1
|
Kakturskiy LV, Mikhaleva LM, Gioeva ZV, Gutyrchik NA. [The role of imbalance of myocardial cell adhesion proteins in cardiac arrhythmia and heart failure]. Arkh Patol 2024; 86:75-80. [PMID: 39434531 DOI: 10.17116/patol20248605175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
A review of the literature data on the role of imbalance of cellular adhesion proteins (CAP) of the heart in the development of cardiac arrhythmias and heart failure. The CAPS of the intercalated discs belonging to the cadherin and desmin groups provide a mechanical connection of cardiomyocytes, proteins from the connexin group are responsible for the transmission of an electrical impulse. The imbalance of CAP has mainly a hereditary origin and is accompanied by the destruction of intercalated discs, blockage of impulse transmission with the development of electrical instability of the myocardium and cardiac arrhythmias, including ventricular and atrial fibrillation. This is the case with cardiomyopathies, coronary heart disease. Endothelial dysfunction also plays an essential role in atrial fibrillation, which is associated with an imbalance in the CAP of the endothelial lining of the endocardium and blood vessels.
Collapse
Affiliation(s)
- L V Kakturskiy
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - L M Mikhaleva
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Z V Gioeva
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - N A Gutyrchik
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
- Patrice Lumumba Peoples' Friendship University of Russia, Moscow, Russia
| |
Collapse
|
2
|
Zheng M, Li RG, Song J, Zhao X, Tang L, Erhardt S, Chen W, Nguyen BH, Li X, Li M, Wang J, Evans SM, Christoffels VM, Li N, Wang J. Hippo-Yap Signaling Maintains Sinoatrial Node Homeostasis. Circulation 2022; 146:1694-1711. [PMID: 36317529 PMCID: PMC9897204 DOI: 10.1161/circulationaha.121.058777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The sinoatrial node (SAN) functions as the pacemaker of the heart, initiating rhythmic heartbeats. Despite its importance, the SAN is one of the most poorly understood cardiac entities because of its small size and complex composition and function. The Hippo signaling pathway is a molecular signaling pathway fundamental to heart development and regeneration. Although abnormalities of the Hippo pathway are associated with cardiac arrhythmias in human patients, the role of this pathway in the SAN is unknown. METHODS We investigated key regulators of the Hippo pathway in SAN pacemaker cells by conditionally inactivating the Hippo signaling kinases Lats1 and Lats2 using the tamoxifen-inducible, cardiac conduction system-specific Cre driver Hcn4CreERT2 with Lats1 and Lats2 conditional knockout alleles. In addition, the Hippo-signaling effectors Yap and Taz were conditionally inactivated in the SAN. To determine the function of Hippo signaling in the SAN and other cardiac conduction system components, we conducted a series of physiological and molecular experiments, including telemetry ECG recording, echocardiography, Masson Trichrome staining, calcium imaging, immunostaining, RNAscope, cleavage under targets and tagmentation sequencing using antibodies against Yap1 or H3K4me3, quantitative real-time polymerase chain reaction, and Western blotting. We also performed comprehensive bioinformatics analyses of various datasets. RESULTS We found that Lats1/2 inactivation caused severe sinus node dysfunction. Compared with the controls, Lats1/2 conditional knockout mutants exhibited dysregulated calcium handling and increased fibrosis in the SAN, indicating that Lats1/2 function through both cell-autonomous and non-cell-autonomous mechanisms. It is notable that the Lats1/2 conditional knockout phenotype was rescued by genetic deletion of Yap and Taz in the cardiac conduction system. These rescued mice had normal sinus rhythm and reduced fibrosis of the SAN, indicating that Lats1/2 function through Yap and Taz. Cleavage Under Targets and Tagmentation sequencing data showed that Yap potentially regulates genes critical for calcium homeostasis such as Ryr2 and genes encoding paracrine factors important in intercellular communication and fibrosis induction such as Tgfb1 and Tgfb3. Consistent with this, Lats1/2 conditional knockout mutants had decreased Ryr2 expression and increased Tgfb1 and Tgfb3 expression compared with control mice. CONCLUSIONS We reveal, for the first time to our knowledge, that the canonical Hippo-Yap pathway plays a pivotal role in maintaining SAN homeostasis.
Collapse
Affiliation(s)
- Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Rich G Li
- Texas Heart Institute, Houston (R.G.L., X.L.)
| | - Jia Song
- Department of Medicine (Section of Cardiovascular Research), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S., N.L.)
| | - Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Li Tang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas, Houston (S.E., Jun Wang)
| | - Wen Chen
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Bao H Nguyen
- Department of Molecular Physiology and Biophysics (B.H.N.)
| | - Xiao Li
- Texas Heart Institute, Houston (R.G.L., X.L.)
| | - Min Li
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Departments of Pharmacology and Medicine, University of California at San Diego, La Jolla (S.M.E.)
| | - Vincent M Christoffels
- Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, The Netherlands (V.M.C.)
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S., N.L.)
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas, Houston (S.E., Jun Wang)
| |
Collapse
|
3
|
Podyacheva E, Toropova Y. SIRT1 activation and its effect on intercalated disc proteins as a way to reduce doxorubicin cardiotoxicity. Front Pharmacol 2022; 13:1035387. [PMID: 36408244 PMCID: PMC9672938 DOI: 10.3389/fphar.2022.1035387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
According to the World Health Organization, the neoplasm is one of the main reasons for morbidity and mortality worldwide. At the same time, application of cytostatic drugs like an independent type of cancer treatment and in combination with surgical methods, is often associated with the development of cardiovascular complications both in the early and in the delayed period of treatment. Doxorubicin (DOX) is the most commonly used cytotoxic anthracycline antibiotic. DOX can cause both acute and delayed side effects. The problem is still not solved, as evidenced by the continued activity of researchers in terms of developing approaches for the prevention and treatment of cardiovascular complications. It is known, the heart muscle consists of cardiomyocytes connected by intercalated discs (ID), which ensure the structural, electrical, metabolic unity of the heart. Various defects in the ID proteins can lead to the development of cardiovascular diseases of various etiologies, including DOX-induced cardiomyopathy. The search for ways to influence the functioning of ID proteins of the cardiac muscle can become the basis for the creation of new therapeutic approaches to the treatment and prevention of cardiac pathologies. SIRT1 may be an interesting cardioprotective variant due to its wide functional significance. SIRT1 activation triggers nuclear transcription programs that increase the efficiency of cellular, mitochondrial metabolism, increases resistance to oxidative stress, and promotes cell survival. It can be assumed that SIRT1 can not only provide a protective effect at the cardiomyocytes level, leading to an improvement in mitochondrial and metabolic functions, reducing the effects of oxidative stress and inflammatory processes, but also have a protective effect on the functioning of IDs structures of the cardiac muscle.
Collapse
|
4
|
Abstract
Intercalated discs (ICDs) are highly orchestrated structures that connect neighboring cardiomyocytes in the heart. Three major complexes are distinguished in ICD: desmosome, adherens junction (AJ), and gap junction (GJ). Desmosomes are major cell adhesion junctions that anchor cell membrane to the intermediate filament network; AJs connect the actin cytoskeleton of adjacent cells; and gap junctions metabolically and electrically connect the cytoplasm of adjacent cardiomyocytes. All these complexes work as a single unit, the so-called area composita, interdependently rather than individually. Mutation or altered expression of ICD proteins results in various cardiac diseases, such as ARVC (arrhythmogenic right ventricular cardiomyopathy), dilated cardiomyopathy, and hypotrophy cardiomyopathy, eventually leading to heart failure. In this article, we first review the recent findings on the structural organization of ICD and their functions and then focus on the recent advances in molecular pathogenesis of the ICD-related heart diseases, which include two major areas: i) the ICD gene mutations in cardiac diseases, and ii) the involvement of ICD proteins in signal transduction pathways leading to myocardium remodeling and eventual heart failure. These major ICD-related signaling pathways include Wnt/β-catenin pathway, p38 MAPK cascade, Rho-dependent serum response factor (SRF) signaling, calcineurin/NFAT signaling, Hippo kinase cascade, etc., which are differentially regulated in pathological conditions.
Collapse
|
5
|
A Novel DES L115F Mutation Identified by Whole Exome Sequencing is Associated with Inherited Cardiac Conduction Disease. Int J Mol Sci 2019; 20:ijms20246227. [PMID: 31835587 PMCID: PMC6940838 DOI: 10.3390/ijms20246227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/03/2019] [Accepted: 12/08/2019] [Indexed: 12/15/2022] Open
Abstract
Inherited cardiac conduction disease (CCD) is rare; it is caused by a large number of mutations in genes encoding cardiac ion channels and cytoskeletal proteins. Recently, whole-exome sequencing has been successfully used to identify causal mutations for rare monogenic Mendelian diseases. We used trio-based whole-exome sequencing to study a Chinese family with multiple family members affected by CCD, and identified a heterozygous missense mutation (c.343C>T, p.Leu115Phe) in the desmin (DES) gene as the most likely candidate causal mutation for the development of CCD in this family. The mutation is novel and is predicted to affect the conformation of the coiled-coil rod domain of DES according to structural model prediction. Its pathogenicity in desmin protein aggregation was further confirmed by expressing the mutation, both in a cellular model and a CRISPR/CAS9 knock-in mouse model. In conclusion, our results suggest that whole-exome sequencing is a feasible approach to identify candidate genes underlying inherited conduction diseases.
Collapse
|
6
|
Bonilla IM, Belevych AE, Baine S, Stepanov A, Mezache L, Bodnar T, Liu B, Volpe P, Priori S, Weisleder N, Sakuta G, Carnes CA, Radwański PB, Veeraraghavan R, Gyorke S. Enhancement of Cardiac Store Operated Calcium Entry (SOCE) within Novel Intercalated Disk Microdomains in Arrhythmic Disease. Sci Rep 2019; 9:10179. [PMID: 31308393 PMCID: PMC6629850 DOI: 10.1038/s41598-019-46427-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/07/2019] [Indexed: 01/27/2023] Open
Abstract
Store-operated Ca2+ entry (SOCE), a major Ca2+ signaling mechanism in non-myocyte cells, has recently emerged as a component of Ca2+ signaling in cardiac myocytes. Though it has been reported to play a role in cardiac arrhythmias and to be upregulated in cardiac disease, little is known about the fundamental properties of cardiac SOCE, its structural underpinnings or effector targets. An even greater question is how SOCE interacts with canonical excitation-contraction coupling (ECC). We undertook a multiscale structural and functional investigation of SOCE in cardiac myocytes from healthy mice (wild type; WT) and from a genetic murine model of arrhythmic disease (catecholaminergic ventricular tachycardia; CPVT). Here we provide the first demonstration of local, transient Ca2+ entry (LoCE) events, which comprise cardiac SOCE. Although infrequent in WT myocytes, LoCEs occurred with greater frequency and amplitude in CPVT myocytes. CPVT myocytes also evidenced characteristic arrhythmogenic spontaneous Ca2+ waves under cholinergic stress, which were effectively prevented by SOCE inhibition. In a surprising finding, we report that both LoCEs and their underlying protein machinery are concentrated at the intercalated disk (ID). Therefore, localization of cardiac SOCE in the ID compartment has important implications for SOCE-mediated signaling, arrhythmogenesis and intercellular mechanical and electrical coupling in health and disease.
Collapse
Affiliation(s)
- Ingrid M Bonilla
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA.,Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Andriy E Belevych
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Stephen Baine
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Andrei Stepanov
- Laboratory of Cell Pathology, Institute RAS, Saint Petersburg, Russia
| | - Louisa Mezache
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Tom Bodnar
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Bin Liu
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS, USA
| | - Pompeo Volpe
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Silvia Priori
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Noah Weisleder
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Galina Sakuta
- Laboratory of Cell Pathology, Institute RAS, Saint Petersburg, Russia
| | - Cynthia A Carnes
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Division of Pharmacy Practice and Sciences, College of Pharmacy, The Ohio State University, Columbus, OH, USA.,Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Przemysław B Radwański
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA.,Division of Pharmacy Practice and Sciences, College of Pharmacy, The Ohio State University, Columbus, OH, USA.,Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Rengasayee Veeraraghavan
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA. .,Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA.
| | - Sandor Gyorke
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
7
|
Novel Mechanistic Roles for Ankyrin-G in Cardiac Remodeling and Heart Failure. JACC Basic Transl Sci 2018; 3:675-689. [PMID: 30456339 PMCID: PMC6234521 DOI: 10.1016/j.jacbts.2018.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/05/2018] [Accepted: 07/31/2018] [Indexed: 12/25/2022]
Abstract
The pathogenesis of human heart failure is complex, and the creation of new therapeutic strategies for human heart failure is critical. Identifying the molecular pathways underlying heart failure is important to define potential new therapeutic targets. Ankyrin polypeptides serve to target and stabilize membrane proteins in cardiomyocytes. Ankyrin-G levels are altered in humans and mice with heart failure, and mice lacking ankyrin-G in cardiomyocytes develop cardiomyopathy and systolic dysfunction. Mechanistically, ankyrin-G is necessary for the expression and localization of critical myocyte proteins essential for regulating cardiac structural and electrical activity.
Ankyrin polypeptides are intracellular proteins responsible for targeting cardiac membrane proteins. Here, the authors demonstrate that ankyrin-G plays an unexpected role in normal compensatory physiological remodeling in response to myocardial stress and aging; the authors implicate disruption of ankyrin-G in human heart failure. Mechanistically, the authors illustrate that ankyrin-G serves as a key nodal protein required for cardiac myofilament integration with the intercalated disc. Their data define novel in vivo mechanistic roles for ankyrin-G, implicate ankyrin-G as necessary for compensatory cardiac physiological remodeling under stress, and implicate disruption of ankyrin-G in the development and progression of human heart failure.
Collapse
Key Words
- AnkG, ankyrin-G
- DSP, desmoplakin
- ECG, electrocardiogram
- HF, heart failure
- LV, left ventricular
- Nav1.5
- PBS, phosphate-buffered saline
- PKP2, plakophilin-2
- TAC, transverse aortic constriction
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick-end labeling
- WT, wild-type
- ankyrin
- arrhythmia
- cKO, cardiomyocyte-specific knockout
- cytoskeleton
- heart failure
- ion channel
Collapse
|
8
|
Turkowski KL, Tester DJ, Bos JM, Haugaa KH, Ackerman MJ. Whole exome sequencing with genomic triangulation implicatesCDH2-encoded N-cadherin as a novel pathogenic substrate for arrhythmogenic cardiomyopathy. CONGENIT HEART DIS 2017; 12:226-235. [DOI: 10.1111/chd.12462] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Kari L. Turkowski
- Mayo Clinic Graduate School of Biomedical Sciences; Mayo Clinic; Rochester Minnesota USA
| | - David J. Tester
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory; Mayo Clinic; Rochester Minnesota USA
- Department of Cardiovascular Diseases, Division of Heart Rhythm Services; Mayo Clinic; Rochester Minnesota USA
| | - J. Martijn Bos
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory; Mayo Clinic; Rochester Minnesota USA
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology; Mayo Clinic; Rochester Minnesota USA
| | - Kristina H. Haugaa
- Center for Cardiological Innovation, Department of Cardiology; Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo Norway and University of Oslo; Oslo Norway
| | - Michael J. Ackerman
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory; Mayo Clinic; Rochester Minnesota USA
- Department of Cardiovascular Diseases, Division of Heart Rhythm Services; Mayo Clinic; Rochester Minnesota USA
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology; Mayo Clinic; Rochester Minnesota USA
| |
Collapse
|
9
|
Wang HZ, Rosati B, Gordon C, Valiunas V, McKinnon D, Cohen IS, Brink PR. Inhibition of histone deacetylase (HDAC) by 4-phenylbutyrate results in increased junctional conductance between rat corpora smooth muscle cells. Front Pharmacol 2015; 6:9. [PMID: 25691868 PMCID: PMC4315027 DOI: 10.3389/fphar.2015.00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 01/11/2015] [Indexed: 01/02/2023] Open
Abstract
4-phenylbutyrate (4-PB) has been shown to increase the protein content in a number of cells types. One such protein is Connexin43 (Cx43). We show here that 4-phenylbutyrate exposure results in significantly elevated cell to cell coupling, as determined by dual whole cell patch clamp. Incubation with 5 mM 4PB for 24 h or more nearly doubles junctional conductance. Interestingly, mRNA levels for Cx43 declined with exposure to 4-PB while western blot analysis revealed not significant change in protein levels. These data are most consistent with stabilization of the existing Cx43 pool or alterations in the number of functional channels within an existing pool of active and silent channels. These data represent a baseline for testing the efficacy of increased connexin mediated coupling in a variety of multicellular functions including erectile function.
Collapse
Affiliation(s)
- Hong Zhan Wang
- Department of Physiology and Biophysics, Stony Brook University Stony Brook, NY, USA
| | - Barbara Rosati
- Department of Physiology and Biophysics, Stony Brook University Stony Brook, NY, USA ; Department of Physiology and Biophysics, Molecular Cardiology Institute, Stony Brook University Stony Brook, NY, USA
| | - Chris Gordon
- Department of Physiology and Biophysics, Stony Brook University Stony Brook, NY, USA
| | - Virginijus Valiunas
- Department of Physiology and Biophysics, Stony Brook University Stony Brook, NY, USA ; Department of Physiology and Biophysics, Molecular Cardiology Institute, Stony Brook University Stony Brook, NY, USA
| | - David McKinnon
- Department of Physiology and Biophysics, Molecular Cardiology Institute, Stony Brook University Stony Brook, NY, USA ; Department of Neurobiology and Behavior, Stony Brook University Stony Brook, NY, USA
| | - Ira S Cohen
- Department of Physiology and Biophysics, Stony Brook University Stony Brook, NY, USA ; Department of Physiology and Biophysics, Molecular Cardiology Institute, Stony Brook University Stony Brook, NY, USA
| | - Peter R Brink
- Department of Physiology and Biophysics, Stony Brook University Stony Brook, NY, USA ; Department of Physiology and Biophysics, Molecular Cardiology Institute, Stony Brook University Stony Brook, NY, USA
| |
Collapse
|
10
|
Schlipp A, Schinner C, Spindler V, Vielmuth F, Gehmlich K, Syrris P, Mckenna WJ, Dendorfer A, Hartlieb E, Waschke J. Desmoglein-2 interaction is crucial for cardiomyocyte cohesion and function. Cardiovasc Res 2014; 104:245-57. [DOI: 10.1093/cvr/cvu206] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
11
|
Walters B, Hariharan V, Huang H. Dietary levels of acrylamide affect rat cardiomyocyte properties. Food Chem Toxicol 2014; 71:68-73. [DOI: 10.1016/j.fct.2014.05.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/06/2014] [Accepted: 05/28/2014] [Indexed: 01/07/2023]
|
12
|
Wang Q, Lin JLC, Erives AJ, Lin CI, Lin JJC. New insights into the roles of Xin repeat-containing proteins in cardiac development, function, and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 310:89-128. [PMID: 24725425 DOI: 10.1016/b978-0-12-800180-6.00003-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the discovery of Xin repeat-containing proteins in 1996, the importance of Xin proteins in muscle development, function, regeneration, and disease has been continuously implicated. Most Xin proteins are localized to myotendinous junctions of the skeletal muscle and also to intercalated discs (ICDs) of the heart. The Xin gene is only found in vertebrates, which are characterized by a true chambered heart. This suggests that the evolutionary origin of the Xin gene may have played a key role in vertebrate origins. Diverse vertebrates including mammals possess two paralogous genes, Xinα (or Xirp1) and Xinβ (or Xirp2), and this review focuses on the role of their encoded proteins in cardiac muscles. Complete loss of mouse Xinβ (mXinβ) results in the failure of forming ICD, severe growth retardation, and early postnatal lethality. Deletion of mouse Xinα (mXinα) leads to late-onset cardiomyopathy with conduction defects. Molecular studies have identified three classes of mXinα-interacting proteins: catenins, actin regulators/modulators, and ion-channel subunits. Thus, mXinα acts as a scaffolding protein modulating the N-cadherin-mediated adhesion and ion-channel surface expression. Xin expression is significantly upregulated in early stages of stressed hearts, whereas Xin expression is downregulated in failing hearts from various human cardiomyopathies. Thus, mutations in these Xin loci may lead to diverse cardiomyopathies and heart failure.
Collapse
Affiliation(s)
- Qinchuan Wang
- Department of Biology, University of Iowa, Iowa City, Iowa, USA
| | | | - Albert J Erives
- Department of Biology, University of Iowa, Iowa City, Iowa, USA
| | - Cheng-I Lin
- Institute of Physiology, National Defense Medical Center, Taipei, Taiwan, ROC
| | | |
Collapse
|
13
|
Tavora F, Zhang M, Cresswell N, Li L, Fowler D, Franco M, Burke A. Quantitative Immunohistochemistry of Desmosomal Proteins (Plakoglobin, Desmoplakin and Plakophilin), Connexin-43, and N-cadherin in Arrhythmogenic Cardiomyopathy: An Autopsy Study. Open Cardiovasc Med J 2013; 7:28-35. [PMID: 23802019 PMCID: PMC3680985 DOI: 10.2174/1874192401307010028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 02/23/2013] [Accepted: 02/24/2013] [Indexed: 12/19/2022] Open
Abstract
Background: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a genetic disorder related to mutations in desmosomal proteins. The current study tests the hypothesis that immunohistochemical staining for desmosomal proteins is of diagnostic utility by studying autopsy-confirmed cases of ARVC. Methods and Results: We studied 23 hearts from patients dying suddenly with ARVC. Control subject tissues were 21 hearts from people dying from non-cardiac causes (n=15), dilated cardiomyopathy (n=3) and coronary artery disease (n=3). Areas free of fibrofatty change or scarring were assessed on 50 sections from ARVC (24 left ventricle, 26 right ventricle) and 28 sections from controls. Immunohistochemical stains against plakoglobin, plakophilin, desmoplakin, connexin-43, and N-cadherin were applied and area expression analyzed by computerized morphometry. Desmin was stained as a control for fixation and similarly analyzed. The mean area of desmin expression was similar in controls and ARVC (86% vs. 85%, p=0.6). Plakoglobin expression was 4.9% ± 0.3% in controls, vs. 4.6% ± 0.3% in ARVC (p=0.3). Plakophilin staining was 4.8% ± 0.3% in controls vs. 4.4% ± 03% in ARVC (p=0.3). Desmoplakin staining was 3.4% in controls vs. 3.2 ± 0.2% in ARVC (p=0.6). There were no significant differences when staining was compared between right and left ventricles (all p > 0.1). For non-desmosomal proteins, the mean area of connexin-43 staining showed no significant difference by presence of disease. Conclusions: The small and insignificant decrease in junction protein expression in ARVC suggests that immunohistochemistry is not a useful tool for the diagnosis.
Collapse
Affiliation(s)
- Fabio Tavora
- Escola Paulista de Medicina/UNIFESP, Sao Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
14
|
Kline CF, Mohler PJ. Evolving form to fit function: cardiomyocyte intercalated disc and transverse-tubule membranes. CURRENT TOPICS IN MEMBRANES 2013; 72:121-58. [PMID: 24210429 DOI: 10.1016/b978-0-12-417027-8.00004-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The vertebrate cardiac myocyte has evolved a highly organized cellular membrane architecture and cell-cell contacts in order to effectively transmit precisely timed and homogeneous depolarizing waves without failure (>2 billion times/human life span). Two unique specialized membrane domains, the intercalated disc and the transverse tubule (T-tubule), function to ensure the rapid and coordinated propagation of the action potential throughout the heart. Based on their critical roles in structure, signaling, and electric inter- and intracellular communication, it is not surprising that dysfunction in these membrane structures is associated with aberrant vertebrate physiology, resulting in potentially fatal congenital and acquired disease. This chapter will review the fundamental components of cardiomyocyte intercalated disc and transverse-tubule membranes with a focus on linking dysfunction in these membranes with human cardiovascular disease.
Collapse
Affiliation(s)
- Crystal F Kline
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | |
Collapse
|
15
|
Wang Q, Lin JLC, Chan SY, Lin JJC. The Xin repeat-containing protein, mXinβ, initiates the maturation of the intercalated discs during postnatal heart development. Dev Biol 2012; 374:264-80. [PMID: 23261932 DOI: 10.1016/j.ydbio.2012.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/25/2012] [Accepted: 12/04/2012] [Indexed: 10/27/2022]
Abstract
The intercalated disc (ICD) is a unique structure to the heart and plays vital roles in communication and signaling among cardiomyocytes. ICDs are formed and matured during postnatal development through a profound redistribution of the intercellular junctions, as well as recruitment and assembly of more than 200 proteins at the termini of cardiomyocytes. The molecular mechanism underlying this process is not completely understood. The mouse orthologs (mXinα and mXinβ) of human cardiomyopathy-associated (CMYA)/Xin actin-binding repeat-containing protein (XIRP) genes (CMYA1/XIRP1 and CMYA3/XIRP2, respectively) encode proteins localized to ICDs. Ablation of mXinα results in adult late-onset cardiomyopathy with conduction defects and up-regulation of mXinβ. ICD structural defects are found in adult but not juvenile mXinα-null hearts. On the other hand, loss of mXinβ leads to ICD defects at postnatal day 16.5, a developmental stage when the heart is forming ICDs, suggesting mXinβ is required for ICD formation. Using quantitative Western blot, we showed in this study that mXinβ but not mXinα was uniquely up-regulated during the redistribution of intercellular junction from the lateral membrane of cardiomyocytes to their termini. In the absence of mXinβ, the intercellular junctions failed to be restricted to the termini of the cells, and the onset of such defect correlated with the peak expression of mXinβ. Immunofluorescence staining and subcellular fractionation showed that mXinβ preferentially associated with the forming ICDs, further suggesting that mXinβ functioned locally to promote ICD maturation. In contrast, the spatiotemporal expression profile of mXinα and the lack of more severe ICD defects in mXinα-/-;mXinβ-/- double knockout hearts than in mXinβ-/- hearts suggested that mXinα was not essential for the postnatal formation of ICDs. A two-step model for the development of ICD is proposed where mXinβ is essential for the redistribution of intercellular junction components from the lateral puncta to the cell termini.
Collapse
Affiliation(s)
- Qinchuan Wang
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324, USA
| | | | | | | |
Collapse
|
16
|
Chopra A, Patel A, Shieh AC, A. Janmey P, Kresh JY. α-Catenin localization and sarcomere self-organization on N-cadherin adhesive patterns are myocyte contractility driven. PLoS One 2012; 7:e47592. [PMID: 23077648 PMCID: PMC3471892 DOI: 10.1371/journal.pone.0047592] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/12/2012] [Indexed: 12/18/2022] Open
Abstract
The N-cadherin (N-cad) complex plays a crucial role in cardiac cell structure and function. Cadherins are adhesion proteins linking adjacent cardiac cells and, like integrin adhesions, are sensitive to force transmission. Forces through these adhesions are capable of eliciting structural and functional changes in myocytes. Compared to integrins, the mechanisms of force transduction through cadherins are less explored. α-catenin is a major component of the cadherin-catenin complex, thought to provide a link to the cell actin cytoskeleton. Using N-cad micropatterned substrates in an adhesion constrainment model, the results from this study show that α-catenin localizes to regions of highest internal stress in myocytes. This localization suggests that α-catenin acts as an adaptor protein associated with the cadherin mechanosensory apparatus, which is distinct from mechanosensing through integrins. Myosin inhibition in cells bound by integrins to fibronectin-coated patterns disrupts myofibiril organization, whereas on N-cad coated patterns, myosin inhibition leads to better organized myofibrils. This result indicates that the two adhesion systems provide independent mechanisms for regulating myocyte structural organization.
Collapse
Affiliation(s)
- Anant Chopra
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Akash Patel
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Adrian C. Shieh
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Paul A. Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (JYK); (PAJ)
| | - J. Yasha Kresh
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (JYK); (PAJ)
| |
Collapse
|
17
|
Simmons CS, Petzold BC, Pruitt BL. Microsystems for biomimetic stimulation of cardiac cells. LAB ON A CHIP 2012; 12:3235-48. [PMID: 22782590 DOI: 10.1039/c2lc40308k] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The heart is a complex integrated system that leverages mechanoelectrical signals to synchronize cardiomyocyte contraction and push blood throughout the body. The correct magnitude, timing, and distribution of these signals is critical for proper functioning of the heart; aberrant signals can lead to acute incidents, long-term pathologies, and even death. Due to the heart's limited regenerative capacity and the wide variety of pathologies, heart disease is often studied in vitro. However, it is difficult to accurately replicate the cardiac environment outside of the body. Studying the biophysiology of the heart in vitro typically consists of studying single cells in a tightly controlled static environment or whole tissues in a complex dynamic environment. Micro-electromechanical systems (MEMS) allow us to bridge these two extremes by providing increasing complexity for cell culture without having to use a whole tissue. Here, we carefully describe the electromechanical environment of the heart and discuss MEMS specifically designed to replicate these stimulation modes. Strengths, limitations and future directions of various designs are discussed for a variety of applications.
Collapse
Affiliation(s)
- Chelsey S Simmons
- Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | | | | |
Collapse
|
18
|
Cooperative coupling of cell-matrix and cell-cell adhesions in cardiac muscle. Proc Natl Acad Sci U S A 2012; 109:9881-6. [PMID: 22675119 DOI: 10.1073/pnas.1203007109] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Adhesion between cardiac myocytes is essential for the heart to function as an electromechanical syncytium. Although cell-matrix and cell-cell adhesions reorganize during development and disease, the hierarchical cooperation between these subcellular structures is poorly understood. We reasoned that, during cardiac development, focal adhesions mechanically stabilize cells and tissues during myofibrillogenesis and intercalated disc assembly. As the intercalated disc matures, we postulated that focal adhesions disassemble as systolic stresses are transmitted intercellularly. Finally, we hypothesized that pathological remodeling of cardiac microenvironments induces excessive mechanical loading of intercalated discs, leading to assembly of stabilizing focal adhesions adjacent to the junction. To test our model, we engineered μtissues composed of two ventricular myocytes on deformable substrates of tunable elasticity to measure the dynamic organization and functional remodeling of myofibrils, focal adhesions, and intercalated discs as cooperative ensembles. Maturing μtissues increased systolic force while simultaneously developing into an electromechanical syncytium by disassembling focal adhesions at the cell-cell interface and forming mature intercalated discs that transmitted the systolic load. We found that engineering the microenvironment to mimic fibrosis resulted in focal adhesion formation adjacent to the cell-cell interface, suggesting that the intercalated disc required mechanical reinforcement. In these pathological microenvironments, μtissues exhibited further evidence of maladaptive remodeling, including lower work efficiency, longer contraction cycle duration, and weakened relationships between cytoskeletal organization and force generation. These results suggest that the cooperative balance between cell-matrix and cell-cell adhesions in the heart is guided by an architectural and functional hierarchy established during development and disrupted during disease.
Collapse
|
19
|
Genet G, Guilbeau-Frugier C, Honton B, Dague E, Schneider MD, Coatrieux C, Calise D, Cardin C, Nieto C, Payré B, Dubroca C, Marck P, Heymes C, Dubrac A, Arvanitis D, Despas F, Altié MF, Seguelas MH, Delisle MB, Davy A, Sénard JM, Pathak A, Galés C. Ephrin-B1 Is a Novel Specific Component of the Lateral Membrane of the Cardiomyocyte and Is Essential for the Stability of Cardiac Tissue Architecture Cohesion. Circ Res 2012; 110:688-700. [DOI: 10.1161/circresaha.111.262451] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rationale:
Cardiac tissue cohesion relying on highly ordered cardiomyocytes (CM) interactions is critical because most cardiomyopathies are associated with tissue remodeling and architecture alterations.
Objective:
Eph/ephrin system constitutes a ubiquitous system coordinating cellular communications which recently emerged as a major regulator in adult organs. We examined if eph/ephrin could participate in cardiac tissue cyto-organization.
Methods and Results:
We reported the expression of cardiac ephrin-B1 in both endothelial cells and for the first time in CMs where ephrin-B1 localized specifically at the lateral membrane. Ephrin-B1 knock-out (KO) mice progressively developed cardiac tissue disorganization with loss of adult CM rod-shape and sarcomeric and intercalated disk structural disorganization confirmed in CM-specific ephrin-B1 KO mice. CMs lateral membrane exhibited abnormal structure by electron microscopy and notably increased stiffness by atomic force microscopy. In wild-type CMs, ephrin-B1 interacted with claudin-5/ZO-1 complex at the lateral membrane, whereas the complex disappeared in KO/CM-specific ephrin-B1 KO mice. Ephrin-B1 deficiency resulted in decreased mRNA expression of CM basement membrane components and disorganized fibrillar collagen matrix, independently of classical integrin/dystroglycan system. KO/CM-specific ephrin-B1 KO mice exhibited increased left ventricle diameter and delayed atrioventricular conduction. Under pressure overload stress, KO mice were prone to death and exhibited striking tissue disorganization. Finally, failing CMs displayed downregulated ephrin-B1/claudin-5 gene expression linearly related to the ejection fraction.
Conclusions:
Ephrin-B1 is necessary for cardiac tissue architecture cohesion by stabilizing the adult CM morphology through regulation of its lateral membrane. Because decreased ephrin-B1 is associated with molecular/functional cardiac defects, it could represent a new actor in the transition toward heart failure.
Collapse
Affiliation(s)
- Gaël Genet
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Céline Guilbeau-Frugier
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Benjamin Honton
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Etienne Dague
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Michael D. Schneider
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Christelle Coatrieux
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Denis Calise
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Christelle Cardin
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Cécile Nieto
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Bruno Payré
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Caroline Dubroca
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Pauline Marck
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Christophe Heymes
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Alexandre Dubrac
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Dina Arvanitis
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Fabien Despas
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Marie-Françoise Altié
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Marie-Hélène Seguelas
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Marie-Bernadette Delisle
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Alice Davy
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Jean-Michel Sénard
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Atul Pathak
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| | - Céline Galés
- From the Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale UMR 1048 (G.G., B.H., C.C., F.D., M.F.A., M.H.S., J.M.S., A.P., C.G., A.D., D.C., C.D., P.M., C.H.), Department of Histopathology (C.G.F., M.B.D.) and of Clinical Pharmacology (F.D., J.M.S., A.P.), Toulouse University Hospital, CNRS; LAAS, ITAV-UMS3039 (E.D.), Centre de Microscopie Électronique Appliquée à la Biologie, Rangueil Medical Faculty (C.N., B.P.), Development biology
| |
Collapse
|
20
|
Loss of cadherin-binding proteins β-catenin and plakoglobin in the heart leads to gap junction remodeling and arrhythmogenesis. Mol Cell Biol 2012; 32:1056-67. [PMID: 22252313 DOI: 10.1128/mcb.06188-11] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Arrhythmic right ventricular cardiomyopathy (ARVC) is a hereditary heart muscle disease that causes sudden cardiac death (SCD) in young people. Almost half of ARVC patients have a mutation in genes encoding cell adhesion proteins of the desmosome, including plakoglobin (JUP). We previously reported that cardiac tissue-specific plakoglobin (PG) knockout (PG CKO) mice have no apparent conduction abnormality and survive longer than expected. Importantly, the PG homolog, β-catenin (CTNNB1), showed increased association with the gap junction protein connexin43 (Cx43) in PG CKO hearts. To determine whether β-catenin is required to maintain cardiac conduction in the absence of PG, we generated mice lacking both PG and β-catenin specifically in the heart (i.e., double knockout [DKO]). The DKO mice exhibited cardiomyopathy, fibrous tissue replacement, and conduction abnormalities resulting in SCD. Loss of the cadherin linker proteins resulted in dissolution of the intercalated disc (ICD) structure. Moreover, Cx43-containing gap junction plaques were reduced at the ICD, consistent with the arrhythmogenicity of the DKO hearts. Finally, ambulatory electrocardiogram monitoring captured the abrupt onset of spontaneous lethal ventricular arrhythmia in the DKO mice. In conclusion, these studies demonstrate that the N-cadherin-binding partners, PG and β-catenin, are indispensable for maintaining mechanoelectrical coupling in the heart.
Collapse
|
21
|
Guan X, Delo DM, Atala A, Soker S. In vitro cardiomyogenic potential of human amniotic fluid stem cells. J Tissue Eng Regen Med 2011; 5:220-8. [PMID: 20687122 DOI: 10.1002/term.308] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Stem cell therapy for damaged cardiac tissue is currently limited by a number of factors, including inability to obtain sufficient cell numbers, the potential tumorigenicity of certain types of stem cells and the possible link between stem cell therapy and the development of malignant arrhythmias. In this study, we investigated whether human amniotic fluid-derived stem (hAFS) cells could be a potential source of cells for cardiac cell therapy, by testing the in vitro differentiation capabilities. Undifferentiated hAFS cells express several cardiac genes, including the transcription factor mef2, the gap junction connexin43, and H- and N-cadherin. A 24 h incubation with 5-aza-2'-deoxycytidine (5-AZA-dC) induced hAFS cell differentiation along the cardiac lineage. Evidence for this differentiation included morphological changes, upregulation of cardiac-specific genes (cardiac troponin I and cardiac troponin T) and redistribution of connexin43, as well as downregulation of the stem cell marker SRY-box 2 (sox2). When co-cultured with neonatal rat cardiomyocytes (NRCs), hAFS cells formed both mechanical and electrical connections with the NRCs. Dye transfer experiments showed that calcein dye could be transferred from NRCs to hAFS cells through cellular connections. The gap junction connexin43 likely involved in the communication between the two cell types, because 12-O-tetradecanoylphorbol 13-acetate (TPA) could partially block cellular crosstalk. We conclude that hAFS cells can be differentiated into a cardiomyocyte-like phenotype and can establish functional communication with NRCs. Thus, hAFS cells may potentially be used for cardiac cell therapy.
Collapse
Affiliation(s)
- Xuan Guan
- Wake Forest Institute for Regenerative Medicine, Wake Forest University of Health Sciences, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
22
|
Cardiac conduction disturbances and differential effects on atrial and ventricular electrophysiological properties in desmin deficient mice. J Interv Card Electrophysiol 2010; 28:71-80. [PMID: 20390331 DOI: 10.1007/s10840-010-9482-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 03/02/2010] [Indexed: 01/23/2023]
Abstract
PURPOSE Desmin mutations in humans cause desmin-related cardiomyopathy, resulting in heart failure, atrial and ventricular arrhythmias, and sudden cardiac death. The intermediate filament desmin is strongly expressed in striated muscle cells and in Purkinje fibers of the ventricular conduction system. The aim of the present study was to characterize electrophysiological cardiac properties in a desmin-deficient mouse model. METHODS The impact of desmin deficiency on cardiac electrophysiological characteristics was examined in the present study. In vivo electrophysiological studies were carried out in 29 adult desmin deficient (Des-/-) and 19 wild-type (Des+/+) mice. Additionally, epicardial activation mapping was performed in Langendorff-perfused hearts. RESULTS Intracardiac electrograms showed no significant differences in AV, AH, and HV intervals. Functional testing revealed equal AV-nodal refractory periods, sinus-node recovery times, and Wenckebach points. However, compared to the wild-type situation, Des-/- mice were found to have a significantly reduced atrial (23.6+/-10.3 ms vs. 31.8+/-12.5 ms; p=0.045), but prolonged ventricular refractory period (33.0+/-8.7 ms vs. 26.7+/-6.5 ms; p=0.009). The probability of induction of atrial fibrillation was significantly higher in Des-/- mice (Des-/-: 38% vs. Des+/+: 27%; p=0.0255), while ventricular tachycardias significantly were reduced (Des-/-: 7% vs. Des+/+: 21%; p<0.0001). Epicardial activation mapping showed slowing of conduction in the ventricles of Des-/- mice. CONCLUSIONS Des-/- mice exhibit reduced atrial but prolonged ventricular refractory periods and ventricular conduction slowing, accompanied by enhanced inducibility of atrial fibrillation and diminished susceptibility to ventricular arrhythmias. Desmin deficiency does not result in electrophysiological changes present in human desminopathies, suggesting that functional alterations rather than loss of desmin cause the cardiac alterations in these patients.
Collapse
|
23
|
Zhu H, Wang H, Zhang X, Hou X, Cao K, Zou J. Inhibiting N-cadherin-mediated adhesion affects gap junction communication in isolated rat hearts. Mol Cells 2010; 30:193-200. [PMID: 20803092 DOI: 10.1007/s10059-010-0107-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 05/19/2010] [Accepted: 05/27/2010] [Indexed: 12/21/2022] Open
Abstract
Cadherin-mediated adherens junctions is impaired concomitant with a decrease in connexin 43 (Cx43) in diseases or pathological processes. We have investigated the acute effects of adherens junction impairment in isolated rat hearts by introducing Ala-His-Ala-Val-Asp-NH(2) (AHAVD, a synthetic peptide) as a specific inhibitor of N-cadherin. Effect of AHAVD on N-cadherin mediated adhension was analyzed by Cardiomy-ocyte aggregation assay. Laser confocal microscopy showed disrupted cell-cell contacts in cultured neonatal cardiomyocytes co-incubated with 0.2 mM AHAVD. In isolated adult rat hearts, Cx43 was redistributed along the bilateral of cardiomyocytes from the intercalated discs and significant dephosphorylation of Cx43 on serine368 occurred concomitantly with decreased gap junction (GJ) function in dose dependent manner after 1 h perfusion with AHAVD. These results indicate that impairing cad-herin-mediated adhesion by AHAVD rapidly results in Cx43 redistribution and dephosphorylation of serine368, thereby impairing GJ communication function.
Collapse
Affiliation(s)
- Hongjun Zhu
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | | | | | | | | | | |
Collapse
|
24
|
Zhu H, Wang H, Zhang X, Hou X, Cao K, Zou J. Arrhythmogenic properties of dismantling cadherin-mediated adhesion in murine hearts. J Biomed Res 2010; 24:292-300. [PMID: 23554643 PMCID: PMC3596595 DOI: 10.1016/s1674-8301(10)60041-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Indexed: 11/28/2022] Open
Abstract
Objective To evaluate the arrhythmogenic effects of dismantling cadherin-mediated adhesion by recombinant mouse aminopeptidase N (rmAPN) in murine hearts. Methods rmAPN was incubated with cultured neonatal rat cardiomyocytes as well as being infused in adult mice. The cell-cell connections were immunolabelled and observed by laser confocal microscopy. Disruption of the N-terminal of N-cadherin (N-cad) was detected by western blot and quantitative immunofluorescence. The risk of inducible ventricular tachyarrhythmia was evaluated in mice by an electrophysiological study. Results Disrupted cell-cell contact was observed in cultured neonatal rat cardiomyocytes in response to 30-40 ng/µL rmAPN. Loss of the N-terminal in N-cad and altered distribution of connexin 43 (Cx43) were observed in hearts from rmAPN-infused mice. In addition, a reduction of phosphorylated Cx43 was also detected concomitant with redistribution of Cx43. Electrophysiological studies of rmAPN-infused mice showed prolonged QRS duration and increased inducibility of ventricular tachycardias. Conclusion Disruption of N-cad by rmAPN contributes to gap junction remodeling and may elicit arrhythmogenic effects. The disorder of adherent junctions by proteolytic enzymes may play an important role in arrhythmogenic mechanisms in correlated diseases.
Collapse
Affiliation(s)
- Hongjun Zhu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
25
|
Naccarelli GV, Curtis AB. Optimizing the management of atrial fibrillation: focus on current guidelines and the impact of new agents on future recommendations. J Cardiovasc Pharmacol Ther 2010; 15:244-56. [PMID: 20562374 DOI: 10.1177/1074248410370964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia encountered in everyday clinical practice. It affects ~2.3 million individuals in the United States, and the prevalence is expected to increase ~2.5-fold over the next 40 years. Atrial fibrillation accounts for more than 2 million hospitalizations each year and contributes to nearly 67 000 deaths. Our understanding of the pathophysiology of AF has increased dramatically over the past few decades. Recent treatment guidelines have heightened our awareness of the challenges involved in the treatment of AF and provided useful recommendations for its diagnosis and management. Because AF is usually associated with multiple comorbid conditions, greater emphasis must be placed on individualizing treatment. This review focuses on current treatment guidelines for patients with AF, assessing the benefits and shortcomings of current pharmacologic options and discussing new agents and trials that may provide better opportunities to improve and individualize patient management.
Collapse
Affiliation(s)
- Gerald V Naccarelli
- Division of Cardiology, Penn State Heart & Vascular Institute, Hershey, PA 17033, USA.
| | | |
Collapse
|
26
|
Abstract
Intercalated disks (ICDs) are highly organized cell-cell adhesion structures, which connect cardiomyocytes to one another. They are composed of three major complexes: desmosomes, fascia adherens, and gap junctions. Desmosomes and fascia adherens junction are necessary for mechanically coupling and reinforcing cardiomyocytes, whereas gap junctions are essential for rapid electrical transmission between cells. Because human genetics and mouse models have revealed that mutations and/or deficiencies in various ICD components can lead to cardiomyopathies and arrhythmias, considerable attention has focused on the biologic function of the ICD. This review will discuss recent scientific developments related to the ICD and focus on its role in regulating cardiac muscle structure, signaling, and disease.
Collapse
Affiliation(s)
- Farah Sheikh
- Department of Medicine, University of California-San Diego, CA 92093, USA
| | | | | |
Collapse
|
27
|
Wang Q, Lin JLC, Reinking BE, Feng HZ, Chan FC, Lin CI, Jin JP, Gustafson-Wagner EA, Scholz TD, Yang B, Lin JJC. Essential roles of an intercalated disc protein, mXinbeta, in postnatal heart growth and survival. Circ Res 2010; 106:1468-78. [PMID: 20360251 DOI: 10.1161/circresaha.109.212787] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE The Xin repeat-containing proteins mXinalpha and mXinbeta localize to the intercalated disc of mouse heart and are implicated in cardiac development and function. The mXinalpha directly interacts with beta-catenin, p120-catenin, and actin filaments. Ablation of mXinalpha results in adult late-onset cardiomyopathy with conduction defects. An upregulation of the mXinbeta in mXinalpha-deficient hearts suggests a partial compensation. OBJECTIVE The essential roles of mXinbeta in cardiac development and intercalated disc maturation were investigated. METHODS AND RESULTS Ablation of mXinbeta led to abnormal heart shape, ventricular septal defects, severe growth retardation, and postnatal lethality with no upregulation of the mXinalpha. Postnatal upregulation of mXinbeta in wild-type hearts, as well as altered apoptosis and proliferation in mXinbeta-null hearts, suggests that mXinbeta is required for postnatal heart remodeling. The mXinbeta-null hearts exhibited a misorganized myocardium as detected by histological and electron microscopic studies and an impaired diastolic function, as suggested by echocardiography and a delay in switching off the slow skeletal troponin I. Loss of mXinbeta resulted in the failure of forming mature intercalated discs and the mislocalization of mXinalpha and N-cadherin. The mXinbeta-null hearts showed upregulation of active Stat3 (signal transducer and activator of transcription 3) and downregulation of the activities of Rac1, insulin-like growth factor 1 receptor, protein kinase B, and extracellular signal-regulated kinases 1 and 2. CONCLUSIONS These findings identify not only an essential role of mXinbeta in the intercalated disc maturation but also mechanisms of mXinbeta modulating N-cadherin-mediated adhesion signaling and its crosstalk signaling for postnatal heart growth and animal survival.
Collapse
Affiliation(s)
- Qinchuan Wang
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Iacobas DA, Iacobas S, Thomas N, Spray DC. Sex-dependent gene regulatory networks of the heart rhythm. Funct Integr Genomics 2009; 10:73-86. [PMID: 19756788 DOI: 10.1007/s10142-009-0137-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Revised: 08/19/2009] [Accepted: 08/22/2009] [Indexed: 01/27/2023]
Abstract
Expression level, control, and intercoordination of 66 selected heart rhythm determinant (HRD) genes were compared in atria and ventricles of four male and four female adult mice. We found that genes encoding various adrenergic receptors, ankyrins, ion channels and transporters, connexins, cadherins, plakophilins, and other components of the intercalated discs form a complex network that is chamber dependent and differs between the two sexes. In addition, most HRD genes in atria had higher expression in males than in females, while in ventricles, expression levels were mostly higher in females than in males. Moreover, significant chamber differences were observed between the sexes, with higher expression in atria than ventricles for males and higher expression in ventricles than atria for females. We have ranked the selected genes according to their prominence (new concept) within the HRD gene web defined as extent of expression coordination with the other web genes and stability of expression. Interestingly, the prominence hierarchy was substantially different between the two sexes. Taken together, these findings indicate that the organizational principles of the heart rhythm transcriptome are sex dependent, with the newly introduced prominence analysis allowing identification of genes that are pivotal for the sexual dichotomy.
Collapse
Affiliation(s)
- D A Iacobas
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Kennedy Center, New York, NY 10461, USA.
| | | | | | | |
Collapse
|
29
|
Liang X, Sun Y, Ye M, Scimia MC, Cheng H, Martin J, Wang G, Rearden A, Wu C, Peterson KL, Powell HC, Evans SM, Chen J. Targeted ablation of PINCH1 and PINCH2 from murine myocardium results in dilated cardiomyopathy and early postnatal lethality. Circulation 2009; 120:568-76. [PMID: 19652092 DOI: 10.1161/circulationaha.109.864686] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND PINCH proteins are 5 LIM domain-only adaptor proteins that function as key components of the integrin signaling pathway and play crucial roles in multiple cellular processes. Two PINCH proteins, PINCH1 and PINCH2, have been described in mammals and share high homology. Both PINCH1 and PINCH2 are ubiquitously expressed in most tissues and organs, including myocardium. Cardiac-specific PINCH1 knockout or global PINCH2 knockout mice exhibit no basal cardiac phenotype, which may reflect a redundant role for these 2 PINCH proteins in myocardium. A potential role for PINCH proteins in myocardium remains unknown. METHODS AND RESULTS To define the role of PINCH in myocardium, we generated mice that were doubly homozygous null for PINCH1 and PINCH2 in myocardium. Resulting mutants were viable at birth but developed dilated cardiomyopathy and died of heart failure within 4 weeks. Mutant hearts exhibited disruptions of intercalated disks and costameres accompanied by fibrosis. Furthermore, multiple cell adhesion proteins exhibited reduced expression and were mislocalized. Mutant cardiomyocytes were significantly smaller and irregular in size. In addition, we observed that the absence of either PINCH1 or PINCH2 in myocardium leads to exacerbated cardiac injury and deterioration in cardiac function after myocardial infarction. CONCLUSIONS These results demonstrate essential roles for PINCHs in myocardial growth, maturation, remodeling, and function and highlight the importance of studying the role of PINCHs in human cardiac injury and cardiomyopathy.
Collapse
Affiliation(s)
- Xingqun Liang
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0613, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Noorman M, van der Heyden MA, van Veen TA, Cox MG, Hauer RN, de Bakker JM, van Rijen HV. Cardiac cell–cell junctions in health and disease: Electrical versus mechanical coupling. J Mol Cell Cardiol 2009; 47:23-31. [DOI: 10.1016/j.yjmcc.2009.03.016] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 03/12/2009] [Accepted: 03/19/2009] [Indexed: 10/21/2022]
|
31
|
Fischer R, Poller W, Schultheiss HP, Gotthardt M. CAR-diology--a virus receptor in the healthy and diseased heart. J Mol Med (Berl) 2009; 87:879-84. [PMID: 19495568 DOI: 10.1007/s00109-009-0489-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Revised: 05/13/2009] [Accepted: 05/18/2009] [Indexed: 12/19/2022]
Abstract
The interplay of diverse cell-contact proteins is required for normal cardiac function and determines the mechanical and electrical properties of the heart. A specialized structure between cardiomyocytes-the intercalated disk-contains a high density of these proteins, which are assembled into adherens junctions, desmosomes, and gap junctions. The Coxsackievirus-adenovirus receptor (CAR) as a tight junction protein of the intercalated disk has recently been implied in cardiac remodeling and electrical conductance between atria and ventricle. This review summarizes recent in vivo studies that relate CAR to heart disease and how they could translate to improved diagnosis and therapy of viral myocarditis and arrhythmia.
Collapse
Affiliation(s)
- Robert Fischer
- Electrophysiology, Experimental and Clinical Research Center, Charité University Medical Center, Humboldt-University, Berlin, 13125, Germany
| | | | | | | |
Collapse
|
32
|
Derangeon M, Spray DC, Bourmeyster N, Sarrouilhe D, Hervé JC. Reciprocal influence of connexins and apical junction proteins on their expressions and functions. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2008; 1788:768-78. [PMID: 19046940 DOI: 10.1016/j.bbamem.2008.10.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 10/29/2008] [Accepted: 10/30/2008] [Indexed: 01/17/2023]
Abstract
Membranes of adjacent cells form intercellular junctional complexes to mechanically anchor neighbour cells (anchoring junctions), to seal the paracellular space and to prevent diffusion of integral proteins within the plasma membrane (tight junctions) and to allow cell-to-cell diffusion of small ions and molecules (gap junctions). These different types of specialised plasma membrane microdomains, sharing common adaptor molecules, particularly zonula occludens proteins, frequently present intermingled relationships where the different proteins co-assemble into macromolecular complexes and their expressions are co-ordinately regulated. Proteins forming gap junction channels (connexins, particularly) and proteins fulfilling cell attachment or forming tight junction strands mutually influence expression and functions of one another.
Collapse
Affiliation(s)
- Mickaël Derangeon
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers, Poitiers, F-86022, France
| | | | | | | | | |
Collapse
|
33
|
Mays TA, Binkley PF, Lesinski A, Doshi AA, Quaile MP, Margulies KB, Janssen PM, Rafael-Fortney JA. Claudin-5 levels are reduced in human end-stage cardiomyopathy. J Mol Cell Cardiol 2008; 45:81-7. [DOI: 10.1016/j.yjmcc.2008.04.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 04/15/2008] [Accepted: 04/16/2008] [Indexed: 11/27/2022]
|
34
|
Severs NJ, Bruce AF, Dupont E, Rothery S. Remodelling of gap junctions and connexin expression in diseased myocardium. Cardiovasc Res 2008; 80:9-19. [PMID: 18519446 PMCID: PMC2533424 DOI: 10.1093/cvr/cvn133] [Citation(s) in RCA: 396] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gap junctions form the cell-to-cell pathways for propagation of the precisely orchestrated patterns of current flow that govern the regular rhythm of the healthy heart. As in most tissues and organs, multiple connexin types are expressed in the heart: connexin43 (Cx43), Cx40 and Cx45 are found in distinctive combinations and relative quantities in different, functionally-specialized subsets of cardiac myocyte. Mutations in genes that encode connexins have only rarely been identified as being a cause of human cardiac disease, but remodelling of connexin expression and gap junction organization are well documented in acquired adult heart disease, notably ischaemic heart disease and heart failure. Remodelling may take the form of alterations in (i) the distribution of gap junctions and (ii) the amount and type of connexins expressed. Heterogeneous reduction in Cx43 expression and disordering in gap junction distribution feature in human ventricular disease and correlate with electrophysiologically identified arrhythmic changes and contractile dysfunction in animal models. Disease-related alterations in Cx45 and Cx40 expression have also been reported, and some of the functional implications of these are beginning to emerge. Apart from ventricular disease, various features of gap junction organization and connexin expression have been implicated in the initiation and persistence of the most common form of atrial arrhythmia, atrial fibrillation, though the disparate findings in this area remain to be clarified. Other major tasks ahead focus on the Purkinje/working ventricular myocyte interface and its role in normal and abnormal impulse propagation, connexin-interacting proteins and their regulatory functions, and on defining the precise functional properties conferred by the distinctive connexin co-expression patterns of different myocyte types in health and disease.
Collapse
Affiliation(s)
- Nicholas J Severs
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK.
| | | | | | | |
Collapse
|
35
|
Bistola V, Nikolopoulou M, Derventzi A, Kataki A, Sfyras N, Nikou N, Toutouza M, Toutouzas P, Stefanadis C, Konstadoulakis MM. Long-term primary cultures of human adult atrial cardiac myocytes: cell viability, structural properties and BNP secretion in vitro. Int J Cardiol 2008; 131:113-22. [PMID: 18255169 DOI: 10.1016/j.ijcard.2007.10.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 09/27/2007] [Accepted: 10/20/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Human adult cardiomyocytes (CM) have been used in short-term cultures for in vitro studies of the adult myocardium. However, little information is available regarding human adult CMs cultured for long term (>2 weeks). METHODS Human adult CMs were isolated from atrial specimens of 43 patients undergoing cardiopulmonary bypass surgery. Cell viability, cytoskeletal properties, intercellular junctional mediators and responsiveness to extracellular stimuli were monitored in CM cultures for 8 weeks. RESULTS Absolute numbers of CMs decreased through the first 2 weeks, with substantially lower rates of cell loss thereafter. Apoptosis predominated over necrosis as the principal mode of cell death, affecting 4.1+/-1.6% of freshly dissociated cells, that declined in culture (3.6+/-1.0% week 1, 1.3+/-0.5% week 2). CMs maintained rod-shaped morphology and cross-striated expression pattern of sarcomeric proteins desmin and beta-myosin heavy chain for the first 4 weeks. Levels of desmin remained stable on first 3 weeks, but declined thereafter. CMs expressed cardiac-specific adherence molecule N-cadherin throughout the culture duration, indicating conserved contractile potential. CMs remained functional early in culture, as indicated by BNP secretion, with maximal levels on 1st week that declined gradually by week 4. Cell responsiveness to metabolic stresses (serum deprivation) was detected, inducing an early (6 h) 1.8-fold increase in levels of BNP. CONCLUSION Long-term cultured human adult CMs maintain morphological integrity, adult-type cytoskeletal protein expression, cell-cell communication potential and functionality for 3-4 weeks in vitro.
Collapse
Affiliation(s)
- Vasiliki Bistola
- Laboratory of Surgical Research, 1st Propaedeutic Department of Surgery, University of Athens Medical School, Hippocration Hospital, Greece.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Kuwabara M, Kakinuma Y, Katare RG, Ando M, Yamasaki F, Doi Y, Sato T. Granulocyte colony-stimulating factor activates Wnt signal to sustain gap junction function through recruitment of β-catenin and cadherin. FEBS Lett 2007; 581:4821-30. [PMID: 17888912 DOI: 10.1016/j.febslet.2007.09.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2007] [Revised: 09/04/2007] [Accepted: 09/04/2007] [Indexed: 11/15/2022]
Abstract
Our previous study reveals that connexin (Cx) 43 is targeted by ACh to prevent lethal arrhythmia. Granulocyte colony-stimulating factor (G-CSF), used against ischemic heart failure, may be another candidate, however, with unknown mechanisms. Therefore, we investigated the cellular effects of G-CSF. G-CSF activated the Wnt and Jak2 signals in cardiomyocytes, and up-regulated Cx43 protein and phosphorylation levels. In addition, G-CSF enhanced the localization of Cx43, beta-catenin and cadherin on the plasma membrane. G-CSF inhibited the reduction of Cx43 by enhancing Cx43 anchoring and sustained the cell-cell communication during hypoxia. Consequently, G-CSF suppressed ventricular arrhythmia induced by myocardial infarction. As a result, G-CSF could be used as a therapeutic tool for arrhythmia.
Collapse
Affiliation(s)
- Masanori Kuwabara
- Department of Medicine and Geriatrics, Kochi Medical School, Nankoku, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Celes MRN, Torres-Dueñas D, Alves-Filho JC, Duarte DB, Cunha FQ, Rossi MA. Reduction of gap and adherens junction proteins and intercalated disc structural remodeling in the hearts of mice submitted to severe cecal ligation and puncture sepsis*. Crit Care Med 2007; 35:2176-85. [PMID: 17855834 DOI: 10.1097/01.ccm.0000281454.97901.01] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The present study describes intercalated disc remodeling under both protein expression and structural features in experimental severe sepsis induced by cecal ligation and puncture in mice. DESIGN Controlled animal study. SETTING University research laboratory. SUBJECTS Male C57BL/6 mice. INTERVENTIONS Mice were submitted to moderate and severe septic injury by cecal ligation and puncture. MEASUREMENT AND MAIN RESULTS Severe septic injury was accompanied by a large number of bacteria in the peritoneal cavity and blood, high levels of tumor necrosis factor-alpha, and monocyte inflammatory protein-1alpha in the septic focus and serum, marked hypotension, and a high mortality rate. Western blot analysis and immunofluorescence showed a marked decrease of key gap and adherens junction proteins (connexin43 and N-cadherin, respectively) in mice submitted to severe septic injury. These changes may result in the loss of intercalated disc structural integrity, characterized in the electron microscopic study by partial separation or dehiscence of gap junctions and adherens junctions. CONCLUSIONS Our data provide important insight regarding the alterations in intercalated disc components resulting from severe septic injury. The intercalated disc remodeling under both protein expression and structural features in experimental severe sepsis induced by cecal ligation and puncture may be partly responsible for myocardial depression in sepsis/septic shock. Although further electrophysiological studies in animals and humans are needed to determine the effect of these alterations on myocardial conduction velocity, the abnormal variables may emerge as therapeutic targets, and their modulation might provide beneficial effects on future cardiovascular outcomes and mortality in sepsis.
Collapse
Affiliation(s)
- Mara Rúbia N Celes
- Department of Pathology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|