1
|
Foo B, Amedei H, Kaur S, Jaawan S, Boshnakovska A, Gall T, de Boer RA, Silljé HHW, Urlaub H, Rehling P, Lenz C, Lehnart SE. Unbiased complexome profiling and global proteomics analysis reveals mitochondrial impairment and potential changes at the intercalated disk in presymptomatic R14Δ/+ mice hearts. PLoS One 2024; 19:e0311203. [PMID: 39446877 PMCID: PMC11501035 DOI: 10.1371/journal.pone.0311203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/15/2024] [Indexed: 10/26/2024] Open
Abstract
Phospholamban (PLN) is a sarco-endoplasmic reticulum (SER) membrane protein that regulates cardiac contraction/relaxation by reversibly inhibiting the SERCA2a Ca2+-reuptake pump. The R14Δ-PLN mutation causes severe cardiomyopathy that is resistant to conventional treatment. Protein complexes and higher-order supercomplexes such as intercalated disk components and Ca+2-cycling domains underlie many critical cardiac functions, a subset of which may be disrupted by R14Δ-PLN. Complexome profiling (CP) is a proteomics workflow for systematic analysis of high molecular weight (MW) protein complexes and supercomplexes. We hypothesize that R14Δ-PLN may alter a subset of these assemblies, and apply CP workflows to explore these changes in presymptomatic R14Δ/+ mice hearts. Ventricular tissues from presymptomatic 28wk-old WT and R14Δ/+ mice were homogenized under non-denaturing conditions, fractionated by size-exclusion chromatography (SEC) with a linear MW-range exceeding 5 MDa, and subjected to quantitative data-independent acquisition mass spectrometry (DIA-MS) analysis. Unfortunately, current workflows for the systematic analysis of CP data proved ill-suited for use in cardiac samples. Most rely upon curated protein complex databases to provide ground-truth for analysis; however, these are derived primarily from cancerous or immortalized cell lines and, consequently, cell-type specific complexes (including cardiac-specific machinery potentially affected in R14Δ-PLN hearts) are poorly covered. We thus developed PERCOM: a novel CP data-analysis strategy that does not rely upon these databases and can, furthermore, be implemented on widely available spreadsheet software. Applying PERCOM to our CP dataset resulted in the identification of 296 proteins with disrupted elution profiles. Hits were significantly enriched for mitochondrial and intercalated disk (ICD) supercomplex components. Changes to mitochondrial supercomplexes were associated with reduced expression of mitochondrial proteins and maximal oxygen consumption rate. The observed alterations to mitochondrial and ICD supercomplexes were replicated in a second cohort of "juvenile" 9wk-old mice. These early-stage changes to key cardiac machinery may contribute to R14Δ-PLN pathogenesis.
Collapse
Affiliation(s)
- Brian Foo
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Hugo Amedei
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Surmeet Kaur
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Samir Jaawan
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Tanja Gall
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, Rotterdam, the Netherlands
| | - Herman H. W. Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henning Urlaub
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Christof Lenz
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stephan E. Lehnart
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
2
|
Tan S, Yang J, Hu S, Lei W. Cell-cell interactions in the heart: advanced cardiac models and omics technologies. Stem Cell Res Ther 2024; 15:362. [PMID: 39396018 PMCID: PMC11470663 DOI: 10.1186/s13287-024-03982-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/06/2024] [Indexed: 10/14/2024] Open
Abstract
A healthy heart comprises various cell types, including cardiomyocytes, endothelial cells, fibroblasts, immune cells, and among others, which work together to maintain optimal cardiac function. These cells engage in complex communication networks, known as cell-cell interactions (CCIs), which are essential for homeostasis, cardiac structure, and efficient function. However, in the context of cardiac diseases, the heart undergoes damage, leading to alterations in the cellular composition. Such pathological conditions trigger significant changes in CCIs, causing cell rearrangement and the transition between cell types. Studying these interactions can provide valuable insights into cardiac biology and disease mechanisms, enabling the development of new therapeutic strategies. While the development of cardiac organoids and advanced 3D co-culture technologies has revolutionized in vitro studies of CCIs, recent advancements in single-cell and spatial multi-omics technologies provide researchers with powerful and convenient tools to investigate CCIs at unprecedented resolution. This article provides a concise overview of CCIs observed in both normal and injured heart, with an emphasis on the cutting-edge methods used to study these interactions. It highlights recent advancements such as 3D co-culture systems, single-cell and spatial omics technologies, that have enhanced the understanding of CCIs. Additionally, it summarizes the practical applications of CCI research in advancing cardiovascular therapies, offering potential solutions for treating heart disease by targeting intercellular communication.
Collapse
Affiliation(s)
- Shuai Tan
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Jingsi Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
3
|
Badowski C, Benny P, Verma CS, Lane EB. Desmoplakin CSM models unravel mechanisms regulating the binding to intermediate filaments and putative therapeutics for cardiocutaneous diseases. Sci Rep 2024; 14:23206. [PMID: 39369039 PMCID: PMC11455855 DOI: 10.1038/s41598-024-73705-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a common cause of sudden cardiac arrest and death in young adults. It can be induced by different types of mutations throughout the desmoplakin gene including the R2834H mutation in the extreme carboxyterminus tail of desmoplakin (DP CT) which remains structurally uncharacterized and poorly understood. Here, we have created 3D models of DP CT which show the structural effects of AC-inducing mutations as well as the implications of post-translational modifications (PTMs). Our results suggest that, in absence of PTMs, positively charged wildtype DP CT likely folds back onto negatively-charged plectin repeat 14 of nearby plakin repeat domain C (PRD C) contributing to the recruitment of intermediate filaments (IFs). When phosphorylated and methylated, negatively-charged wildtype DP CT would then fold back onto positively-charged plectin repeat 17 of PRD C, promoting the repulsion of intermediate filaments. However, by preventing PTMs, the R2834H mutation would lead to the formation of a cytoplasmic mutant desmoplakin with a constitutively positive DP CT tail that would be aberrantly recruited by cytoplasmic IFs instead of desmosomes, potentially weakening cell-cell contacts and promoting AC. Virtual screening of FDA-approved drug libraries identified several promising drug candidates for the treatment of cardiocutaneous diseases through drug repurposing.
Collapse
Affiliation(s)
- Cedric Badowski
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore.
| | - Paula Benny
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
- Department of Obstetrics and Gynaecology, National University Hospital, National University of Singapore, 1E Kent Ridge Rd, Level 12 NUHS Tower Block, Singapore, 119228, Singapore
- NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Immunos Building, 8A Biomedical Grove, Singapore, Singapore
| | - Chandra S Verma
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, 138671, Singapore
| | - E Birgitte Lane
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore.
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Singapore, 138648, Singapore.
| |
Collapse
|
4
|
Kiener S, Lehner G, Jagannathan V, Welle M, Leeb T. Heterozygous DSP in-frame deletion in a poodle with syndromic ichthyosis involving additional hair and tooth abnormalities. Anim Genet 2024; 55:725-732. [PMID: 39136317 DOI: 10.1111/age.13467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/28/2024] [Accepted: 07/29/2024] [Indexed: 10/23/2024]
Abstract
Ichthyoses comprise a large heterogeneous group of skin disorders, characterized by generalized scaly and hyperkeratotic skin. We investigated a miniature poodle with early onset generalized scaling, dry and irregularly thickened skin, paw pad hyperkeratosis and abnormalities in hair and teeth. The clinical signs of ichthyosis were confirmed by histopathological examination, which revealed mild epidermal hyperplasia and lamellar orthokeratotic hyperkeratosis. A hereditary condition was suspected and a genetic investigation was initiated. We sequenced the whole genome of the affected dog and searched for potentially causative variants in functional candidate genes for the observed phenotype. The analysis revealed a heterozygous in-frame deletion in DSP, NC_049256.1:g.8804542_8804544del resulting from a de novo mutation event as evidenced by genotyping leukocyte DNA from both parents. The 3 bp deletion is predicted to remove one aspartic acid without disrupting the open reading frame (XM_038584124.1:c.1821_1823del, XP_038440052.1:p.(Asp608del)). The DSP gene encodes desmoplakin, a desmosomal plaque protein, responsible for cell-cell adhesion to provide resistance to mechanical stress in epidermal and cardiac tissues. We hypothesize that the deletion of one amino acid in the N-terminal globular head domain acts in a dominant negative manner and thus impairs the proper connection with other proteins. Several variants in DSP in humans and cattle have been described to result in different phenotypes associated with hair and skin abnormalities, sometimes in combination with variable cardiac and/or dental manifestations. In conclusion, we characterized a new syndromic ichthyosis phenotype in a dog and identified a de novo 3 bp deletion in the DSP gene as causal variant.
Collapse
Affiliation(s)
- Sarah Kiener
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Dermfocus, University of Bern, Bern, Switzerland
| | - Georg Lehner
- Kleintierpraxis Lehner - Fachpraxis für Dermatologie, Buch, Germany
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Monika Welle
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Dermfocus, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
D’Elia S, Caputo A, Natale F, Pezzullo E, Limongelli G, Golino P, Cimmino G, Loffredo FS. The Desmoplakin Phenotype Spectrum: Is the Inflammation the "Fil Rouge" Linking Myocarditis, Arrhythmogenic Cardiomyopathy, and Uncommon Autoinflammatory Systemic Disease? Genes (Basel) 2024; 15:1234. [PMID: 39336825 PMCID: PMC11431300 DOI: 10.3390/genes15091234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Myocarditis is an inflammatory condition of cardiac tissue presenting significant variability in clinical manifestations and outcomes. Its etiology is diverse, encompassing infectious agents (primarily viruses, but also bacteria, protozoa, and helminths) and non-infectious factors (autoimmune responses, toxins, and drugs), though often the specific cause remains unidentified. Recent research has highlighted the potential role of genetic susceptibility in the development of myocarditis (and in some cases the development of inflammatory dilated cardiomyopathy, i.e., the condition in which there is chronic inflammation (>3 months) and left ventricular dysfunction\dilatation), with several studies indicating a correlation between myocarditis and genetic backgrounds. Notably, pathogenic genetic variants linked to dilated or arrhythmic cardiomyopathy are found in 8-16% of myocarditis patients. Genetic predispositions can lead to recurrent myocarditis and a higher incidence of ventricular arrhythmias and heart failure. Moreover, the presence of DSP mutations has been associated with distinct pathological patterns and clinical outcomes in arrhythmogenic cardiomyopathy (hot phases). The interplay between genetic factors and environmental triggers, such as viral infections and physical stress, is crucial in understanding the pathogenesis of myocarditis. Identifying these genetic markers can improve the diagnosis, risk stratification, and management of patients with myocarditis, potentially guiding tailored therapeutic interventions. This review aims to synthesize current knowledge on the genetic underpinnings of myocarditis, with an emphasis on desmoplakin-related arrhythmogenic cardiomyopathy, to enhance clinical understanding and inform future research directions.
Collapse
Affiliation(s)
- Saverio D’Elia
- Cardiology Unit, Azienda Ospedaliera Universitaria Luigi Vanvitelli, Piazza Miraglia 2, 80138 Napoli, Italy;
| | - Adriano Caputo
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.C.); (G.L.); (P.G.); (F.S.L.)
| | - Francesco Natale
- Vanvitelli Cardiology and Intensive Care Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (E.P.)
| | - Enrica Pezzullo
- Vanvitelli Cardiology and Intensive Care Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (E.P.)
| | - Giuseppe Limongelli
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.C.); (G.L.); (P.G.); (F.S.L.)
- Inherited and Rare Cardiovascular Diseases, Monaldi Hospital, 80131 Naples, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.C.); (G.L.); (P.G.); (F.S.L.)
| | - Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.C.); (G.L.); (P.G.); (F.S.L.)
| | - Francesco S. Loffredo
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.C.); (G.L.); (P.G.); (F.S.L.)
| |
Collapse
|
6
|
Corrado D, Thiene G, Bauce B, Calore C, Cipriani A, De Lazzari M, Migliore F, Perazzolo Marra M, Pilichou K, Rigato I, Rizzo S, Zorzi A, Basso C. The "Padua classification" of cardiomyopathies: Combining pathobiological basis and morpho-functional remodeling. Int J Cardiol 2024; 418:132571. [PMID: 39306295 DOI: 10.1016/j.ijcard.2024.132571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Over the last 20 years, the scientific progresses in molecular biology and genetics in combination with the increasing use in the clinical setting of contrast-enhanced cardiac magnetic resonance (CMR) for morpho-functional imaging and structural myocardial tissue characterization have provided important new insights into our understanding of the distinctive aspects of cardiomyopathy, regarding both the genetic and biologic background and the clinical phenotypic features. This has led to the need of an appropriate revision and upgrading of current nosographic framework and pathobiological categorization of heart muscle disorders. This article proposes a new definition and classification of cardiomyopathies that rely on the combination of the distinctive pathobiological basis (genetics, molecular biology and pathology) and the clinical phenotypic pattern (morpho-functional and structural features), leading to the proposal of three different disease categories, each of either genetic or non-genetic etiology and characterized by a combined designation based on both "anatomic" and "functional" features, i.e., hypertrophic/restrictive (H/RC), dilated/hypokinetic (D/HC) and scarring/arrhythmogenic cardiomyopathy (S/AC). The clinical application of the newly proposed classification approach in the real-world practice appears crucial to design a targeted clinical management and evaluation of outcomes of affected patients. Although current treatment of cardiomyopathies is largely palliative and based on drugs, catheter ablation, device or surgical interventions aimed to prevent and manage heart failure and malignant arrhythmias, better knowledge of basic mechanisms involved in the onset and progression of pathobiologically different heart muscle diseases may allow to the development of disease-specific curative therapy.
Collapse
Affiliation(s)
- Domenico Corrado
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy.
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Barbara Bauce
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Chiara Calore
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Alberto Cipriani
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Manuel De Lazzari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Federico Migliore
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Martina Perazzolo Marra
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Ilaria Rigato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Alessandro Zorzi
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| |
Collapse
|
7
|
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
|
8
|
Vencato S, Romanato C, Rampazzo A, Calore M. Animal Models and Molecular Pathogenesis of Arrhythmogenic Cardiomyopathy Associated with Pathogenic Variants in Intercalated Disc Genes. Int J Mol Sci 2024; 25:6208. [PMID: 38892395 PMCID: PMC11172742 DOI: 10.3390/ijms25116208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a rare genetic cardiac disease characterized by the progressive substitution of myocardium with fibro-fatty tissue. Clinically, ACM shows wide variability among patients; symptoms can include syncope and ventricular tachycardia but also sudden death, with the latter often being its sole manifestation. Approximately half of ACM patients have been found with variations in one or more genes encoding cardiac intercalated discs proteins; the most involved genes are plakophilin 2 (PKP2), desmoglein 2 (DSG2), and desmoplakin (DSP). Cardiac intercalated discs provide mechanical and electro-metabolic coupling among cardiomyocytes. Mechanical communication is guaranteed by the interaction of proteins of desmosomes and adheren junctions in the so-called area composita, whereas electro-metabolic coupling between adjacent cardiac cells depends on gap junctions. Although ACM has been first described almost thirty years ago, the pathogenic mechanism(s) leading to its development are still only partially known. Several studies with different animal models point to the involvement of the Wnt/β-catenin signaling in combination with the Hippo pathway. Here, we present an overview about the existing murine models of ACM harboring variants in intercalated disc components with a particular focus on the underlying pathogenic mechanisms. Prospectively, mechanistic insights into the disease pathogenesis will lead to the development of effective targeted therapies for ACM.
Collapse
Affiliation(s)
- Sara Vencato
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (S.V.); (C.R.); (A.R.)
| | - Chiara Romanato
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (S.V.); (C.R.); (A.R.)
| | - Alessandra Rampazzo
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (S.V.); (C.R.); (A.R.)
| | - Martina Calore
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (S.V.); (C.R.); (A.R.)
- Department of Molecular Genetics, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| |
Collapse
|
9
|
Rathod M, Franz H, Beyersdorfer V, Wanuske MT, Leal-Fischer K, Hanns P, Stüdle C, Zimmermann A, Buczak K, Schinner C, Spindler V. DPM1 modulates desmosomal adhesion and epidermal differentiation through SERPINB5. J Cell Biol 2024; 223:e202305006. [PMID: 38477878 PMCID: PMC10937187 DOI: 10.1083/jcb.202305006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/30/2023] [Accepted: 01/23/2024] [Indexed: 03/14/2024] Open
Abstract
Glycosylation is essential to facilitate cell-cell adhesion and differentiation. We determined the role of the dolichol phosphate mannosyltransferase (DPM) complex, a central regulator for glycosylation, for desmosomal adhesive function and epidermal differentiation. Deletion of the key molecule of the DPM complex, DPM1, in human keratinocytes resulted in weakened cell-cell adhesion, impaired localization of the desmosomal components desmoplakin and desmoglein-2, and led to cytoskeletal organization defects in human keratinocytes. In a 3D organotypic human epidermis model, loss of DPM1 caused impaired differentiation with abnormally increased cornification, reduced thickness of non-corneal layers, and formation of intercellular gaps in the epidermis. Using proteomic approaches, SERPINB5 was identified as a DPM1-dependent interaction partner of desmoplakin. Mechanistically, SERPINB5 reduced desmoplakin phosphorylation at serine 176, which was required for strong intercellular adhesion. These results uncover a novel role of the DPM complex in connecting desmosomal adhesion with epidermal differentiation.
Collapse
Affiliation(s)
- Maitreyi Rathod
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Anatomy and Experimental Morphology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Henriette Franz
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Vivien Beyersdorfer
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Anatomy and Experimental Morphology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Pauline Hanns
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Chiara Stüdle
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Aude Zimmermann
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Katarzyna Buczak
- Proteomics Core Facility, Biocentre, University of Basel, Basel, Switzerland
| | - Camilla Schinner
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Anatomy and Experimental Morphology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
10
|
Fu M, Hua X, Shu S, Xu X, Zhang H, Peng Z, Mo H, Liu Y, Chen X, Yang Y, Zhang N, Wang X, Liu Z, Yue G, Hu S, Song J. Single-cell RNA sequencing in donor and end-stage heart failure patients identifies NLRP3 as a therapeutic target for arrhythmogenic right ventricular cardiomyopathy. BMC Med 2024; 22:11. [PMID: 38185631 PMCID: PMC10773142 DOI: 10.1186/s12916-023-03232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/14/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Dilation may be the first right ventricular change and accelerates the progression of threatening ventricular tachyarrhythmias and heart failure for patients with arrhythmogenic right ventricular cardiomyopathy (ARVC), but the treatment for right ventricular dilation remains limited. METHODS Single-cell RNA sequencing (scRNA-seq) of blood and biventricular myocardium from 8 study participants was performed, including 6 end-stage heart failure patients with ARVC and 2 normal controls. ScRNA-seq data was then deeply analyzed, including cluster annotation, cellular proportion calculation, and characterization of cellular developmental trajectories and interactions. An integrative analysis of our single-cell data and published genome-wide association study-based data provided insights into the cell-specific contributions to the cardiac arrhythmia phenotype of ARVC. Desmoglein 2 (Dsg2)mut/mut mice were used as the ARVC model to verify the therapeutic effects of pharmacological intervention on identified cellular cluster. RESULTS Right ventricle of ARVC was enriched of CCL3+ proinflammatory macrophages and TNMD+ fibroblasts. Fibroblasts were preferentially affected in ARVC and perturbations associated with ARVC overlap with those reside in genetic variants associated with cardiac arrhythmia. Proinflammatory macrophages strongly interact with fibroblast. Pharmacological inhibition of Nod-like receptor protein 3 (NLRP3), a transcriptional factor predominantly expressed by the CCL3+ proinflammatory macrophages and several other myeloid subclusters, could significantly alleviate right ventricular dilation and dysfunction in Dsg2mut/mut mice (an ARVC mouse model). CONCLUSIONS This study provided a comprehensive analysis of the lineage-specific changes in the blood and myocardium from ARVC patients at a single-cell resolution. Pharmacological inhibition of NLRP3 could prevent right ventricular dilation and dysfunction of mice with ARVC.
Collapse
Affiliation(s)
- Mengxia Fu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Galactophore Department, Galactophore Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Songren Shu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Xinjie Xu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Hang Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Zhiming Peng
- Department of Orthopedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Mo
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| | - Yanyun Liu
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Shaanxi, 710126, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Yicheng Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Ningning Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Xiaohu Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Zirui Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China
| | - Guangxin Yue
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Shengshou Hu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China.
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10037, China.
- The Cardiomyopathy Research Group, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10037, China.
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 10037, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China.
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10037, China.
- The Cardiomyopathy Research Group, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10037, China.
| |
Collapse
|
11
|
Orgil BO, Purevjav E. Molecular Pathways and Animal Models of Cardiomyopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:991-1019. [PMID: 38884766 DOI: 10.1007/978-3-031-44087-8_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
12
|
Zhu Z, Zhang M, Qiu X. Functions and Clinical Significance of Myocardial Cell-Derived Immunoglobulins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1445:119-128. [PMID: 38967754 DOI: 10.1007/978-981-97-0511-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Immunoglobulins (Igs) have been widely accepted to be exclusively expressed by B cells. Nonetheless, this theory is challenged by mounting evidence which suggests that Igs can also be generated by non B cells (non B-Ig), including cardiomyocytes (CM). Non B-Ig exhibits unique physical and chemical characteristics, unique variable region sequences and functions, which diverge from those of B-Ig. For instance, non B-Ig demonstrates hydrophobicity, limited diversity in the variable region, and extracellular matrix protein activity. Likewise, cardiomyocytes can express different classes of Igs, including IgM, IgG, and free Igκ light chains (cardiomyocyte derived-Igs, CM-Igs). In particular, CM-Igs can be secreted into the extracellular space in various cardiovascular diseases, such as myocardial ischaemia and myocardial fibrosis where they might be involved in complement activation and direct damage to cardiomyocytes. Nevertheless, the precise pathological activity of CM-Igs remains unclear. Recently, Zhu et al. focused on studying the sequence characteristics and functions of CM-Igκ; they discovered that the CM-Igκ exhibits a unique VJ recombination pattern, high hydrophobicity, and is principally located on the intercalated discs and cross striations of the cardiomyocytes. Interestingly, loss of Igκ in cardiomyocytes results in structural disorders in intercalated discs and dysfunction in myocardial contraction and conduction. Mechanically, Igκ promotes the stabilisation of plectin, a cytoskeleton cross-linker protein that connects desmin to desomsome, to maintain the normal structure of the intercalated disc. This finding indicates that CM-Igκ plays an integral role in maintaining cytoskeleton structure. Consequently, it is imperative to reveal the physiological functions and mechanisms of pathological injury associated with CM-Igs.
Collapse
Affiliation(s)
- Zhu Zhu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Meng Zhang
- Department of Cardiology, Aerospace Center Hospital, Beijing, China
| | - Xiaoyan Qiu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
13
|
Stevens TL, Coles S, Sturm AC, Hoover CA, Borzok MA, Mohler PJ, El Refaey M. Molecular Pathways and Animal Models of Arrhythmias. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:1057-1090. [PMID: 38884769 DOI: 10.1007/978-3-031-44087-8_67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Arrhythmias account for over 300,000 annual deaths in the United States, and approximately half of all deaths are associated with heart disease. Mechanisms underlying arrhythmia risk are complex; however, work in humans and animal models over the past 25 years has identified a host of molecular pathways linked with both arrhythmia substrates and triggers. This chapter will focus on select arrhythmia pathways solved by linking human clinical and genetic data with animal models.
Collapse
Affiliation(s)
- Tyler L Stevens
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sara Coles
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Amy C Sturm
- Genomic Medicine Institute, 23andMe, Sunnyvale, CA, USA
| | - Catherine A Hoover
- Department of Biochemistry, Chemistry, Engineering and Physics, Commonwealth University of Pennsylvania, Mansfield, PA, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Maegen A Borzok
- Department of Biochemistry, Chemistry, Engineering and Physics, Commonwealth University of Pennsylvania, Mansfield, PA, USA
| | - Peter J Mohler
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mona El Refaey
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
14
|
Fan X, Yang G, Duru F, Grilli M, Akin I, Zhou X, Saguner AM, Ei-Battrawy I. Arrhythmogenic Cardiomyopathy: from Preclinical Models to Genotype-phenotype Correlation and Pathophysiology. Stem Cell Rev Rep 2023; 19:2683-2708. [PMID: 37731079 PMCID: PMC10661732 DOI: 10.1007/s12015-023-10615-0] [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] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a hereditary myocardial disease characterized by the replacement of the ventricular myocardium with fibrous fatty deposits. ACM is usually inherited in an autosomal dominant pattern with variable penetrance and expressivity, which is mainly related to ventricular tachyarrhythmia and sudden cardiac death (SCD). Importantly, significant progress has been made in determining the genetic background of ACM due to the development of new techniques for genetic analysis. The exact molecular pathomechanism of ACM, however, is not completely clear and the genotype-phenotype correlations have not been fully elucidated, which are useful to predict the prognosis and treatment of ACM patients. Different gene-targeted and transgenic animal models, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models, and heterologous expression systems have been developed. Here, this review aims to summarize preclinical ACM models and platforms promoting our understanding of the pathogenesis of ACM and assess their value in elucidating the ACM genotype-phenotype relationship.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Guoqiang Yang
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Acupuncture and Rehabilitation, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Research Unit of Molecular Imaging Probes, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Firat Duru
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Maurizio Grilli
- Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Xiaobo Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- First Department of Medicine, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Ardan Muammer Saguner
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Ibrahim Ei-Battrawy
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- Department of Cardiology and Angiology, Ruhr University, Bochum, Germany; Institute of Physiology, Department of Cellular and Translational Physiology and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr- University Bochum, Bochum, Germany.
| |
Collapse
|
15
|
Nielsen MS, van Opbergen CJM, van Veen TAB, Delmar M. The intercalated disc: a unique organelle for electromechanical synchrony in cardiomyocytes. Physiol Rev 2023; 103:2271-2319. [PMID: 36731030 PMCID: PMC10191137 DOI: 10.1152/physrev.00021.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The intercalated disc (ID) is a highly specialized structure that connects cardiomyocytes via mechanical and electrical junctions. Although described in some detail by light microscopy in the 19th century, it was in 1966 that electron microscopy images showed that the ID represented apposing cell borders and provided detailed insight into the complex ID nanostructure. Since then, much has been learned about the ID and its molecular composition, and it has become evident that a large number of proteins, not all of them involved in direct cell-to-cell coupling via mechanical or gap junctions, reside at the ID. Furthermore, an increasing number of functional interactions between ID components are emerging, leading to the concept that the ID is not the sum of isolated molecular silos but an interacting molecular complex, an "organelle" where components work in concert to bring about electrical and mechanical synchrony. The aim of the present review is to give a short historical account of the ID's discovery and an updated overview of its composition and organization, followed by a discussion of the physiological implications of the ID architecture and the local intermolecular interactions. The latter will focus on both the importance of normal conduction of cardiac action potentials as well as the impact on the pathophysiology of arrhythmias.
Collapse
Affiliation(s)
- Morten S Nielsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chantal J M van Opbergen
- The Leon Charney Division of Cardiology, New York University Grossmann School of Medicine, New York, New York, United States
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mario Delmar
- The Leon Charney Division of Cardiology, New York University Grossmann School of Medicine, New York, New York, United States
| |
Collapse
|
16
|
van Kampen SJ, Han SJ, van Ham WB, Kyriakopoulou E, Stouthart EW, Goversen B, Monshouwer-Kloots J, Perini I, de Ruiter H, van der Kraak P, Vink A, van Laake LW, Groeneweg JA, de Boer TP, Tsui H, Boogerd CJ, van Veen TAB, van Rooij E. PITX2 induction leads to impaired cardiomyocyte function in arrhythmogenic cardiomyopathy. Stem Cell Reports 2023; 18:749-764. [PMID: 36868229 PMCID: PMC10031305 DOI: 10.1016/j.stemcr.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 03/05/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited progressive disease characterized by electrophysiological and structural remodeling of the ventricles. However, the disease-causing molecular pathways, as a consequence of desmosomal mutations, are poorly understood. Here, we identified a novel missense mutation within desmoplakin in a patient clinically diagnosed with ACM. Using CRISPR-Cas9, we corrected this mutation in patient-derived human induced pluripotent stem cells (hiPSCs) and generated an independent knockin hiPSC line carrying the same mutation. Mutant cardiomyocytes displayed a decline in connexin 43, NaV1.5, and desmosomal proteins, which was accompanied by a prolonged action potential duration. Interestingly, paired-like homeodomain 2 (PITX2), a transcription factor that acts a repressor of connexin 43, NaV1.5, and desmoplakin, was induced in mutant cardiomyocytes. We validated these results in control cardiomyocytes in which PITX2 was either depleted or overexpressed. Importantly, knockdown of PITX2 in patient-derived cardiomyocytes is sufficient to restore the levels of desmoplakin, connexin 43, and NaV1.5.
Collapse
Affiliation(s)
- Sebastiaan J van Kampen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Su Ji Han
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Willem B van Ham
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eirini Kyriakopoulou
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Elizabeth W Stouthart
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Birgit Goversen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, the Netherlands
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Ilaria Perini
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Hesther de Ruiter
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Petra van der Kraak
- Department of Pathology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Aryan Vink
- Department of Pathology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Linda W van Laake
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Judith A Groeneweg
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hoyee Tsui
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Cornelis J Boogerd
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
17
|
Wang Y, Dobreva G. Epigenetics in LMNA-Related Cardiomyopathy. Cells 2023; 12:cells12050783. [PMID: 36899919 PMCID: PMC10001118 DOI: 10.3390/cells12050783] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/18/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Mutations in the gene for lamin A/C (LMNA) cause a diverse range of diseases known as laminopathies. LMNA-related cardiomyopathy is a common inherited heart disease and is highly penetrant with a poor prognosis. In the past years, numerous investigations using mouse models, stem cell technologies, and patient samples have characterized the phenotypic diversity caused by specific LMNA variants and contributed to understanding the molecular mechanisms underlying the pathogenesis of heart disease. As a component of the nuclear envelope, LMNA regulates nuclear mechanostability and function, chromatin organization, and gene transcription. This review will focus on the different cardiomyopathies caused by LMNA mutations, address the role of LMNA in chromatin organization and gene regulation, and discuss how these processes go awry in heart disease.
Collapse
Affiliation(s)
- Yinuo Wang
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), 68167 Mannheim, Germany
- Correspondence: (Y.W.); (G.D.)
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), 68167 Mannheim, Germany
- Correspondence: (Y.W.); (G.D.)
| |
Collapse
|
18
|
Mendelson JB, Sternbach JD, Doyle MJ, Mills L, Hartweck LM, Tollison W, Carney JP, Lahti MT, Bianco RW, Kalra R, Kazmirczak F, Hindmarch C, Archer SL, Prins KW, Martin CM. A Multi-omic and Multi-Species Analysis of Right Ventricular Failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527661. [PMID: 36798212 PMCID: PMC9934613 DOI: 10.1101/2023.02.08.527661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Right ventricular failure (RVF) is a leading cause of morbidity and mortality in multiple cardiovascular diseases, but there are no approved treatments for RVF as therapeutic targets are not clearly defined. Contemporary transcriptomic/proteomic evaluations of RVF are predominately conducted in small animal studies, and data from large animal models are sparse. Moreover, a comparison of the molecular mediators of RVF across species is lacking. Here, we used transcriptomics and proteomics analyses to define the molecular pathways associated with cardiac MRI-derived values of RV hypertrophy, dilation, and dysfunction in pulmonary artery banded (PAB) piglets. Publicly available data from rat monocrotaline-induced RVF and pulmonary arterial hypertension patients with preserved or impaired RV function were used to compare the three species. Transcriptomic and proteomic analyses identified multiple pathways that were associated with RV dysfunction and remodeling in PAB pigs. Surprisingly, disruptions in fatty acid oxidation (FAO) and electron transport chain (ETC) proteins were different across the three species. FAO and ETC proteins and transcripts were mostly downregulated in rats, but were predominately upregulated in PAB pigs, which more closely matched the human data. Thus, the pig PAB metabolic molecular signature was more similar to human RVF than rodents. These data suggest there may be divergent molecular responses of RVF across species, and that pigs more accurately recapitulate the metabolic aspects of human RVF.
Collapse
|
19
|
Borie R, Cardwell J, Konigsberg IR, Moore CM, Zhang W, Sasse SK, Gally F, Dobrinskikh E, Walts A, Powers J, Brancato J, Rojas M, Wolters PJ, Brown KK, Blackwell TS, Nakanishi T, Richards JB, Gerber AN, Fingerlin TE, Sachs N, Pulit SL, Zappala Z, Schwartz DA, Yang IV. Colocalization of Gene Expression and DNA Methylation with Genetic Risk Variants Supports Functional Roles of MUC5B and DSP in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2022; 206:1259-1270. [PMID: 35816432 PMCID: PMC9746850 DOI: 10.1164/rccm.202110-2308oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Common genetic variants have been associated with idiopathic pulmonary fibrosis (IPF). Objectives: To determine functional relevance of the 10 IPF-associated common genetic variants we previously identified. Methods: We performed expression quantitative trait loci (eQTL) and methylation quantitative trait loci (mQTL) mapping, followed by co-localization of eQTL and mQTL with genetic association signals and functional validation by luciferase reporter assays. Illumina multi-ethnic genotyping arrays, mRNA sequencing, and Illumina 850k methylation arrays were performed on lung tissue of participants with IPF (234 RNA and 345 DNA samples) and non-diseased controls (188 RNA and 202 DNA samples). Measurements and Main Results: Focusing on genetic variants within 10 IPF-associated genetic loci, we identified 27 eQTLs in controls and 24 eQTLs in cases (false-discovery-rate-adjusted P < 0.05). Among these signals, we identified associations of lead variants rs35705950 with expression of MUC5B and rs2076295 with expression of DSP in both cases and controls. mQTL analysis identified CpGs in gene bodies of MUC5B (cg17589883) and DSP (cg08964675) associated with the lead variants in these two loci. We also demonstrated strong co-localization of eQTL/mQTL and genetic signal in MUC5B (rs35705950) and DSP (rs2076295). Functional validation of the mQTL in MUC5B using luciferase reporter assays demonstrates that the CpG resides within a putative internal repressor element. Conclusions: We have established a relationship of the common IPF genetic risk variants rs35705950 and rs2076295 with respective changes in MUC5B and DSP expression and methylation. These results provide additional evidence that both MUC5B and DSP are involved in the etiology of IPF.
Collapse
Affiliation(s)
| | | | | | - Camille M. Moore
- Department of Biostatistics and Bioinformatics and
- Center for Genes, Environment, and Health
| | | | | | - Fabienne Gally
- Department of Medicine
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | | | | | | | | | - Mauricio Rojas
- Department of Internal Medicine, Ohio State College of Medicine, The Ohio State University, Columbus, Ohio
| | - Paul J. Wolters
- Department of Medicine, University of California, San Francisco, California
| | | | - Timothy S. Blackwell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tomoko Nakanishi
- Department of Human Genetics, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Canada
| | - J. Brent Richards
- Department of Human Genetics, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Canada
| | - Anthony N. Gerber
- Department of Medicine
- Department of Medicine, and
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Tasha E. Fingerlin
- Department of Biostatistics and Bioinformatics and
- Center for Genes, Environment, and Health
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Norman Sachs
- Cell Biology, Vertex Pharmaceuticals, San Diego, California; and
| | - Sara L. Pulit
- Computational Genomics, Vertex Pharmaceuticals, Boston, Massachusetts
| | - Zachary Zappala
- Computational Genomics, Vertex Pharmaceuticals, Boston, Massachusetts
| | - David A. Schwartz
- Department of Medicine
- Department of Microbiology and Immunology, University of Colorado Anschutz Medical Campus; Aurora, Colorado
| | - Ivana V. Yang
- Department of Medicine
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado
| |
Collapse
|
20
|
Laurita KR, Vasireddi SK, Mackall JA. Elucidating arrhythmogenic right ventricular cardiomyopathy with stem cells. Birth Defects Res 2022; 114:948-958. [PMID: 35396927 PMCID: PMC9790231 DOI: 10.1002/bdr2.2010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 12/31/2022]
Abstract
Human stems cells have sparked many novel strategies for treating heart disease and for elucidating their underlying mechanisms. For example, arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited heart muscle disorder that is associated with fatal arrhythmias often occurring in healthy young adults. Fibro-fatty infiltrate, a clinical hallmark, progresses with the disease and can develop across both ventricles. Pathogenic variants in genes have been identified, with most being responsible for encoding cardiac desmosome proteins that reside at myocyte boundaries that are critical for cell-to-cell coupling. Despite some understanding of the molecular signaling mechanisms associated with ARVC mutations, their relationship with arrhythmogenesis is complex and not well understood for a monogenetic disorder. This review article focuses on arrhythmia mechanisms in ARVC based on clinical and animal studies and their relationship with disease causing variants. We also discuss the ways in which stem cells can be leveraged to improve our understanding of the role cardiac myocytes, nonmyocytes, metabolic signals, and inflammatory mediators play in an early onset disease such as ARVC.
Collapse
Affiliation(s)
- Kenneth R. Laurita
- Heart and Vascular Research CenterMetroHealth Campus, Case Western Reserve UniversityClevelandOhioUSA
| | - Sunil K. Vasireddi
- Stanford Cardiovascular Institute, Department of MedicineStanford UniversityCaliforniaUSA
| | - Judith A. Mackall
- Harrington Heart and Vascular InstituteUniversity Hospitals Cleveland Medical Center, Case Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
21
|
Stevens TL, Manring HR, Wallace MJ, Argall A, Dew T, Papaioannou P, Antwi-Boasiako S, Xu X, Campbell SG, Akar FG, Borzok MA, Hund TJ, Mohler PJ, Koenig SN, El Refaey M. Humanized Dsp ACM Mouse Model Displays Stress-Induced Cardiac Electrical and Structural Phenotypes. Cells 2022; 11:3049. [PMID: 36231013 PMCID: PMC9562631 DOI: 10.3390/cells11193049] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited disorder characterized by fibro-fatty infiltration with an increased propensity for ventricular arrhythmias and sudden death. Genetic variants in desmosomal genes are associated with ACM. Incomplete penetrance is a common feature in ACM families, complicating the understanding of how external stressors contribute towards disease development. To analyze the dual role of genetics and external stressors on ACM progression, we developed one of the first mouse models of ACM that recapitulates a human variant by introducing the murine equivalent of the human R451G variant into endogenous desmoplakin (DspR451G/+). Mice homozygous for this variant displayed embryonic lethality. While DspR451G/+ mice were viable with reduced expression of DSP, no presentable arrhythmogenic or structural phenotypes were identified at baseline. However, increased afterload resulted in reduced cardiac performance, increased chamber dilation, and accelerated progression to heart failure. In addition, following catecholaminergic challenge, DspR451G/+ mice displayed frequent and prolonged arrhythmic events. Finally, aberrant localization of connexin-43 was noted in the DspR451G/+ mice at baseline, becoming more apparent following cardiac stress via pressure overload. In summary, cardiovascular stress is a key trigger for unmasking both electrical and structural phenotypes in one of the first humanized ACM mouse models.
Collapse
Affiliation(s)
- Tyler L. Stevens
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Physiology and Cellular Biology, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Heather R. Manring
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Michael J. Wallace
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Physiology and Cellular Biology, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Aaron Argall
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Physiology and Cellular Biology, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Trevor Dew
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Physiology and Cellular Biology, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Peter Papaioannou
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Steve Antwi-Boasiako
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Xianyao Xu
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Stuart G. Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Fadi G. Akar
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Maegen A. Borzok
- Biochemistry, Chemistry, Engineering, and Physics Department, Commonwealth University of Pennsylvania, Mansfield, PA 16933, USA
| | - Thomas J. Hund
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Peter J. Mohler
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Physiology and Cellular Biology, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Sara N. Koenig
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| | - Mona El Refaey
- Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
22
|
Towards a Better Understanding of Genotype-Phenotype Correlations and Therapeutic Targets for Cardiocutaneous Genes: The Importance of Functional Studies above Prediction. Int J Mol Sci 2022; 23:ijms231810765. [PMID: 36142674 PMCID: PMC9503274 DOI: 10.3390/ijms231810765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Genetic variants in gene-encoding proteins involved in cell−cell connecting structures, such as desmosomes and gap junctions, may cause a skin and/or cardiac phenotype, of which the combination is called cardiocutaneous syndrome. The cardiac phenotype is characterized by cardiomyopathy and/or arrhythmias, while the skin particularly displays phenotypes such as keratoderma, hair abnormalities and skin fragility. The reported variants associated with cardiocutaneous syndrome, in genes DSP, JUP, DSC2, KLHL24, GJA1, are classified by interpretation guidelines from the American College of Medical Genetics and Genomics. The genotype−phenotype correlation, however, remains poorly understood. By providing an overview of variants that are assessed for a functional protein pathology, we show that this number (n = 115) is low compared to the number of variants that are assessed by in silico algorithms (>5000). As expected, there is a mismatch between the prediction of variant pathogenicity and the prediction of the functional effect compared to the real functional evidence. Aiding to improve genotype−phenotype correlations, we separate variants into ‘protein reducing’ or ‘altered protein’ variants and provide general conclusions about the skin and heart phenotype involved. We conclude by stipulating that adequate prognoses can only be given, and targeted therapies can only be designed, upon full knowledge of the protein pathology through functional investigation.
Collapse
|
23
|
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
|
24
|
Tan M, Wang X, Liu H, Peng X, Yang Y, Yu H, Xu L, Li J, Cao H. Genetic Diagnostic Yield and Novel Causal Genes of Congenital Heart Disease. Front Genet 2022; 13:941364. [PMID: 35910219 PMCID: PMC9326225 DOI: 10.3389/fgene.2022.941364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Congenital heart disease (CHD) is the most common congenital malformation in fetuses and neonates, which also represents a leading cause of mortality. Although significant progress has been made by emerging advanced technologies in genetic etiology diagnosis, the causative genetic mechanisms behind CHD remain poorly understood and more than half of CHD patients lack a genetic diagnosis. Unlike carefully designed large case-control cohorts by multicenter trials, we designed a reliable strategy to analyze case-only cohorts to utilize clinical samples sufficiently. Combined low-coverage whole-genome sequencing (WGS) and whole-exome sequencing (WES) were simultaneously conducted in a patient-only cohort for identifying genetic etiologies and exploring candidate, or potential causative CHD-related genes. A total of 121 sporadic CHD patients were recruited and 34.71% (95% CI, 26.80 to 43.56) was diagnosed with genetic etiologies by low-coverage WGS and WES. Chromosomal abnormalities and damaging variants of CHD-related genes could explain 24.79% (95% CI, 17.92 to 33.22) and 18.18% (95% CI, 12.26 to 26.06) of CHD patients, separately, and 8.26% (95% CI, 4.39 to 14.70) of them have simultaneously detected two types of variants. Deletion of chromosome 22q11.2 and pathogenic variants of the COL3A1 gene were the most common recurrent variants of chromosomal abnormalities and gene variants, respectively. By in-depth manual interpretation, we identified eight candidate CHD-causing genes. Based on rare disease-causing variants prediction and interaction analysis with definitive CHD association genes, we proposed 86 genes as potential CHD-related genes. Gene Ontology (GO) enrichment analysis of the 86 genes revealed regulation-related processes were significantly enriched and processes response to regulation of muscle adaptation might be one of the underlying molecular mechanisms of CHD. Our findings and results provide new insights into research strategies and underlying mechanisms of CHD.
Collapse
Affiliation(s)
- Meihua Tan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Genomics Co., Ltd, Shenzhen, China
| | - Xinrui Wang
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Hongjie Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Peng
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - You Yang
- BGI Genomics Co., Ltd, Shenzhen, China
| | - Haifei Yu
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital, Fuzhou, China
- Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Affiliated Hospital of Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, China
- *Correspondence: Liangpu Xu, ; Jia Li, ; Hua Cao,
| | - Jia Li
- BGI Genomics Co., Ltd, Shenzhen, China
- Hebei Industrial Technology Research Institute of Genomics in Maternal and Child Health, Shijiazhuang BGI Genomics Co., Ltd, Shijiazhuang, China
- *Correspondence: Liangpu Xu, ; Jia Li, ; Hua Cao,
| | - Hua Cao
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Liangpu Xu, ; Jia Li, ; Hua Cao,
| |
Collapse
|
25
|
Blackwell DJ, Schmeckpeper J, Knollmann BC. Animal Models to Study Cardiac Arrhythmias. Circ Res 2022; 130:1926-1964. [PMID: 35679367 DOI: 10.1161/circresaha.122.320258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cardiac arrhythmias are a significant cause of morbidity and mortality worldwide, accounting for 10% to 15% of all deaths. Although most arrhythmias are due to acquired heart disease, inherited channelopathies and cardiomyopathies disproportionately affect children and young adults. Arrhythmogenesis is complex, involving anatomic structure, ion channels and regulatory proteins, and the interplay between cells in the conduction system, cardiomyocytes, fibroblasts, and the immune system. Animal models of arrhythmia are powerful tools for studying not only molecular and cellular mechanism of arrhythmogenesis but also more complex mechanisms at the whole heart level, and for testing therapeutic interventions. This review summarizes basic and clinical arrhythmia mechanisms followed by an in-depth review of published animal models of genetic and acquired arrhythmia disorders.
Collapse
Affiliation(s)
- Daniel J Blackwell
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Jeffrey Schmeckpeper
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
26
|
Ou S, Cesarato N, Mauran P, Gellé MP, Thiele H, Betz RC, Viguier M, Gusdorf L. A new de novo heterozygous missense mutation in the desmoplakin gene, causing Naxos and Carvajal disease, associating oligodontia and nail fragility. Clin Exp Dermatol 2022; 47:1424-1426. [PMID: 35574671 DOI: 10.1111/ced.15210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 11/28/2022]
Abstract
A new de novo heterozygous mutation in the desmoplakin gene, causing Naxos and Carvajal disease, has been reported in a 13-year-old Caucasian girl, with expanded clinical phenotype. In addition to woolly hair, palmoplantar keratoderma and cardiomyopathy, she had oligodontia and nail fragility. These additional clinical features may help in the diagnosis of Naxos and Carvajal disease, known to be severe on the cardiac level.
Collapse
Affiliation(s)
- Sokounthie Ou
- Services of Dermatology and Venereology, Centre Hospitalier Universitaire (CHU) de Reims, Reims, France
| | - Nicole Cesarato
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Pierre Mauran
- Services of Pediatric and Congenital Cardiology, Centre Hospitalier Universitaire (CHU) de Reims, Reims, France
| | - Marie-Paule Gellé
- Services of Pediatric Oncology, Centre Hospitalier Universitaire (CHU) de Reims, Reims, France
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Regina C Betz
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Manuelle Viguier
- Services of Dermatology and Venereology, Centre Hospitalier Universitaire (CHU) de Reims, Reims, France
| | - Laurence Gusdorf
- Services of Dermatology and Venereology, Centre Hospitalier Universitaire (CHU) de Reims, Reims, France
| |
Collapse
|
27
|
Vermeer MCSC, Andrei D, Kramer D, Nijenhuis AM, Hoedemaekers YM, Westers H, Jongbloed JDH, Pas HH, van den Berg MP, Silljé HHW, van der Meer P, Bolling MC. Functional investigation of two simultaneous or separately segregating DSP variants within a single family support the theory of a dose-dependent disease severity. Exp Dermatol 2022; 31:970-979. [PMID: 35325485 PMCID: PMC9322008 DOI: 10.1111/exd.14571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 02/28/2022] [Accepted: 03/22/2022] [Indexed: 11/30/2022]
Abstract
Desmoplakin (DP) is an important component of desmosomes, essential in cell–cell connecting structures in stress‐bearing tissues. Over the years, many hundreds of pathogenic variants in DSP have been associated with different cutaneous and cardiac phenotypes or a combination, known as a cardiocutaneous syndrome. Of less than 5% of the reported DSP variants, the effect on the protein has been investigated. Here, we describe and have performed RNA, protein and tissue analysis in a large family where DSPc.273+5G>A/c.6687delA segregated with palmoplantar keratoderma (PPK), woolly hair and lethal cardiomyopathy, while DSPWT/c.6687delA segregated with PPK and milder cardiomyopathy. hiPSC‐derived cardiomyocytes and primary keratinocytes from carriers were obtained for analysis. Unlike the previously reported nonsense variants in the last exon of DSP that bypassed the nonsense‐mediated mRNA surveillance system leading to protein truncation, variant c.6687delA was shown to cause the loss of protein expression. Patients carrying both variants and having a considerably more severe phenotype were shown to have 70% DP protein reduction, while patients carrying only c.6687delA had 50% protein reduction and a milder phenotype. The analysis of RNA from patient cells did not show any splicing effect of the c.273+5G>A variant. However, a minigene splicing assay clearly showed alternative spliced transcripts originating from this variant. This study shows the importance of RNA and protein analyses to pinpoint the exact effect of DSP variants instead of solely relying on predictions. In addition, the particular pattern of inheritance, with simultaneous or separately segregating DSP variants within the same family, strongly supports the theory of a dose‐dependent disease severity.
Collapse
Affiliation(s)
- Mathilde C S C Vermeer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Daniela Andrei
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Duco Kramer
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albertine M Nijenhuis
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yvonne M Hoedemaekers
- Department of Genetics, Radboud University Nijmegen, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Helga Westers
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan D H Jongbloed
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hendri H Pas
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maarten P van den Berg
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maria C Bolling
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
28
|
Cardiomyopathies. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
29
|
van der Voorn SM, Te Riele ASJM, Basso C, Calkins H, Remme CA, van Veen TAB. Arrhythmogenic cardiomyopathy: pathogenesis, pro-arrhythmic remodelling, and novel approaches for risk stratification and therapy. Cardiovasc Res 2021; 116:1571-1584. [PMID: 32246823 PMCID: PMC7526754 DOI: 10.1093/cvr/cvaa084] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a life-threatening cardiac disease caused by mutations in genes predominantly encoding for desmosomal proteins that lead to alterations in the molecular composition of the intercalated disc. ACM is characterized by progressive replacement of cardiomyocytes by fibrofatty tissue, ventricular dilatation, cardiac dysfunction, and heart failure but mostly dominated by the occurrence of life-threatening arrhythmias and sudden cardiac death (SCD). As SCD appears mostly in apparently healthy young individuals, there is a demand for better risk stratification of suspected ACM mutation carriers. Moreover, disease severity, progression, and outcome are highly variable in patients with ACM. In this review, we discuss the aetiology of ACM with a focus on pro-arrhythmic disease mechanisms in the early concealed phase of the disease. We summarize potential new biomarkers which might be useful for risk stratification and prediction of disease course. Finally, we explore novel therapeutic strategies to prevent arrhythmias and SCD in the early stages of ACM.
Collapse
Affiliation(s)
- Stephanie M van der Voorn
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, PO Box 85060, Utrecht 3508 AB, The Netherlands
| | - Anneline S J M Te Riele
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, PO Box 85060, Utrecht 3508 AB, The Netherlands
| | - Cristina Basso
- Cardiovascular Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua Medical School, Via A. Gabelli, 61 35121 Padova, Italy
| | - Hugh Calkins
- Johns Hopkins Hospital, Sheikh Zayed Tower 7125R, Baltimore, MD 21287, USA
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Toon A B van Veen
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, PO Box 85060, Utrecht 3508 AB, The Netherlands
| |
Collapse
|
30
|
Liu YQ, Zou HY, Xie JJ, Fang WK. Paradoxical Roles of Desmosomal Components in Head and Neck Cancer. Biomolecules 2021; 11:914. [PMID: 34203070 PMCID: PMC8234459 DOI: 10.3390/biom11060914] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 02/05/2023] Open
Abstract
Desmosomes are intercellular adhesion complexes involved in various aspects of epithelial pathophysiology, including tissue homeostasis, morphogenesis, and disease development. Recent studies have reported that the abnormal expression of various desmosomal components correlates with tumor progression and poor survival. In addition, desmosomes have been shown to act as a signaling platform to regulate the proliferation, invasion, migration, morphogenesis, and apoptosis of cancer cells. The occurrence and progression of head and neck cancer (HNC) is accompanied by abnormal expression of desmosomal components and loss of desmosome structure. However, the role of desmosomal components in the progression of HNC remains controversial. This review aims to provide an overview of recent developments showing the paradoxical roles of desmosomal components in tumor suppression and promotion. It offers valuable insights for HNC diagnosis and therapeutics development.
Collapse
Affiliation(s)
- Yin-Qiao Liu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China; (Y.-Q.L.); (H.-Y.Z.)
| | - Hai-Ying Zou
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China; (Y.-Q.L.); (H.-Y.Z.)
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China; (Y.-Q.L.); (H.-Y.Z.)
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, China
| | - Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China; (Y.-Q.L.); (H.-Y.Z.)
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
31
|
Mohammed F, Chidgey M. Desmosomal protein structure and function and the impact of disease-causing mutations. J Struct Biol 2021; 213:107749. [PMID: 34033898 DOI: 10.1016/j.jsb.2021.107749] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/26/2022]
Abstract
In this graphical review we focus on the structural characteristics of desmosomal proteins, their interactions with each other and with the intermediate filament cytoskeleton. The wealth of structural information that is now available allows predictions to be made about the pathogenic effect of disease-causing mutations. We have selected representative examples of missense mutations that are buried, semi-buried or surface exposed, and demonstrate how such variants could affect the structural fold of desmosomal proteins that are expressed in the heart. We explain how such alterations could compromise desmosomal adhesion, resulting in life threatening diseases including arrhythmogenic right ventricular cardiomyopathy.
Collapse
Affiliation(s)
- Fiyaz Mohammed
- Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK.
| | - Martyn Chidgey
- Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
32
|
Mattesi G, Cipriani A, Bauce B, Rigato I, Zorzi A, Corrado D. Arrhythmogenic Left Ventricular Cardiomyopathy: Genotype-Phenotype Correlations and New Diagnostic Criteria. J Clin Med 2021; 10:jcm10102212. [PMID: 34065276 PMCID: PMC8160676 DOI: 10.3390/jcm10102212] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disease characterized by loss of ventricular myocardium and fibrofatty replacement, which predisposes to scar-related ventricular arrhythmias and sudden cardiac death, particularly in the young and athletes. Although in its original description the disease was characterized by an exclusive or at least predominant right ventricle (RV) involvement, it has been demonstrated that the fibrofatty scar can also localize in the left ventricle (LV), with the LV lesion that can equalize or even overcome that of the RV. While the right-dominant form is typically associated with mutations in genes encoding for desmosomal proteins, other (non-desmosomal) mutations have been showed to cause the biventricular and left-dominant variants. This has led to a critical evaluation of the 2010 International Task Force criteria, which exclusively addressed the right phenotypic manifestations of ACM. An International Expert consensus document has been recently developed to provide upgraded criteria (“the Padua Criteria”) for the diagnosis of the whole spectrum of ACM phenotypes, particularly left-dominant forms, highlighting the use of cardiac magnetic resonance. This review aims to offer an overview of the current knowledge on the genetic basis, the phenotypic expressions, and the diagnosis of left-sided variants, both biventricular and left-dominant, of ACM.
Collapse
|
33
|
Prashanth G, Vastrad B, Tengli A, Vastrad C, Kotturshetti I. Identification of hub genes related to the progression of type 1 diabetes by computational analysis. BMC Endocr Disord 2021; 21:61. [PMID: 33827531 PMCID: PMC8028841 DOI: 10.1186/s12902-021-00709-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a serious threat to childhood life and has fairly complicated pathogenesis. Profound attempts have been made to enlighten the pathogenesis, but the molecular mechanisms of T1D are still not well known. METHODS To identify the candidate genes in the progression of T1D, expression profiling by high throughput sequencing dataset GSE123658 was downloaded from Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were identified, and gene ontology (GO) and pathway enrichment analyses were performed. The protein-protein interaction network (PPI), modules, target gene - miRNA regulatory network and target gene - TF regulatory network analysis were constructed and analyzed using HIPPIE, miRNet, NetworkAnalyst and Cytoscape. Finally, validation of hub genes was conducted by using ROC (Receiver operating characteristic) curve and RT-PCR analysis. A molecular docking study was performed. RESULTS A total of 284 DEGs were identified, consisting of 142 up regulated genes and 142 down regulated genes. The gene ontology (GO) and pathways of the DEGs include cell-cell signaling, vesicle fusion, plasma membrane, signaling receptor activity, lipid binding, signaling by GPCR and innate immune system. Four hub genes were identified and biological process analysis revealed that these genes were mainly enriched in cell-cell signaling, cytokine signaling in immune system, signaling by GPCR and innate immune system. ROC curve and RT-PCR analysis showed that EGFR, GRIN2B, GJA1, CAP2, MIF, POLR2A, PRKACA, GABARAP, TLN1 and PXN might be involved in the advancement of T1D. Molecular docking studies showed high docking score. CONCLUSIONS DEGs and hub genes identified in the present investigation help us understand the molecular mechanisms underlying the advancement of T1D, and provide candidate targets for diagnosis and treatment of T1D.
Collapse
Affiliation(s)
- G Prashanth
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, Karnataka, 577501, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka, 582103, India
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karanataka, 580001, India.
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society's Ayurvedic Medical College, Ron, Karanataka, 582209, India
| |
Collapse
|
34
|
Wanuske M, Brantschen D, Schinner C, Stüdle C, Walter E, Hiermaier M, Vielmuth F, Waschke J, Spindler V. Clustering of desmosomal cadherins by desmoplakin is essential for cell-cell adhesion. Acta Physiol (Oxf) 2021; 231:e13609. [PMID: 33354837 DOI: 10.1111/apha.13609] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/14/2022]
Abstract
AIM Desmoplakin (Dp) is a crucial component of the desmosome, a supramolecular cell junction complex anchoring intermediate filaments. The mechanisms how Dp modulates cell-cell adhesion are only partially understood. Here, we studied the impact of Dp on the function of desmosomal adhesion molecules, desmosome turnover and intercellular adhesion. METHODS CRISPR/Cas9 was used for gene editing of human keratinocytes which were characterized by Western blot and immunostaining. Desmosomal ultrastructure and function were assessed by electron microscopy and cell adhesion assays. Single molecule binding properties and localization of desmosomal cadherins were studied by atomic force microscopy and super-resolution imaging. RESULTS Knockout (ko) of Dp impaired cell cohesion to drastically higher extents as ko of another desmosomal protein, plakoglobin (Pg). In contrast to Pg ko, desmosomes were completely absent in Dp ko. Binding properties of the desmosomal adhesion molecules desmocollin (Dsc) 3 and desmoglein (Dsg) 3 remained unaltered under loss of Dp. Dp was required for assembling desmosomal cadherins into large clusters, as Dsg2 and Dsc3, adhesion molecules primarily localized within desmosomes, were redistributed into small puncta in the cell membrane of Dp ko cells. Additional silencing of desmosomal cadherins in Dp ko did not further increase loss of intercellular adhesion. CONCLUSION Our data demonstrate that Dp is essential for desmosome formation but does not influence intercellular adhesion on the level of individual cadherin binding properties. Rather, macro-clustering of desmosomal adhesion molecules through Dp is crucial. These results may help to better understand severe diseases which are caused by Dp dysfunction.
Collapse
Affiliation(s)
- Marie‐Therès Wanuske
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | | | - Camilla Schinner
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Chiara Stüdle
- Department of Biomedicine University of Basel Basel Switzerland
| | - Elias Walter
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Matthias Hiermaier
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Franziska Vielmuth
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Jens Waschke
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Volker Spindler
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| |
Collapse
|
35
|
Iop L. Toward the Effective Bioengineering of a Pathological Tissue for Cardiovascular Disease Modeling: Old Strategies and New Frontiers for Prevention, Diagnosis, and Therapy. Front Cardiovasc Med 2021; 7:591583. [PMID: 33748193 PMCID: PMC7969521 DOI: 10.3389/fcvm.2020.591583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases (CVDs) still represent the primary cause of mortality worldwide. Preclinical modeling by recapitulating human pathophysiology is fundamental to advance the comprehension of these diseases and propose effective strategies for their prevention, diagnosis, and treatment. In silico, in vivo, and in vitro models have been applied to dissect many cardiovascular pathologies. Computational and bioinformatic simulations allow developing algorithmic disease models considering all known variables and severity degrees of disease. In vivo studies based on small or large animals have a long tradition and largely contribute to the current treatment and management of CVDs. In vitro investigation with two-dimensional cell culture demonstrates its suitability to analyze the behavior of single, diseased cellular types. The introduction of induced pluripotent stem cell technology and the application of bioengineering principles raised the bar toward in vitro three-dimensional modeling by enabling the development of pathological tissue equivalents. This review article intends to describe the advantages and disadvantages of past and present modeling approaches applied to provide insights on some of the most relevant congenital and acquired CVDs, such as rhythm disturbances, bicuspid aortic valve, cardiac infections and autoimmunity, cardiovascular fibrosis, atherosclerosis, and calcific aortic valve stenosis.
Collapse
Affiliation(s)
- Laura Iop
- Department of Cardiac Thoracic Vascular Sciences, and Public Health, University of Padua Medical School, Padua, Italy
| |
Collapse
|
36
|
Kanoldt V, Kluger C, Barz C, Schweizer AL, Ramanujam D, Windgasse L, Engelhardt S, Chrostek-Grashoff A, Grashoff C. Metavinculin modulates force transduction in cell adhesion sites. Nat Commun 2020; 11:6403. [PMID: 33335089 PMCID: PMC7747745 DOI: 10.1038/s41467-020-20125-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 11/13/2020] [Indexed: 12/15/2022] Open
Abstract
Vinculin is a ubiquitously expressed protein, crucial for the regulation of force transduction in cells. Muscle cells express a vinculin splice-isoform called metavinculin, which has been associated with cardiomyopathies. However, the molecular function of metavinculin has remained unclear and its role for heart muscle disorders undefined. Here, we have employed a set of piconewton-sensitive tension sensors to probe metavinculin mechanics in cells. Our experiments reveal that metavinculin bears higher molecular forces but is less frequently engaged as compared to vinculin, leading to altered force propagation in cell adhesions. In addition, we have generated knockout mice to investigate the consequences of metavinculin loss in vivo. Unexpectedly, these animals display an unaltered tissue response in a cardiac hypertrophy model. Together, the data reveal that the transduction of cell adhesion forces is modulated by expression of metavinculin, yet its role for heart muscle function seems more subtle than previously thought.
Collapse
Affiliation(s)
- Verena Kanoldt
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, 48149, Münster, Germany
- Max Planck Institute of Biochemistry, Group of Molecular Mechanotransduction, 82152, Martinsried, Germany
| | - Carleen Kluger
- Max Planck Institute of Biochemistry, Group of Molecular Mechanotransduction, 82152, Martinsried, Germany
| | - Christiane Barz
- Max Planck Institute of Biochemistry, Group of Molecular Mechanotransduction, 82152, Martinsried, Germany
| | - Anna-Lena Schweizer
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, 48149, Münster, Germany
- Max Planck Institute of Biochemistry, Group of Molecular Mechanotransduction, 82152, Martinsried, Germany
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technical University of Munich, 80802, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802, Munich, Germany
| | - Lukas Windgasse
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, 48149, Münster, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich, 80802, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802, Munich, Germany
| | - Anna Chrostek-Grashoff
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, 48149, Münster, Germany
- Max Planck Institute of Biochemistry, Group of Molecular Mechanotransduction, 82152, Martinsried, Germany
| | - Carsten Grashoff
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, 48149, Münster, Germany.
- Max Planck Institute of Biochemistry, Group of Molecular Mechanotransduction, 82152, Martinsried, Germany.
| |
Collapse
|
37
|
Towbin JA, McKenna WJ, Abrams DJ, Ackerman MJ, Calkins H, Darrieux FCC, Daubert JP, de Chillou C, DePasquale EC, Desai MY, Estes NAM, Hua W, Indik JH, Ingles J, James CA, John RM, Judge DP, Keegan R, Krahn AD, Link MS, Marcus FI, McLeod CJ, Mestroni L, Priori SG, Saffitz JE, Sanatani S, Shimizu W, van Tintelen JP, Wilde AAM, Zareba W. 2019 HRS expert consensus statement on evaluation, risk stratification, and management of arrhythmogenic cardiomyopathy: Executive summary. Heart Rhythm 2020; 16:e373-e407. [PMID: 31676023 DOI: 10.1016/j.hrthm.2019.09.019] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Indexed: 01/14/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an arrhythmogenic disorder of the myocardium not secondary to ischemic, hypertensive, or valvular heart disease. ACM incorporates a broad spectrum of genetic, systemic, infectious, and inflammatory disorders. This designation includes, but is not limited to, arrhythmogenic right/left ventricular cardiomyopathy, cardiac amyloidosis, sarcoidosis, Chagas disease, and left ventricular noncompaction. The ACM phenotype overlaps with other cardiomyopathies, particularly dilated cardiomyopathy with arrhythmia presentation that may be associated with ventricular dilatation and/or impaired systolic function. This expert consensus statement provides the clinician with guidance on evaluation and management of ACM and includes clinically relevant information on genetics and disease mechanisms. PICO questions were utilized to evaluate contemporary evidence and provide clinical guidance related to exercise in arrhythmogenic right ventricular cardiomyopathy. Recommendations were developed and approved by an expert writing group, after a systematic literature search with evidence tables, and discussion of their own clinical experience, to present the current knowledge in the field. Each recommendation is presented using the Class of Recommendation and Level of Evidence system formulated by the American College of Cardiology and the American Heart Association and is accompanied by references and explanatory text to provide essential context. The ongoing recognition of the genetic basis of ACM provides the opportunity to examine the diverse triggers and potential common pathway for the development of disease and arrhythmia.
Collapse
Affiliation(s)
- Jeffrey A Towbin
- Le Bonheur Children's Hospital, Memphis, Tennessee; University of Tennessee Health Science Center, Memphis, Tennessee
| | - William J McKenna
- University College London, Institute of Cardiovascular Science, London, United Kingdom
| | | | | | | | | | | | | | | | | | - N A Mark Estes
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Wei Hua
- Fu Wai Hospital, Beijing, China
| | - Julia H Indik
- University of Arizona, Sarver Heart Center, Tucson, Arizona
| | - Jodie Ingles
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
| | | | - Roy M John
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel P Judge
- Medical University of South Carolina, Charleston, South Carolina
| | - Roberto Keegan
- Hospital Privado Del Sur, Buenos Aires, Argentina; Hospital Español, Bahia Blanca, Argentina
| | | | - Mark S Link
- UT Southwestern Medical Center, Dallas, Texas
| | - Frank I Marcus
- University of Arizona, Sarver Heart Center, Tucson, Arizona
| | | | - Luisa Mestroni
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Silvia G Priori
- University of Pavia, Pavia, Italy; European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart); ICS Maugeri, IRCCS, Pavia, Italy
| | | | | | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - J Peter van Tintelen
- University of Amsterdam, Academic Medical Center, Amsterdam, the Netherlands; Utrecht University Medical Center Utrecht, University of Utrecht, Department of Genetics, Utrecht, the Netherlands
| | - Arthur A M Wilde
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart); University of Amsterdam, Academic Medical Center, Amsterdam, the Netherlands; Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | | |
Collapse
|
38
|
Beffagna G, Sommariva E, Bellin M. Mechanotransduction and Adrenergic Stimulation in Arrhythmogenic Cardiomyopathy: An Overview of in vitro and in vivo Models. Front Physiol 2020; 11:568535. [PMID: 33281612 PMCID: PMC7689294 DOI: 10.3389/fphys.2020.568535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/19/2020] [Indexed: 01/09/2023] Open
Abstract
Arrhythmogenic Cardiomyopathy (AC) is a rare inherited heart disease, manifesting with progressive myocardium degeneration and dysfunction, and life-threatening arrhythmic events that lead to sudden cardiac death. Despite genetic determinants, most of AC patients admitted to hospital are athletes or very physically active people, implying the existence of other disease-causing factors. It is recognized that AC phenotypes are enhanced and triggered by strenuous physical activity, while excessive mechanical stretch and load, and repetitive adrenergic stimulation are mechanisms influencing disease penetrance. Different approaches have been undertaken to recapitulate and study both mechanotransduction and adrenergic signaling in AC, including the use of in vitro cellular and tissue models, and the development of in vivo models (particularly rodents but more recently also zebrafish). However, it remains challenging to reproduce mechanical load stimuli and physical activity in laboratory experimental settings. Thus, more work to drive the innovation of advanced AC models is needed to recapitulate these subtle physiological influences. Here, we review the state-of-the-art in this field both in clinical and laboratory-based modeling scenarios. Specific attention will be focused on highlighting gaps in the knowledge and how they may be resolved by utilizing novel research methodology.
Collapse
Affiliation(s)
- Giorgia Beffagna
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy.,Department of Biology, University of Padua, Padua, Italy
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Milena Bellin
- Department of Biology, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy.,Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
39
|
Ding Y, Yang J, Chen P, Lu T, Jiao K, Tester DJ, Giudicessi JR, Jiang K, Ackerman MJ, Li Y, Wang DW, Lee H, Wang DW, Xu X. Knockout of SORBS2 Protein Disrupts the Structural Integrity of Intercalated Disc and Manifests Features of Arrhythmogenic Cardiomyopathy. J Am Heart Assoc 2020; 9:e017055. [PMID: 32808564 PMCID: PMC7660791 DOI: 10.1161/jaha.119.017055] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022]
Abstract
Background Sorbs2b (sorbin and SH3 domain-containing 2b) was recently identified as a cardiomyopathy gene from a zebrafish mutagenesis screen. However, cardiac functions of its mammalian ortholog remain elusive. Methods and Results We conducted a detailed expression and subcellular localization analysis of Sorbs2 ortholog in mice and a phenotypic characterization in Sorbs2 knockout mice. Sorbs2 is highly expressed in the mouse heart and encodes an adhesion junction/desmosome protein that is mainly localized to the intercalated disc. A mutation with near complete depletion of the Sorbs2 protein in mice results in phenotypes characteristic of human arrhythmogenic cardiomyopathy (ACM), including right ventricular dilation, right ventricular dysfunction, spontaneous ventricular tachycardia, and premature death. Sorbs2 is required to maintain the structural integrity of intercalated disc. Its absence resulted in profound cardiac electrical remodeling with impaired impulse conduction and action potential derangements. Targeted sequencing of human patients with ACM identified 2 rare splicing variants classified as likely pathogenic were in 2 unrelated individuals with ACM from a cohort of 59 patients with ACM. Conclusions The Sorbs2 knockout mouse manifests several key features reminiscent of human ACM. Although the candidacy of SORBS2 as a new ACM-susceptibility gene is supported by preliminary human genetics study, future validation in larger cohorts with ACM is needed.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Jingchun Yang
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Peng Chen
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tong Lu
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Kunli Jiao
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | | | | | - Kai Jiang
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | - Michael J. Ackerman
- Department of Cardiovascular Medicine (Division of Heart Rhythm Services)Mayo ClinicRochesterMN
- Pediatric and Adolescent Medicine (Division of Pediatric Cardiology)Mayo ClinicRochesterMN
- Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory)Mayo ClinicRochesterMN
| | - Yigang Li
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Dao Wu Wang
- State Key Laboratory of Reproductive MedicineClinical Center of Reproductive Medicine and Department of CardiologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - HoN‐chi Lee
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Dao Wen Wang
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaolei Xu
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| |
Collapse
|
40
|
Song E, Wang R, Leopold JA, Loscalzo J. Network determinants of cardiovascular calcification and repositioned drug treatments. FASEB J 2020; 34:11087-11100. [PMID: 32638415 PMCID: PMC7497212 DOI: 10.1096/fj.202001062r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 01/31/2023]
Abstract
Ectopic cardiovascular calcification is a highly prevalent pathology for which there are no effective novel or repurposed pharmacotherapeutics to prevent disease progression. We created a human calcification endophenotype module (ie, the "calcificasome") by mapping vascular calcification genes (proteins) to the human vascular smooth muscle-specific protein-protein interactome (218 nodes and 632 edges, P < 10-5 ). Network proximity analysis was used to demonstrate that the calcificasome overlapped significantly with endophenotype modules governing inflammation, thrombosis, and fibrosis in the human interactome (P < 0.001). A network-based drug repurposing analysis further revealed that everolimus, temsirolimus, and pomalidomide are predicted to target the calcificasome. The efficacy of these agents in limiting calcification was confirmed experimentally by treating human coronary artery smooth muscle cells in an in vitro calcification assay. Each of the drugs affected expression or activity of their predicted target in the network, and decreased calcification significantly (P < 0.009). An integrated network analytical approach identified novel mediators of ectopic cardiovascular calcification and biologically plausible candidate drugs that could be repurposed to target calcification. This methodological framework for drug repurposing has broad applicability to other diseases.
Collapse
Affiliation(s)
- Euijun Song
- Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Rui‐Sheng Wang
- Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Joseph Loscalzo
- Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
41
|
Ali A, Al-Tobasei R, Lourenco D, Leeds T, Kenney B, Salem M. Genome-wide scan for common variants associated with intramuscular fat and moisture content in rainbow trout. BMC Genomics 2020; 21:529. [PMID: 32736521 PMCID: PMC7393730 DOI: 10.1186/s12864-020-06932-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/20/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Genetic improvement of fillet quality attributes is a priority of the aquaculture industry. Muscle composition impacts quality attributes such as flavor, appearance, texture, and juiciness. Fat and moisture make up about ~ 80% of the tissue weight. The genetic architecture underlying the fat and moisture content of the muscle is still to be fully explored in fish. A 50 K gene transcribed SNP chip was used for genotyping 789 fish with available phenotypic data for fat and moisture content. Genotyped fish were obtained from two consecutive generations produced in the National Center for Cool and Cold Water Aquaculture (NCCCWA) growth-selective breeding program. Estimates of SNP effects from weighted single-step GBLUP (WssGBLUP) were used to perform genome-wide association (GWA) analysis to identify quantitative trait loci (QTL) associated with the studied traits. RESULTS Using genomic sliding windows of 50 adjacent SNPs, 137 and 178 SNPs were identified as associated with fat and moisture content, respectively. Chromosomes 19 and 29 harbored the highest number of SNPs explaining at least 2% of the genetic variation in fat and moisture content. A total of 61 common SNPs on chromosomes 19 and 29 affected the aforementioned traits; this association suggests common mechanisms underlying intramuscular fat and moisture content. Additionally, based on single-marker GWA analyses, 8 and 24 SNPs were identified in association with fat and moisture content, respectively. CONCLUSION SNP-harboring genes were primarily involved in lipid metabolism, cytoskeleton remodeling, and protein turnover. This work provides putative SNP markers that could be prioritized and used for genomic selection in breeding programs.
Collapse
Affiliation(s)
- Ali Ali
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Rafet Al-Tobasei
- Computational Science Program, Middle Tennessee State University, Murfreesboro, TN, 37132, USA
| | - Daniela Lourenco
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA
| | - Tim Leeds
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, United States Department of Agriculture, Kearneysville, WV, USA
| | - Brett Kenney
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, 26506, USA
| | - Mohamed Salem
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
42
|
Mohammed F, Trieber C, Overduin M, Chidgey M. Molecular mechanism of intermediate filament recognition by plakin proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118801. [PMID: 32712070 DOI: 10.1016/j.bbamcr.2020.118801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/26/2022]
Abstract
The plakin family of cytolinkers interacts with intermediate filaments (IFs) through plakin repeat domain (PRD) and linker modules. Recent structure/function studies have established the molecular basis of envoplakin-PRD and periplakin-linker interactions with vimentin. Both plakin modules share a broad basic groove which recognizes acidic rod elements on IFs, a mechanism that is applicable to other plakin family members. This review postulates a universal IF engagement mechanism that illuminates the specific effects of pathogenic mutations associated with diseases including arrhythmogenic right ventricular cardiomyopathy, and reveals how diverse plakin proteins offer tailored IF tethering to ensure stable, dynamic and regulated cellular structures.
Collapse
Affiliation(s)
- Fiyaz Mohammed
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK.
| | - Catharine Trieber
- Department of Biochemistry, Faculty of Medicine and Dentistry, 474 Medical Sciences Building, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Michael Overduin
- Department of Biochemistry, Faculty of Medicine and Dentistry, 474 Medical Sciences Building, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Martyn Chidgey
- Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
43
|
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
|
44
|
Arrhythmogenic Cardiomyopathy: Molecular Insights for Improved Therapeutic Design. J Cardiovasc Dev Dis 2020; 7:jcdd7020021. [PMID: 32466575 PMCID: PMC7345706 DOI: 10.3390/jcdd7020021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited disorder characterized by structural and electrical cardiac abnormalities, including myocardial fibro-fatty replacement. Its pathological ventricular substrate predisposes subjects to an increased risk of sudden cardiac death (SCD). ACM is a notorious cause of SCD in young athletes, and exercise has been documented to accelerate its progression. Although the genetic culprits are not exclusively limited to the intercalated disc, the majority of ACM-linked variants reside within desmosomal genes and are transmitted via Mendelian inheritance patterns; however, penetrance is highly variable. Its natural history features an initial “concealed phase” that results in patients being vulnerable to malignant arrhythmias prior to the onset of structural changes. Lack of effective therapies that target its pathophysiology renders management of patients challenging due to its progressive nature, and has highlighted a critical need to improve our understanding of its underlying mechanistic basis. In vitro and in vivo studies have begun to unravel the molecular consequences associated with disease causing variants, including altered Wnt/β-catenin signaling. Characterization of ACM mouse models has facilitated the evaluation of new therapeutic approaches. Improved molecular insight into the condition promises to usher in novel forms of therapy that will lead to improved care at the clinical bedside.
Collapse
|
45
|
van Wijngaarden AL, Hiemstra YL, Koopmann TT, Ruivenkamp CAL, Aten E, Schalij MJ, Bax JJ, Delgado V, Barge-Schaapveld DQCM, Ajmone Marsan N. Identification of known and unknown genes associated with mitral valve prolapse using an exome slice methodology. J Med Genet 2020; 57:843-850. [PMID: 32277046 DOI: 10.1136/jmedgenet-2019-106715] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/18/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Although a familial distribution has been documented, the genetic aetiology of mitral valve prolapse (MVP) is largely unknown, with only four genes identified so far: FLNA, DCHS1, DZIP1 and PLD1. The aim of this study was to evaluate the genetic yield in known causative genes and to identify possible novel genes associated with MVP using a heart gene panel based on exome sequencing. METHODS Patients with MVP were referred for genetic counselling when a positive family history for MVP was reported and/or Barlow's disease was diagnosed. In total, 101 probands were included to identify potentially pathogenic variants in a set of 522 genes associated with cardiac development and/or diseases. RESULTS 97 (96%) probands were classified as Barlow's disease and 4 (4%) as fibroelastic deficiency. Only one patient (1%) had a likely pathogenic variant in the known causative genes (DCHS1). However, an interesting finding was that 10 probands (11%) had a variant that was classified as likely pathogenic in six different, mostly cardiomyopathy genes: DSP (1×), HCN4 (1×), MYH6 (1×), TMEM67 (1×), TRPS1 (1×) and TTN (5×). CONCLUSION Exome slice sequencing analysis performed in MVP probands reveals a low genetic yield in known causative genes but may expand the cardiac phenotype of other genes. This study suggests for the first time that also genes related to cardiomyopathy may be associated with MVP. This highlights the importance to screen these patients and their family for the presence of arrhythmias and of 'disproportionate' LV remodelling as compared with the severity of mitral regurgitation, unravelling a possible coexistent cardiomyopathy.
Collapse
Affiliation(s)
| | - Yasmine L Hiemstra
- Cardiology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Tamara T Koopmann
- Clinical Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Claudia A L Ruivenkamp
- Clinical Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Emmelien Aten
- Clinical Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Martin J Schalij
- Cardiology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Jeroen J Bax
- Cardiology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Victoria Delgado
- Cardiology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | | | - Nina Ajmone Marsan
- Cardiology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
46
|
Abstract
Arrhythmogenic cardiomyopathy is a genetic disorder characterized by the risk of life-threatening arrhythmias, myocardial dysfunction and fibrofatty replacement of myocardial tissue. Mutations in genes that encode components of desmosomes, the adhesive junctions that connect cardiomyocytes, are the predominant cause of arrhythmogenic cardiomyopathy and can be identified in about half of patients with the condition. However, the molecular mechanisms leading to myocardial destruction, remodelling and arrhythmic predisposition remain poorly understood. Through the development of animal, induced pluripotent stem cell and other models of disease, advances in our understanding of the pathogenic mechanisms of arrhythmogenic cardiomyopathy over the past decade have brought several signalling pathways into focus. These pathways include canonical and non-canonical WNT signalling, the Hippo-Yes-associated protein (YAP) pathway and transforming growth factor-β signalling. These studies have begun to identify potential therapeutic targets whose modulation has shown promise in preclinical models. In this Review, we summarize and discuss the reported molecular mechanisms underlying the pathogenesis of arrhythmogenic cardiomyopathy.
Collapse
|
47
|
Green KJ, Jaiganesh A, Broussard JA. Desmosomes: Essential contributors to an integrated intercellular junction network. F1000Res 2019; 8. [PMID: 31942240 PMCID: PMC6944264 DOI: 10.12688/f1000research.20942.1] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
The development of adhesive connections between cells was critical for the evolution of multicellularity and for organizing cells into complex organs with discrete compartments. Four types of intercellular junction are present in vertebrates: desmosomes, adherens junctions, tight junctions, and gap junctions. All are essential for the development of the embryonic layers and organs as well as adult tissue homeostasis. While each junction type is defined as a distinct entity, it is now clear that they cooperate physically and functionally to create a robust and functionally diverse system. During evolution, desmosomes first appeared in vertebrates as highly specialized regions at the plasma membrane that couple the intermediate filament cytoskeleton at points of strong cell–cell adhesion. Here, we review how desmosomes conferred new mechanical and signaling properties to vertebrate cells and tissues through their interactions with the existing junctional and cytoskeletal network.
Collapse
Affiliation(s)
- Kathleen J Green
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Avinash Jaiganesh
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joshua A Broussard
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
48
|
Abstract
The ECM (extracellular matrix) network plays a crucial role in cardiac homeostasis, not only by providing structural support, but also by facilitating force transmission, and by transducing key signals to cardiomyocytes, vascular cells, and interstitial cells. Changes in the profile and biochemistry of the ECM may be critically implicated in the pathogenesis of both heart failure with reduced ejection fraction and heart failure with preserved ejection fraction. The patterns of molecular and biochemical ECM alterations in failing hearts are dependent on the type of underlying injury. Pressure overload triggers early activation of a matrix-synthetic program in cardiac fibroblasts, inducing myofibroblast conversion, and stimulating synthesis of both structural and matricellular ECM proteins. Expansion of the cardiac ECM may increase myocardial stiffness promoting diastolic dysfunction. Cardiomyocytes, vascular cells and immune cells, activated through mechanosensitive pathways or neurohumoral mediators may play a critical role in fibroblast activation through secretion of cytokines and growth factors. Sustained pressure overload leads to dilative remodeling and systolic dysfunction that may be mediated by changes in the interstitial protease/antiprotease balance. On the other hand, ischemic injury causes dynamic changes in the cardiac ECM that contribute to regulation of inflammation and repair and may mediate adverse cardiac remodeling. In other pathophysiologic conditions, such as volume overload, diabetes mellitus, and obesity, the cell biological effectors mediating ECM remodeling are poorly understood and the molecular links between the primary insult and the changes in the matrix environment are unknown. This review article discusses the role of ECM macromolecules in heart failure, focusing on both structural ECM proteins (such as fibrillar and nonfibrillar collagens), and specialized injury-associated matrix macromolecules (such as fibronectin and matricellular proteins). Understanding the role of the ECM in heart failure may identify therapeutic targets to reduce geometric remodeling, to attenuate cardiomyocyte dysfunction, and even to promote myocardial regeneration.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
49
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
50
|
Vadgama N, Lamont D, Hardy J, Nasir J, Lovering RC. Distinct proteomic profiles in monozygotic twins discordant for ischaemic stroke. Mol Cell Biochem 2019; 456:157-165. [PMID: 30694515 DOI: 10.1007/s11010-019-03501-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/19/2019] [Indexed: 01/13/2023]
Abstract
Stroke is a common disorder with significant morbidity and mortality, and complex aetiology involving both environmental and genetic risk factors. Although some of the major risk factors for stoke, such as smoking and hypertension, are well-documented, the underlying genetic and detailed molecular mechanisms remain elusive. Exploring the relevant biochemical pathways may contribute to the clinical diagnosis of stroke and shed light on its aetiology. A comparative proteomic analysis of blood serum of a pair of monozygotic (MZ) twins discordant for ischaemic stroke (IS) was performed using a label-free quantitative proteomics approach. To overcome the limit of reproducibility in the serum preparation, two separate runs were performed, each consisting of three technical replicates per sample. Biological processes associated with proteins differentially expressed between the twins were explored with gene ontology (GO) classification using the functional analysis tool g:Profiler. ANOVA test performed in Progenesis LC-MS identified 179 (run 1) and 209 (run 2) proteins as differentially expressed between the affected and unaffected twin (p < 0.05). Furthermore, the level of serum fibulin 1, an extracellular matrix protein associated with arterial stiffness, was on average 13.37-fold higher in the affected twin. Each dataset was then analysed independently, and the proteins were classified according to GO terms. The categories overrepresented in the affected twin predominantly corresponded to stroke-relevant processes, including wound healing, blood coagulation and haemostasis, with a high proportion of the proteins overexpressed in the affected twin associated with these terms. By contrast, in the unaffected twin diagnosed with atopic dermatitis, there were increased levels of keratin proteins and GO terms associated with skin development. The identification of cellular pathways enriched in IS as well as the upregulation of fibulin 1 sheds new light on the underlying disease-causing mechanisms at the molecular level. Our findings of distinct proteomic signatures associated with IS and atopic dermatitis suggest proteomic profiling could be used as a general approach for improved diagnostic, prognostic and therapeutic strategies.
Collapse
Affiliation(s)
- Nirmal Vadgama
- Institute of Neurology, University College London, London, UK
- Cell Biology and Genetics Research Centre, St. George's University of London, London, UK
| | - Douglas Lamont
- College of Life Sciences, University of Dundee, Dundee, UK
| | - John Hardy
- Institute of Neurology, University College London, London, UK
| | - Jamal Nasir
- Cell Biology and Genetics Research Centre, St. George's University of London, London, UK.
- Molecular Biosciences Research Group, Faculty of Health & Society, University of Northampton, Northampton, UK.
| | - Ruth C Lovering
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK
| |
Collapse
|