein structure that connects the intermediate filament nucleoskeleton across the lumen of the double membrane nuclear envelope to multiple cytoplasmic filament systems that themselves could act simultaneously both like mediaeval buttresses and like lines on a suspension bridge. Although many details of the greater LINC structure remain to be discerned, a number of recent findings are giving clues as to how its structural organization can yield such striking dynamic yet stable properties. Combining double- and triple-helical coiled-coils, intrinsic disorder and order, tissue-specific components, and intermediate filaments enables these unique properties.
Collapse
Affiliation(s)
- Peter Meinke
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK.
| | - Eric C Schirmer
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
37
|
Siragam V, Cui X, Masse S, Ackerley C, Aafaqi S, Strandberg L, Tropak M, Fridman MD, Nanthakumar K, Liu J, Sun Y, Su B, Wang C, Liu X, Yan Y, Mendlowitz A, Hamilton RM. TMEM43 mutation p.S358L alters intercalated disc protein expression and reduces conduction velocity in arrhythmogenic right ventricular cardiomyopathy. PLoS One 2014; 9:e109128. [PMID: 25343256 PMCID: PMC4208740 DOI: 10.1371/journal.pone.0109128] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 09/08/2014] [Indexed: 01/04/2023] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a myocardial disease characterized by fibro-fatty replacement of myocardium in the right ventricular free wall and frequently results in life-threatening ventricular arrhythmias and sudden cardiac death. A heterozygous missense mutation in the transmembrane protein 43 (TMEM43) gene, p.S358L, has been genetically identified to cause autosomal dominant ARVC type 5 in a founder population from the island of Newfoundland, Canada. Little is known about the function of the TMEM43 protein or how it leads to the pathogenesis of ARVC. We sought to determine the distribution of TMEM43 and the effect of the p.S358L mutation on the expression and distribution of various intercalated (IC) disc proteins as well as functional effects on IC disc gap junction dye transfer and conduction velocity in cell culture. Through Western blot analysis, transmission electron microscopy (TEM), immunofluorescence (IF), and electrophysiological analysis, our results showed that the stable expression of p.S358L mutation in the HL-1 cardiac cell line resulted in decreased Zonula Occludens (ZO-1) expression and the loss of ZO-1 localization to cell-cell junctions. Junctional Plakoglobin (JUP) and α-catenin proteins were redistributed to the cytoplasm with decreased localization to cell-cell junctions. Connexin-43 (Cx43) phosphorylation was altered, and there was reduced gap junction dye transfer and conduction velocity in mutant TMEM43-transfected cells. These observations suggest that expression of the p.S358L mutant of TMEM43 found in ARVC type 5 may affect localization of proteins involved in conduction, alter gap junction function and reduce conduction velocity in cardiac tissue.
Collapse
Affiliation(s)
- Vinayakumar Siragam
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Xuezhi Cui
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Stephane Masse
- Division of Cardiology, University Health Network, Toronto, Ontario, Canada
| | - Cameron Ackerley
- Division of Pathology, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Shabana Aafaqi
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Linn Strandberg
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Michael Tropak
- Genetics and Genome Biology, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Michael D. Fridman
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | | | - Jun Liu
- Advanced Micro and Nanosystems Laboratory, Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Yu Sun
- Advanced Micro and Nanosystems Laboratory, Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Bin Su
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Caroline Wang
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Xiaoru Liu
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Yuqing Yan
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Ariel Mendlowitz
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| | - Robert M. Hamilton
- Physiology and Experimental Medicine, The Hospital for Sick Children and Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Asimaki A, Saffitz JE. Remodeling of cell-cell junctions in arrhythmogenic cardiomyopathy. ACTA ACUST UNITED AC 2014; 21:13-23. [PMID: 24460198 DOI: 10.3109/15419061.2013.876016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Arrhythmogenic cardiomyopathy (AC) is a primary myocardial disorder characterized by a high incidence of ventricular arrhythmias often preceding the onset of ventricular remodeling and dysfunction. Approximately 50% of patients diagnosed with AC have one or more mutations in genes encoding desmosomal proteins, although non-desmosomal genes have also been associated with the disease. Increasing evidence implicates remodeling of intercalated disk proteins reflecting abnormal responses to mechanical load and aberrant cell signaling pathways in the pathogenesis of AC. This review summarizes recent advances in understanding disease mechanisms in AC that have come from studies of human myocardium and experimental models.
Collapse
Affiliation(s)
- Angeliki Asimaki
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, MA , USA
| | | |
Collapse
|
39
|
Fedorchak GR, Kaminski A, Lammerding J. Cellular mechanosensing: getting to the nucleus of it all. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:76-92. [PMID: 25008017 PMCID: PMC4252489 DOI: 10.1016/j.pbiomolbio.2014.06.009] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 06/28/2014] [Indexed: 12/12/2022]
Abstract
Cells respond to mechanical forces by activating specific genes and signaling pathways that allow the cells to adapt to their physical environment. Examples include muscle growth in response to exercise, bone remodeling based on their mechanical load, or endothelial cells aligning under fluid shear stress. While the involved downstream signaling pathways and mechanoresponsive genes are generally well characterized, many of the molecular mechanisms of the initiating 'mechanosensing' remain still elusive. In this review, we discuss recent findings and accumulating evidence suggesting that the cell nucleus plays a crucial role in cellular mechanotransduction, including processing incoming mechanoresponsive signals and even directly responding to mechanical forces. Consequently, mutations in the involved proteins or changes in nuclear envelope composition can directly impact mechanotransduction signaling and contribute to the development and progression of a variety of human diseases, including muscular dystrophy, cancer, and the focus of this review, dilated cardiomyopathy. Improved insights into the molecular mechanisms underlying nuclear mechanotransduction, brought in part by the emergence of new technologies to study intracellular mechanics at high spatial and temporal resolution, will not only result in a better understanding of cellular mechanosensing in normal cells but may also lead to the development of novel therapies in the many diseases linked to defects in nuclear envelope proteins.
Collapse
Affiliation(s)
- Gregory R Fedorchak
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Ashley Kaminski
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jan Lammerding
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
40
|
Wong X, Luperchio TR, Reddy KL. NET gains and losses: the role of changing nuclear envelope proteomes in genome regulation. Curr Opin Cell Biol 2014; 28:105-20. [PMID: 24886773 DOI: 10.1016/j.ceb.2014.04.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 03/21/2014] [Accepted: 04/11/2014] [Indexed: 01/13/2023]
Abstract
In recent years, our view of the nucleus has changed considerably with an increased awareness of the roles dynamic higher order chromatin structure and nuclear organization play in nuclear function. More recently, proteomics approaches have identified differential expression of nuclear lamina and nuclear envelope transmembrane (NET) proteins. Many NETs have been implicated in a range of developmental disorders as well as cell-type specific biological processes, including genome organization and nuclear morphology. While further studies are needed, it is clear that the differential nuclear envelope proteome contributes to cell-type specific nuclear identity and functions. This review discusses the importance of proteome diversity at the nuclear periphery and highlights the putative roles of NET proteins, with a focus on nuclear architecture.
Collapse
Affiliation(s)
- Xianrong Wong
- Johns Hopkins University, School of Medicine, Department of Biological Chemistry and Center for Epigenetics, 855N. Wolfe St., Rangos 574, Baltimore, MD 21044, United States
| | - Teresa R Luperchio
- Johns Hopkins University, School of Medicine, Department of Biological Chemistry and Center for Epigenetics, 855N. Wolfe St., Rangos 574, Baltimore, MD 21044, United States
| | - Karen L Reddy
- Johns Hopkins University, School of Medicine, Department of Biological Chemistry and Center for Epigenetics, 855N. Wolfe St., Rangos 574, Baltimore, MD 21044, United States.
| |
Collapse
|
41
|
Protein LUMA is a cytoplasmic plaque constituent of various epithelial adherens junctions and composite junctions of myocardial intercalated disks: a unifying finding for cell biology and cardiology. Cell Tissue Res 2014; 357:159-72. [PMID: 24770932 DOI: 10.1007/s00441-014-1865-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/06/2014] [Indexed: 10/25/2022]
Abstract
In a series of recent reports, mutations in the gene encoding a protein called LUMA (or TMEM43), widely speculated to be a tetraspan transmembrane protein of the nuclear envelope, have been associated with a specific subtype of cardiomyopathy (arrhythmogenic cardiomyopathies) and cases of sudden death. However, using antibodies of high specificity in immunolocalization experiments, we have discovered that, in mammals, LUMA is a component of zonula adhaerens and punctum adhaerens plaques of diverse epithelia and epithelial cell cultures and is also located in (or in some species associated with) the plaques of composite junctions (CJs) in myocardiac intercalated disks (IDs). In CJs, LUMA often colocalizes with several other CJ marker proteins. In all these cells, LUMA has not been detected in the nuclear envelope. Surprisingly, under certain conditions, similar CJ localizations have also been seen with some antibodies commercially available for some time. The identification of LUMA as a plaque component of myocardiac CJs leads to reconsiderations of the molecular composition and architecture, the development, the functions, and the pathogenic states of CJs and IDs. These findings now also allow the general conclusion that LUMA has to be added to the list of mutations of cardiomyocyte junction proteins that may be involved in cardiomyopathies.
Collapse
|
42
|
Patel DM, Green KJ. Desmosomes in the Heart: A Review of Clinical and Mechanistic Analyses. ACTA ACUST UNITED AC 2014; 21:109-28. [DOI: 10.3109/15419061.2014.906533] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
Stroud MJ, Banerjee I, Veevers J, Chen J. Linker of nucleoskeleton and cytoskeleton complex proteins in cardiac structure, function, and disease. Circ Res 2014; 114:538-48. [PMID: 24481844 DOI: 10.1161/circresaha.114.301236] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex, composed of proteins within the inner and the outer nuclear membranes, connects the nuclear lamina to the cytoskeleton. The importance of this complex has been highlighted by the discovery of mutations in genes encoding LINC complex proteins, which cause skeletal or cardiac myopathies. Herein, this review summarizes structure, function, and interactions of major components of the LINC complex, highlights how mutations in these proteins may lead to cardiac disease, and outlines future challenges in the field.
Collapse
Affiliation(s)
- Matthew J Stroud
- From the Department of Cardiology, University of California San Diego School of Medicine, La Jolla, CA
| | | | | | | |
Collapse
|
44
|
Milting H, Klauke B, Christensen AH, Musebeck J, Walhorn V, Grannemann S, Munnich T, ari T, Rasmussen TB, Jensen HK, Mogensen J, Baecker C, Romaker E, Laser KT, zu Knyphausen E, Kassner A, Gummert J, Judge DP, Connors S, Hodgkinson K, Young TL, van der Zwaag PA, van Tintelen JP, Anselmetti D. The TMEM43 Newfoundland mutation p.S358L causing ARVC-5 was imported from Europe and increases the stiffness of the cell nucleus. Eur Heart J 2014; 36:872-81. [DOI: 10.1093/eurheartj/ehu077] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 02/03/2014] [Indexed: 02/03/2023] Open
|
45
|
Kaminski A, Fedorchak GR, Lammerding J. The cellular mastermind(?)-mechanotransduction and the nucleus. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 126:157-203. [PMID: 25081618 PMCID: PMC4591053 DOI: 10.1016/b978-0-12-394624-9.00007-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cells respond to mechanical stimulation by activation of specific signaling pathways and genes that allow the cell to adapt to its dynamic physical environment. How cells sense the various mechanical inputs and translate them into biochemical signals remains an area of active investigation. Recent reports suggest that the cell nucleus may be directly implicated in this cellular mechanotransduction process. Taken together, these findings paint a picture of the nucleus as a central hub in cellular mechanotransduction-both structurally and biochemically-with important implications in physiology and disease.
Collapse
Affiliation(s)
- Ashley Kaminski
- Department of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| | - Gregory R Fedorchak
- Department of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| | - Jan Lammerding
- Department of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| |
Collapse
|
46
|
Abstract
Nuclear envelope
spectrin-repeat
proteins (Nesprins), are a novel family of
nuclear and cytoskeletal proteins with rapidly expanding roles as intracellular scaffolds
and linkers. Originally described as proteins that localise to the nuclear envelope (NE)
and establish nuclear-cytoskeletal connections, nesprins have now been found to comprise a
diverse spectrum of tissue specific isoforms that localise to multiple sub-cellular
compartments. Here, we describe how nesprins are necessary in maintaining cellular
architecture by acting as essential scaffolds and linkers at both the NE and other
sub-cellular domains. More importantly, we speculate how nesprin mutations may disrupt
tissue specific nesprin scaffolds and explain the tissue specific nature of many
nesprin-associated diseases, including laminopathies.
Collapse
|
47
|
Baskin B, Skinner JR, Sanatani S, Terespolsky D, Krahn AD, Ray PN, Scherer SW, Hamilton RM. TMEM43 mutations associated with arrhythmogenic right ventricular cardiomyopathy in non-Newfoundland populations. Hum Genet 2013; 132:1245-52. [DOI: 10.1007/s00439-013-1323-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/09/2013] [Indexed: 11/30/2022]
|
48
|
Farina A, Santarelli R, Bloise R, Gonnella R, Granato M, Bei R, Modesti A, Cirone M, Bengtsson L, Angeloni A, Faggioni A. KSHV ORF67 encoded lytic protein localizes on the nuclear membrane and alters emerin distribution. Virus Res 2013; 175:143-50. [PMID: 23623980 DOI: 10.1016/j.virusres.2013.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 03/28/2013] [Accepted: 04/01/2013] [Indexed: 11/18/2022]
Abstract
p29, a newly identified Kaposi's sarcoma-associated herpesvirus (KSHV) protein, is the product of ORF67, the positional homolog of the conserved herpesvirus protein UL34. Like its homologues in other herpesviruses, p29 is expressed early during viral lytic cycle, and is localized on the nuclear rim. Upon chemical induction of viral replication in primary effusion lymphoma cells, p29 interacts with p33, encoded by ORF69, the positional homolog of the conserved herpesvirus protein UL31, and both proteins colocalize on the nuclear membrane. IFA and biochemical analysis of infected or transfected cells showed that p29 expression resulted in delocalization and hyperphosphorylation of emerin, whereas other nuclear lamin associated proteins, such as LUMA, LB1 and LBR were not affected. Mislocalization of emerin was robustly increased upon combined expression of p29 and p33, suggesting that emerin destabilization might represent the first step in nuclear lamina disassembling, a process necessary for nucleocapsid maturation.
Collapse
Affiliation(s)
- Antonella Farina
- Istituto Pasteur Fondazione Cenci Bolognetti, Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hodgkinson KA, Connors SP, Merner N, Haywood A, Young TL, McKenna WJ, Gallagher B, Curtis F, Bassett AS, Parfrey PS. The natural history of a genetic subtype of arrhythmogenic right ventricular cardiomyopathy caused by a p.S358L mutation in TMEM43. Clin Genet 2013; 83:321-31. [DOI: 10.1111/j.1399-0004.2012.01919.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 06/19/2012] [Accepted: 06/19/2012] [Indexed: 01/08/2023]
Affiliation(s)
| | - SP Connors
- Division of Cardiology; Memorial University; St. John's; Newfoundland; Canada
| | - N Merner
- Discipline of Genetics; Memorial University, Health Sciences Centre; St. John's; Newfoundland; Canada
| | - A Haywood
- Discipline of Genetics; Memorial University, Health Sciences Centre; St. John's; Newfoundland; Canada
| | - T-L Young
- Discipline of Genetics; Memorial University, Health Sciences Centre; St. John's; Newfoundland; Canada
| | - WJ McKenna
- Institute of Cardiovascular Science; University College London and The Heart Hospital, University College London Hospitals Trust; London; UK
| | - B Gallagher
- Division of Pathology; James Paton Memorial Hospital; Gander; Newfoundland; Canada
| | - F Curtis
- Discipline of Genetics; Memorial University, Health Sciences Centre; St. John's; Newfoundland; Canada
| | - AS Bassett
- Clinical Genetics Research Program; Centre for Addiction & Mental Health, University of Toronto; Toronto; Ontario; Canada
| | - PS Parfrey
- Clinical Epidemiology Unit; Memorial University, Health Sciences Centre; St. John's; Newfoundland; Canada
| |
Collapse
|
50
|
Simon DN, Wilson KL. Partners and post-translational modifications of nuclear lamins. Chromosoma 2013; 122:13-31. [PMID: 23475188 DOI: 10.1007/s00412-013-0399-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 12/16/2022]
Abstract
Nuclear intermediate filament networks formed by A- and B-type lamins are major components of the nucleoskeleton that are required for nuclear structure and function, with many links to human physiology. Mutations in lamins cause diverse human diseases ('laminopathies'). At least 54 partners interact with human A-type lamins directly or indirectly. The less studied human lamins B1 and B2 have 23 and seven reported partners, respectively. These interactions are likely to be regulated at least in part by lamin post-translational modifications. This review summarizes the binding partners and post-translational modifications of human lamins and discusses their known or potential implications for lamin function.
Collapse
Affiliation(s)
- Dan N Simon
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|