1
|
Gholipour A, Zahedmehr A, Arabian M, Shakerian F, Maleki M, Oveisee M, Malakootian M. MiR-6721-5p as a natural regulator of Meta-VCL is upregulated in the serum of patients with coronary artery disease. Noncoding RNA Res 2025; 10:25-34. [PMID: 39296643 PMCID: PMC11406674 DOI: 10.1016/j.ncrna.2024.08.006] [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: 01/06/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/21/2024] Open
Abstract
Background Coronary artery disease (CAD), the leading cause of mortality globally, arises from atherosclerotic blockage of the coronary arteries. Meta-vinculin (meta-VCL), a large spliced isoform of VCL, co-localizes in muscular adhesive structures and plays significant roles in cardiac physiology and pathophysiology. This study aimed to identify microRNAs (miRNAs) regulating meta-VCL expression and investigate the expression alterations of the miRNAs of interest and meta-VCL as potential biomarkers in the serum of CAD patients. Methods Bioinformatics tools were employed to select miRNAs targeting meta-VCL. Cell-based ectopic expression analysis and a dual-luciferase assay were used to examine the interactions between miRNAs and meta-VCL. An ELISA assessed the concentrations of interleukin-6 (IL-6), IL-10, and tumor necrosis factor-α (TNF-α). MiRNA and meta-VCL expression patterns and biomarker suitability were evaluated in serum samples from CAD and non-CAD individuals using real-time PCR. A cardiac cell-line data set and CAD blood exosome samples were analyzed using bioinformatics and ROC curve analyses, respectively. Results miR-6721-5p directly interacted with the putative target sites at the 3'-UTR of meta-VCL and regulated its expression. IL-10 and TNF-α concentrations, which may act as anti-inflammatory factors, decreased following miR-6721-5p upregulation and meta-VCL downregulation. Bioinformatics and experimental expression analyses confirmed downregulated meta-VCL expression and upregulated miR-6721-5p expression in CAD samples. ROC curve analysis yielded an AUC score of 0.705 (P = 0.018), indicating the potential suitability of miR-6721-5p as a biomarker for CAD. Conclusions miR-6721-5p plays a regulatory role in meta-VCL expression and may contribute to CAD development by reducing anti-inflammatory factors. These findings suggest that miR-6721-5p could serve as a novel biomarker in the pathogenesis of CAD.
Collapse
Affiliation(s)
- Akram Gholipour
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zahedmehr
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maedeh Arabian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Farshad Shakerian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mahshid Malakootian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
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
|
3
|
Andrysiak K, Ferdek PE, Sanetra AM, Machaj G, Schmidt L, Kraszewska I, Sarad K, Palus-Chramiec K, Lis O, Targosz-Korecka M, Krüger M, Lewandowski MH, Ylla G, Stępniewski J, Dulak J. Upregulation of utrophin improves the phenotype of Duchenne muscular dystrophy hiPSC-derived CMs. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102247. [PMID: 39035791 PMCID: PMC11259739 DOI: 10.1016/j.omtn.2024.102247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 06/07/2024] [Indexed: 07/23/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a genetic neuromuscular disease. Although it leads to muscle weakness, affected individuals predominantly die from cardiomyopathy, which remains uncurable. Accumulating evidence suggests that an overexpression of utrophin may counteract some of the pathophysiological outcomes of DMD. The aim of this study was to investigate the role of utrophin in dystrophin-deficient human cardiomyocytes (CMs) and to test whether an overexpression of utrophin, implemented via the CRISPR-deadCas9-VP64 system, can improve their phenotype. We used human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) lacking either dystrophin (DMD) or both dystrophin and utrophin (DMD KO/UTRN(+/-)). We carried out proteome analysis, which revealed considerable differences in the proteins related to muscle contraction, cell-cell adhesion, and extracellular matrix organization. Furthermore, we evaluated the role of utrophin in maintaining the physiological properties of DMD hiPSC-CMs using atomic force microscopy, patch-clamp, and Ca2+ oscillation analysis. Our results showed higher values of afterhyperpolarization and altered patterns of cytosolic Ca2+ oscillations in DMD; the latter was further disturbed in DMD KO/UTRN(+/-) hiPSC-CMs. Utrophin upregulation improved both parameters. Our findings demonstrate for the first time that utrophin maintains the physiological functions of DMD hiPSC-CMs, and that its upregulation can compensate for the loss of dystrophin.
Collapse
Affiliation(s)
- Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Paweł E. Ferdek
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Anna M. Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland
| | - Gabriela Machaj
- Laboratory of Bioinformatics and Genome Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Luisa Schmidt
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - Izabela Kraszewska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Katarzyna Sarad
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Kraków, Poland
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland
| | - Olga Lis
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
| | - Marta Targosz-Korecka
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - Marian H. Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland
| | - Guillem Ylla
- Laboratory of Bioinformatics and Genome Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| |
Collapse
|
4
|
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
|
5
|
Eden M, Leye M, Hahn J, Heilein E, Luzarowski M, Völschow B, Tannert C, Sossalla S, Lucena-Porcel C, Frank D, Frey N. Mst4, a novel cardiac STRIPAK complex-associated kinase, regulates cardiomyocyte growth and survival and is upregulated in human cardiomyopathy. J Biol Chem 2024; 300:107255. [PMID: 38579991 PMCID: PMC11087964 DOI: 10.1016/j.jbc.2024.107255] [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: 07/05/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
Myocardial failure is associated with adverse remodeling, including loss of cardiomyocytes, hypertrophy, and alterations in cell-cell contacts. Striatin-interacting phosphatase and kinase (STRIPAK) complexes and their mammalian STE20-like kinase 4 (Mst4) have been linked to development of different diseases. The role and targets of Mst4 in cardiomyocytes have not been investigated yet. Multitissue immunoblot experiments show highly enriched Mst4 expression in rodent hearts. Analyses of human biopsy samples from patients suffering from dilated cardiomyopathy revealed that Mst4 is upregulated (5- to 8-fold p < 0.001) compared with nonfailing controls. Increased abundance of Mst4 could also be detected in mouse models of cardiomyopathy. We confirmed that Mst4 interacts with STRIPAK components in neonatal rat ventricular cardiomyocytes, indicating that STRIPAK is present in the heart. Immunofluorescence stainings and molecular interaction studies revealed that Mst4 is localized to the intercalated disc and interacts with several intercalated disc proteins. Overexpression of Mst4 in cardiomyocytes results in hypertrophy compared with controls. In adult rat cardiomyocytes, Mst4 overexpression increases cellular and sarcomeric fractional shortening (p < 0.05), indicating enhanced contractility. Overexpression of Mst4 also inhibits apoptosis shown by reduction of cleaved caspase3 (-69%, p < 0.0001), caspase7 (-80%, p < 0.0001), and cleaved Parp1 (-27%, p < 0.001). To elucidate potential Mst4 targets, we performed phosphoproteomics analyses in neonatal rat cardiomyocytes after Mst4 overexpression and inhibition. The results revealed target candidates of Mst4 at the intercalated disc. We identified Mst4 as a novel cardiac kinase that is upregulated in cardiomyopathy-regulating cardiomyocyte growth and survival.
Collapse
Affiliation(s)
- Matthias Eden
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Marius Leye
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Justus Hahn
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Emanuel Heilein
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Marcin Luzarowski
- Core Facility for Mass Spectrometry and Proteomics, Center for Molecular Biology at Heidelberg University (ZMBH), Heidelberg, Germany
| | - Bill Völschow
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Christin Tannert
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Internal Medicine III (Cardiology and Angiology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Samuel Sossalla
- Department of Cardiology, University of Giessen, Giessen and Kerckhoff Heart and Lung Centre, Giessen, Germany
| | - Carlota Lucena-Porcel
- Tissue Bank of the National Center of Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Derk Frank
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Internal Medicine III (Cardiology and Angiology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany.
| |
Collapse
|
6
|
Federspiel JM, Reil JC, Xu A, Scholtz S, Batzner A, Maack C, Sequeira V. Retrofitting the Heart: Explaining the Enigmatic Septal Thickening in Hypertrophic Cardiomyopathy. Circ Heart Fail 2024; 17:e011435. [PMID: 38695186 DOI: 10.1161/circheartfailure.123.011435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/26/2024] [Indexed: 05/23/2024]
Abstract
Hypertrophic cardiomyopathy is the most common genetic cardiac disease and is characterized by left ventricular hypertrophy. Although this hypertrophy often associates with sarcomeric gene mutations, nongenetic factors also contribute to the disease, leading to diastolic dysfunction. Notably, this dysfunction manifests before hypertrophy and is linked to hypercontractility, as well as nonuniform contraction and relaxation (myofibril asynchrony) of the myocardium. Although the distribution of hypertrophy in hypertrophic cardiomyopathy can vary both between and within individuals, in most cases, it is primarily confined to the interventricular septum. The reasons for septal thickening remain largely unknown. In this article, we propose that alterations in muscle fiber geometry, present from birth, dictate the septal shape. When combined with hypercontractility and exacerbated by left ventricular outflow tract obstruction, these factors predispose the septum to an isometric type of contraction during systole, consequently constraining its mobility. This contraction, or more accurately, this focal increase in biomechanical stress, prompts the septum to adapt and undergo remodeling. Drawing a parallel, this is reminiscent of how earthquake-resistant buildings are retrofitted with vibration dampers to absorb the majority of the shock motion and load. Similarly, the heart adapts by synthesizing viscoelastic elements such as microtubules, titin, desmin, collagen, and intercalated disc components. This pronounced remodeling in the cytoskeletal structure leads to noticeable septal hypertrophy. This structural adaptation acts as a protective measure against damage by attenuating myofibril shortening while reducing cavity tension according to Laplace Law. By examining these events, we provide a coherent explanation for the septum's predisposition toward hypertrophy.
Collapse
Affiliation(s)
- Jan M Federspiel
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
- Saarland University, Faculty of Medicine, Institute for Legal Medicine, Homburg (Saar), Germany (J.M.F.)
| | - Jan-Christian Reil
- Klinik für allgemeine und interventionelle Kardiologie, Herz- und Diabetes-Zentrum Nordrhein-Westphalen, Germany (J.-C.R., S.S.)
| | - Anton Xu
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
| | - Smita Scholtz
- Klinik für allgemeine und interventionelle Kardiologie, Herz- und Diabetes-Zentrum Nordrhein-Westphalen, Germany (J.-C.R., S.S.)
| | - Angelika Batzner
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
- Department of Internal Medicine I, University Hospital Würzburg, Germany (A.B.)
| | - Christoph Maack
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
| | - Vasco Sequeira
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
| |
Collapse
|
7
|
Osten F, Weber N, Wendland M, Holler T, Piep B, Kröhn S, Teske J, Bodenschatz AK, Devadas SB, Menge KS, Chatterjee S, Schwanke K, Kosanke M, Montag J, Thum T, Zweigerdt R, Kraft T, Iorga B, Meissner JD. Myosin expression and contractile function are altered by replating stem cell-derived cardiomyocytes. J Gen Physiol 2023; 155:e202313377. [PMID: 37656049 PMCID: PMC10473967 DOI: 10.1085/jgp.202313377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/16/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
Myosin heavy chain (MyHC) is the main determinant of contractile function. Human ventricular cardiomyocytes (CMs) predominantly express the β-isoform. We previously demonstrated that ∼80% of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) express exclusively β-MyHC after long-term culture on laminin-coated glass coverslips. Here, we investigated the impact of enzymatically detaching hESC-CMs after long-term culture and subsequently replating them for characterization of cellular function. We observed that force-related kinetic parameters, as measured in a micromechanical setup, resembled α- rather than β-MyHC-expressing myofibrils, as well as changes in calcium transients. Single-cell immunofluorescence analysis revealed that replating hESC-CMs led to rapid upregulation of α-MyHC, as indicated by increases in exclusively α-MyHC- and in mixed α/β-MyHC-expressing hESC-CMs. A comparable increase in heterogeneity of MyHC isoform expression was also found among individual human induced pluripotent stem cell (hiPSC)-derived CMs after replating. Changes in MyHC isoform expression and cardiomyocyte function induced by replating were reversible in the course of the second week after replating. Gene enrichment analysis based on RNA-sequencing data revealed changes in the expression profile of mechanosensation/-transduction-related genes and pathways, especially integrin-associated signaling. Accordingly, the integrin downstream mediator focal adhesion kinase (FAK) promoted β-MyHC expression on a stiff matrix, further validating gene enrichment analysis. To conclude, detachment and replating induced substantial changes in gene expression, MyHC isoform composition, and function of long-term cultivated human stem cell-derived CMs, thus inducing alterations in mechanosensation/-transduction, that need to be considered, particularly for downstream in vitro assays.
Collapse
Affiliation(s)
- Felix Osten
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Natalie Weber
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Meike Wendland
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Tim Holler
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Birgit Piep
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Simon Kröhn
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Jana Teske
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Alea K. Bodenschatz
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Santoshi Biswanath Devadas
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Kaja S. Menge
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Shambhabi Chatterjee
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Kristin Schwanke
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Maike Kosanke
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Judith Montag
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- REBIRTH Center for Translational Regenerative Therapies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Robert Zweigerdt
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Bogdan Iorga
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Bucharest, Romania
| | - Joachim D. Meissner
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Liu J, Cheng B, Fan X, Zhou X, Wang J, Zhou W, Li H, Zeng W, Yang P, Chen X. Click-iG: Simultaneous Enrichment and Profiling of Intact N-linked, O-GalNAc, and O-GlcNAcylated Glycopeptides. Angew Chem Int Ed Engl 2023; 62:e202303410. [PMID: 37431278 DOI: 10.1002/anie.202303410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/12/2023]
Abstract
Proteins are ubiquitously modified with glycans of varied chemical structures through distinct glycosidic linkages, making the landscape of protein glycosylation challenging to map. Profiling of intact glycopeptides with mass spectrometry (MS) has recently emerged as a powerful tool for revealing matched information of the glycosylation sites and attached glycans (i.e., intact glycosites), but is largely limited to individual glycosylation types. Herein, we describe Click-iG, which integrates metabolic labeling of glycans with clickable unnatural sugars, an optimized MS method, and a tailored version of pGlyco3 software to enable simultaneous enrichment and profiling of three types of intact glycopeptides: N-linked, mucin-type O-linked, and O-GlcNAcylated glycopeptides. We demonstrate the utility of Click-iG by the identification of thousands of intact glycosites in cell lines and living mice. From the mouse lung, heart, and spleen, a total of 2053 intact N-glycosites, 262 intact O-GalNAc glycosites, and 1947 O-GlcNAcylation sites were identified. Click-iG-enabled comprehensive coverage of the protein glycosylation landscape lays the foundation for interrogating crosstalk between different glycosylation pathways.
Collapse
Affiliation(s)
- Jialin Liu
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
- Institute of Biomedical Sciences and Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Bo Cheng
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Xinqi Fan
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Xinyue Zhou
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Jiankun Wang
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Wen Zhou
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Hengyu Li
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Wenfeng Zeng
- Key Lab of Intelligent Information Processing of Chinese Academy of Sciences (CAS) and Institute of Computing Technology, CAS, Beijing, 100190, China
| | - Pengyuan Yang
- Institute of Biomedical Sciences and Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| |
Collapse
|
9
|
Karatsai O, Lehka L, Wojton D, Grabowska AI, Duda MK, Lenartowski R, Redowicz MJ. Unconventional myosin VI in the heart: Involvement in cardiac dysfunction progressing with age. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166748. [PMID: 37169038 DOI: 10.1016/j.bbadis.2023.166748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
Hypertrophic cardiomyopathy is the most common cardiovascular disease, which is characterized by structural and functional myocardial abnormalities. It is caused predominantly by autosomal dominant mutations, mainly in genes encoding cardiac sarcomeric proteins, resulting in diverse phenotypical patterns and a heterogenic clinical course. Unconventional myosin VI (MVI) is one of the proteins important for heart function, as it was shown that a point mutation within MYO6 is associated with left ventricular hypertrophy. Previously, we showed that MVI is expressed in the cardiac muscle, where it localizes to the sarcoplasmic reticulum and intercalated discs. Here, we addressed the mechanisms of its involvement in cardiac dysfunction in Snell's waltzer mice (natural MVI knockouts) during heart development. We showed that heart enlargement was already seen in the E14.5 embryos and newborn animals (P0), and was maintained throughout the examined lifespan (up to 12 months). The higher levels of MVI were observed in the hearts of E14.5 embryos and P0 of control heterozygous mice. A search for the mechanisms behind the observed phenotype revealed several changes, accumulation of which resulted in age-progressing heart dysfunction. The main changes that mostly contribute to this functional impairment are the increase in cardiomyocyte proliferation in newborns, disorganization of intercalated discs, and overexpression of SERCA2 in hearts isolated from 12-month-old mice, indicative of functional alterations of sarcoplasmic reticulum. Also, possible aberrations in the heart vascularization, observed in 12-month-old animals could be additional factors responsible for MVI-associated heart dysfunction.
Collapse
Affiliation(s)
- Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Lilya Lehka
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Dominika Wojton
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Anna Izabela Grabowska
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Monika Katarzyna Duda
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka St., 01-813 Warsaw, Poland.
| | - Robert Lenartowski
- Faculty of Biological and Veterinary Sciences, The Nicolaus Copernicus University in Torun, 1 Lwowska St., 87-100 Torun, Poland.
| | - Maria Jolanta Redowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| |
Collapse
|
10
|
Wu T, Xu Y, Zhang L, Liang Z, Zhou X, Evans SM, Chen J. Filamin C is Essential for mammalian myocardial integrity. PLoS Genet 2023; 19:e1010630. [PMID: 36706168 PMCID: PMC9907827 DOI: 10.1371/journal.pgen.1010630] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/08/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
FLNC, encoding filamin C, is one of the most mutated genes in dilated and hypertrophic cardiomyopathy. However, the precise role of filamin C in mammalian heart remains unclear. In this study, we demonstrated Flnc global (FlncgKO) and cardiomyocyte-specific knockout (FlnccKO) mice died in utero from severely ruptured ventricular myocardium, indicating filamin C is required to maintain the structural integrity of myocardium in the mammalian heart. Contrary to the common belief that filamin C acts as an integrin inactivator, we observed attenuated activation of β1 integrin specifically in the myocardium of FlncgKO mice. Although deleting β1 integrin from cardiomyocytes did not recapitulate the heart rupture phenotype in Flnc knockout mice, deleting both β1 integrin and filamin C from cardiomyocytes resulted in much more severe heart ruptures than deleting filamin C alone. Our results demonstrated that filamin C works in concert with β1 integrin to maintain the structural integrity of myocardium during mammalian heart development.
Collapse
Affiliation(s)
- Tongbin Wu
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Yujun Xu
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Lunfeng Zhang
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Zhengyu Liang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Xiaohai Zhou
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Sylvia M. Evans
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
11
|
Hu HY, Wei SY, Lin SY, Chen LH, Pan CH. “Breaking heart” due to hydrofluoric acid burns in a case of homicide. Forensic Sci Int 2022; 341:111468. [DOI: 10.1016/j.forsciint.2022.111468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/06/2022] [Accepted: 09/18/2022] [Indexed: 11/27/2022]
|
12
|
Chi J, Wang M, Chen J, Hu L, Chen Z, Backman LJ, Zhang W. Topographic Orientation of Scaffolds for Tissue Regeneration: Recent Advances in Biomaterial Design and Applications. Biomimetics (Basel) 2022; 7:131. [PMID: 36134935 PMCID: PMC9496066 DOI: 10.3390/biomimetics7030131] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/30/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Tissue engineering to develop alternatives for the maintenance, restoration, or enhancement of injured tissues and organs is gaining more and more attention. In tissue engineering, the scaffold used is one of the most critical elements. Its characteristics are expected to mimic the native extracellular matrix and its unique topographical structures. Recently, the topographies of scaffolds have received increasing attention, not least because different topographies, such as aligned and random, have different repair effects on various tissues. In this review, we have focused on various technologies (electrospinning, directional freeze-drying, magnetic freeze-casting, etching, and 3-D printing) to fabricate scaffolds with different topographic orientations, as well as discussed the physicochemical (mechanical properties, porosity, hydrophilicity, and degradation) and biological properties (morphology, distribution, adhesion, proliferation, and migration) of different topographies. Subsequently, we have compiled the effect of scaffold orientation on the regeneration of vessels, skin, neural tissue, bone, articular cartilage, ligaments, tendons, cardiac tissue, corneas, skeletal muscle, and smooth muscle. The compiled information in this review will facilitate the future development of optimal topographical scaffolds for the regeneration of certain tissues. In the majority of tissues, aligned scaffolds are more suitable than random scaffolds for tissue repair and regeneration. The underlying mechanism explaining the various effects of aligned and random orientation might be the differences in "contact guidance", which stimulate certain biological responses in cells.
Collapse
Affiliation(s)
- Jiayu Chi
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Mingyue Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| | - Lizhi Hu
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhixuan Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Ludvig J. Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87 Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, SE-901 87 Umeå, Sweden
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| |
Collapse
|
13
|
Dittloff KT, Spanghero E, Solís C, Banach K, Russell B. Transthyretin deposition alters cardiomyocyte sarcomeric architecture, calcium transients, and contractile force. Physiol Rep 2022; 10:e15207. [PMID: 35262277 PMCID: PMC8906053 DOI: 10.14814/phy2.15207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/20/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023] Open
Abstract
Age-related wild-type transthyretin amyloidosis (wtATTR) is characterized by systemic deposition of amyloidogenic fibrils of misfolded transthyretin (TTR) in the connective tissue of many organs. In the heart, this leads to age-related heart failure with preserved ejection fraction (HFpEF). The hypothesis tested is that TTR deposited in vitro disrupts cardiac myocyte cell-to-cell and cell-to-matrix adhesion complexes, resulting in altered calcium handling, force generation, and sarcomeric disorganization. Human iPSC-derived cardiomyocytes and neonatal rat ventricular myocytes (NRVMs), when grown on TTR-coated polymeric substrata mimicking the stiffness of the healthy human myocardium (10 kPa), had decreased contraction and relaxation velocities as well as decreased force production measured using traction force microscopy. Both NRVMs and adult mouse atrial cardiomyocytes had altered calcium kinetics with prolonged transients when cultured on TTR fibril-coated substrates. Furthermore, NRVMs grown on stiff (~GPa), flat or microgrooved substrates coated with TTR fibrils exhibited significantly decreased intercellular electrical coupling as shown by FRAP dynamics of cells loaded with the gap junction-permeable dye calcein-AM, along with decreased gap junction content as determined by quantitative connexin 43 staining. Significant sarcomeric disorganization and loss of sarcomere content, with increased ubiquitin localization to the sarcomere, were seen in NRVMs on various TTR fibril-coated substrata. TTR presence decreased intercellular mechanical junctions as evidenced by quantitative immunofluorescence staining of N-cadherin and vinculin. Current therapies for wtATTR are cost-prohibitive and only slow the disease progression; therefore, better understanding of cardiomyocyte maladaptation induced by TTR amyloid may identify novel therapeutic targets.
Collapse
Affiliation(s)
- Kyle T. Dittloff
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Emanuele Spanghero
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Christopher Solís
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Kathrin Banach
- Department of Internal Medicine/CardiologyRush University Medical CenterChicagoIllinoisUSA
| | - Brenda Russell
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
14
|
Ušaj M, Moretto L, Månsson A. Critical Evaluation of Current Hypotheses for the Pathogenesis of Hypertrophic Cardiomyopathy. Int J Mol Sci 2022; 23:2195. [PMID: 35216312 PMCID: PMC8880276 DOI: 10.3390/ijms23042195] [Citation(s) in RCA: 10] [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: 01/18/2022] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Hereditary hypertrophic cardiomyopathy (HCM), due to mutations in sarcomere proteins, occurs in more than 1/500 individuals and is the leading cause of sudden cardiac death in young people. The clinical course exhibits appreciable variability. However, typically, heart morphology and function are normal at birth, with pathological remodeling developing over years to decades, leading to a phenotype characterized by asymmetric ventricular hypertrophy, scattered fibrosis and myofibrillar/cellular disarray with ultimate mechanical heart failure and/or severe arrhythmias. The identity of the primary mutation-induced changes in sarcomere function and how they trigger debilitating remodeling are poorly understood. Support for the importance of mutation-induced hypercontractility, e.g., increased calcium sensitivity and/or increased power output, has been strengthened in recent years. However, other ideas that mutation-induced hypocontractility or non-uniformities with contractile instabilities, instead, constitute primary triggers cannot yet be discarded. Here, we review evidence for and criticism against the mentioned hypotheses. In this process, we find support for previous ideas that inefficient energy usage and a blunted Frank-Starling mechanism have central roles in pathogenesis, although presumably representing effects secondary to the primary mutation-induced changes. While first trying to reconcile apparently diverging evidence for the different hypotheses in one unified model, we also identify key remaining questions and suggest how experimental systems that are built around isolated primarily expressed proteins could be useful.
Collapse
Affiliation(s)
| | | | - Alf Månsson
- Department of Chemistry and Biomedical Sciences, Faculty of Health and Life Sciences, Linnaeus University, SE-39182 Kalmar, Sweden; (M.U.); (L.M.)
| |
Collapse
|
15
|
Yeruva S, Waschke J. Structure and regulation of desmosomes in intercalated discs: Lessons from epithelia. J Anat 2022; 242:81-90. [PMID: 35128661 PMCID: PMC9773171 DOI: 10.1111/joa.13634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 12/25/2022] Open
Abstract
For electromechanical coupling of cardiomyocytes, intercalated discs (ICDs) are pivotal as highly specialized intercellular contact areas. ICD consists of adhesive contacts, such as desmosomes and adherens junctions (AJs) that are partially intermingled and thereby form an area composita to provide mechanical strength, as well as gap junctions (GJ) and sodium channels for excitation propagation. In contrast, in epithelia, mixed junctions with features of desmosomes and AJs are regarded as transitory primarily during the formation of desmosomes. The anatomy of desmosomes is defined by a typical ultrastructure with dense intracellular plaques anchoring the cadherin-type adhesion molecules to the intermediate filament cytoskeleton. Desmosomal diseases characterized by impaired adhesive and signalling functions of desmosomal contacts lead to arrhythmogenic cardiomyopathy when affecting cardiomyocytes and cause pemphigus when manifesting in keratinocytes or present as cardiocutaneous syndromes when both cell types are targeted by the disease, which underscores the high biomedical relevance of these cell contacts. Therefore, comparative analyses regarding the structure and regulation of desmosomal contacts in cardiomyocytes and epithelial cells are helpful to better understand disease pathogenesis. In this brief review, we describe the structural properties of ICD compared to epithelial desmosomes and suggest that mechanisms regulating adhesion may at least in part be comparable. Also, we discuss whether phenomena such as hyperadhesion or the bidirectional regulation of desmosomes to serve as signalling hubs in epithelial cells may also be relevant for ICD.
Collapse
Affiliation(s)
- Sunil Yeruva
- Ludwig‐Maximilian‐Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I – Vegetative AnatomieMunichGermany
| | - Jens Waschke
- Ludwig‐Maximilian‐Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I – Vegetative AnatomieMunichGermany
| |
Collapse
|
16
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
17
|
Pitsch M, Kant S, Mytzka C, Leube RE, Krusche CA. Autophagy and Endoplasmic Reticulum Stress during Onset and Progression of Arrhythmogenic Cardiomyopathy. Cells 2021; 11:96. [PMID: 35011658 PMCID: PMC8750195 DOI: 10.3390/cells11010096] [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: 11/10/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a heritable, potentially lethal disease without a causal therapy. AC is characterized by focal cardiomyocyte death followed by inflammation and progressive formation of connective tissue. The pathomechanisms leading to structural disease onset and progression, however, are not fully elucidated. Recent studies revealed that dysregulation of autophagy and endoplasmic/sarcoplasmic reticulum (ER/SR) stress plays an important role in cardiac pathophysiology. We therefore examined the temporal and spatial expression patterns of autophagy and ER/SR stress indicators in murine AC models by qRT-PCR, immunohistochemistry, in situ hybridization and electron microscopy. Cardiomyocytes overexpressing the autophagy markers LC3 and SQSTM1/p62 and containing prominent autophagic vacuoles were detected next to regions of inflammation and fibrosis during onset and chronic disease progression. mRNAs of the ER stress markers Chop and sXbp1 were elevated in both ventricles at disease onset. During chronic disease progression Chop mRNA was upregulated in right ventricles. In addition, reduced Ryr2 mRNA expression together with often drastically enlarged ER/SR cisternae further indicated SR dysfunction during this disease phase. Our observations support the hypothesis that locally altered autophagy and enhanced ER/SR stress play a role in AC pathogenesis both at the onset and during chronic progression.
Collapse
Affiliation(s)
| | | | | | - Rudolf E. Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; (M.P.); (S.K.); (C.M.)
| | - Claudia A. Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; (M.P.); (S.K.); (C.M.)
| |
Collapse
|
18
|
Li X, Pan F, He B, Fang C. Inhibition of ADAM10 ameliorates doxorubicin-induced cardiac remodeling by suppressing N-cadherin cleavage. Open Life Sci 2021; 16:856-866. [PMID: 34522779 PMCID: PMC8402944 DOI: 10.1515/biol-2021-0081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/14/2021] [Accepted: 07/17/2021] [Indexed: 12/22/2022] Open
Abstract
The present research was designed to examine the effects of disintegrin metalloproteinases 10 (ADAM10) on the doxorubicin (DOX)-induced dilated cardiomyopathy (DCM) and the mechanisms involved, with a focus on ADAM10-dependent cleavage of N-cadherin. The present study constructed recombinant lentiviral vectors expressing short hairpin RNA (shRNA) targeting the ADAM10 gene. H9C2 cells were treated with the recombinant lentivirus or GI254023 (an ADAM10 inhibitor). The expression level of N-cadherin and its C-terminal fragment1 (CTF1) was tested by western blotting and flow cytometry. The adhesion ability was analyzed using a plate adhesion model. Cardiac function and morphology were assessed in control and lentivirus-transfected rats with or without DOX treatment. The inhibition of ADAM10 activity significantly increased the expression of full-length N-cadherin on the cellular surface and reduced CTF1 generation in vivo and in vitro. The adhesion ability was also increased in ADAM10-knockdown H9C2 cells. Furthermore, DOX-induced myocardial dysfunction was ameliorated in rats transfected with ADAM10-shRNA lentivirus. These findings demonstrated that ADAM10 specifically cleaves N-cadherin in cardiomyocytes. ADAM10-induced N-cadherin cleavage results in changes in the adhesive behavior of cells. Therefore, ADAM10 may serve as a therapeutic target to reverse cardiac remodeling in DCM.
Collapse
Affiliation(s)
- Xiaoou Li
- Department of Neonatology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Feng Pan
- Department of Orthopedics, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Bing He
- Department of Pediatrics, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Chengzhi Fang
- Department of Neonatology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| |
Collapse
|
19
|
Abstract
After first describing the issue contents (Biophysical Reviews-Volume 12 Issue 6), this Editorial goes on to provide a short round-up of the activities of the journal in 2020. Directly following this Editorial are two obituaries marking the recent deaths of Prof. Fumio Oosawa (Japan) and Dr. Herbert Tabor (USA)-two major figures in Biophysical/Biochemical science from the last 100 years.
Collapse
Affiliation(s)
- Damien Hall
- Department of Life Sciences and Applied Chemistry, Nagoya Institute of Technology, Gokiso Showa, Nagoya, 466-8555 Japan
| |
Collapse
|
20
|
Sun Y, Lee SM, Ku BJ, Moon MJ. Fine structure of the intercalated disc and cardiac junctions in the black widow spider Latrodectus mactans. Appl Microsc 2020; 50:20. [PMID: 33580457 PMCID: PMC7818339 DOI: 10.1186/s42649-020-00040-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/15/2020] [Indexed: 11/18/2022] Open
Abstract
Arthropods have an open circulatory system with a simple tubular heart, so it has been estimated that the contractile pumping structure of the cardiac muscle will be less efficient than that of vertebrates. Nevertheless, certain arthropods are known to have far superior properties and characteristics than vertebrates, so we investigated the fine structural features of intercalated discs and cardiac junctions of cardiac muscle cells in the black widow spider Latrodectus mactans. Characteristically, the spider cardiac muscle has typical striated features and represents a functional syncytium that supports multiple connections to adjacent cells by intercalated discs. Histologically, the boundary lamina of each sarcolemma connects to the basement membrane to form an elastic sheath, and the extracellular matrix allows the cells to be anchored to other tissues. Since the intercalated disc is also part of sarcolemma, it contains gap junctions for depolarization and desmosomes that keep the fibers together during cardiac muscle contraction. Furthermore, fascia adherens and macula adherens (desmosomes) were also identified as cell junctions in both sarcolemma and intercalated discs. To enable the coordinated heartbeat of the cardiac muscle, the muscle fibers have neuronal innervations by multiple axons from the motor ganglion.
Collapse
Affiliation(s)
- Yan Sun
- Department of Biological Sciences, Dankook University, 119 Dandae-ro, Cheonan, 31116, South Korea
| | - Seung-Min Lee
- Department of Biological Sciences, Dankook University, 119 Dandae-ro, Cheonan, 31116, South Korea
| | - Bon-Jin Ku
- Department of Biological Sciences, Dankook University, 119 Dandae-ro, Cheonan, 31116, South Korea
| | - Myung-Jin Moon
- Department of Biological Sciences, Dankook University, 119 Dandae-ro, Cheonan, 31116, South Korea.
| |
Collapse
|
21
|
Hall D, Li A, Cooke R. Biophysics of human anatomy and physiology-a Special Issue in honor of Prof. Cristobal dos Remedios on the occasion of his 80 th birthday. Biophys Rev 2020; 12:731-739. [PMID: 32729063 PMCID: PMC7390459 DOI: 10.1007/s12551-020-00745-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
In 2001, Cristobal dos Remedios was made Professor of Anatomy (now emeritus) within Australia's highest-ranked university (University of Sydney). For the majority of his career, he has examined the biomechanics and biophysics of human muscle contraction. To coincide with the occasion of his 80th birthday, this Special Issue has commissioned a collection of review articles from experts exploring biophysical subjects within the general areas of human anatomy and physiology. After introducing the scope and contents of the Issue, we provide a short scientific biography, placing his scientific achievements within the context of the course of his life's developments.
Collapse
Affiliation(s)
- Damien Hall
- Department of Life Sciences and Applied Chemistry, Nagoya Institute of Technology, Gokiso Showa, Nagoya, Aichi, 466-8555, Japan.
| | - Amy Li
- Department of Pharmacy & Biomedical Sciences, La Trobe University, Bendigo, VIC, 3552, Australia
| | - Roger Cooke
- Department of Biochemistry, University of California San Francisco, San Francisco, CA, 94158, USA
| |
Collapse
|