1
|
Campos I, Richter B, Thomas SM, Czaya B, Yanucil C, Kentrup D, Fajol A, Li Q, Secor SM, Faul C. FGFR4 Is Required for Concentric Growth of Cardiac Myocytes during Physiologic Cardiac Hypertrophy. J Cardiovasc Dev Dis 2024; 11:320. [PMID: 39452290 PMCID: PMC11508992 DOI: 10.3390/jcdd11100320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Fibroblast growth factor (FGF) 23 is a bone-derived hormone that promotes renal phosphate excretion. Serum FGF23 is increased in chronic kidney disease (CKD) and contributes to pathologic cardiac hypertrophy by activating FGF receptor (FGFR) 4 on cardiac myocytes, which might lead to the high cardiovascular mortality in CKD patients. Increases in serum FGF23 levels have also been observed following endurance exercise and in pregnancy, which are scenarios of physiologic cardiac hypertrophy as an adaptive response of the heart to increased demand. To determine whether FGF23/FGFR4 contributes to physiologic cardiac hypertrophy, we studied FGFR4 knockout mice (FGFR4-/-) during late pregnancy. In comparison to virgin littermates, pregnant wild-type and FGFR4-/- mice showed increases in serum FGF23 levels and heart weight; however, the elevation in myocyte area observed in pregnant wild-type mice was abrogated in pregnant FGFR4-/- mice. This outcome was supported by treatments of cultured cardiac myocytes with serum from fed Burmese pythons, another model of physiologic hypertrophy, where the co-treatment with an FGFR4-specific inhibitor abrogated the serum-induced increase in cell area. Interestingly, we found that in pregnant mice, the heart, and not the bone, shows elevated FGF23 expression, and that increases in serum FGF23 are not accompanied by changes in phosphate metabolism. Our study suggests that in physiologic cardiac hypertrophy, the heart produces FGF23 that contributes to hypertrophic growth of cardiac myocytes in a paracrine and FGFR4-dependent manner, and that the kidney does not respond to heart-derived FGF23.
Collapse
Affiliation(s)
- Isaac Campos
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Beatrice Richter
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Sarah Madison Thomas
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Brian Czaya
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Christopher Yanucil
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Dominik Kentrup
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Abul Fajol
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Qing Li
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Stephen M. Secor
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA;
| | - Christian Faul
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| |
Collapse
|
2
|
Chen JK, Ramesh S, Islam MN, Shibu MA, Kuo CH, Hsieh DJY, Lin SZ, Kuo WW, Huang CY, Ho TJ. Artemisia argyi mitigates doxorubicin-induced cardiotoxicity by inhibiting mitochondrial dysfunction through the IGF-IIR/Drp1/GATA4 signaling pathway. Biotechnol Appl Biochem 2024. [PMID: 39375847 DOI: 10.1002/bab.2671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Doxorubicin (DOX) is mostly utilized as a wide range of antitumor anthracycline to treat different cancers. The severe antagonistic impacts of DOX on cardiotoxicity constrain its clinical application. Many mechanisms are involved in cardiac toxicity induced by DOX in the human body. Mitochondria is a central part of fatty acid and glucose metabolism. Thus, impaired mitochondrial metabolism can increase heart failure risk, which can play a vital role in cardiomyocyte mitochondrial dysfunction. This study aimed to assess the possible cardioprotective effect of water-extracted Artemisia argyi (AA) against the side effect of DOX in H9c2 cells and whether these protective effects are mediated through IGF-IIR/Drp1/GATA4 signaling pathways. Although several studies proved that AA extract has benefits for various diseases, its cardiac effects have not yet been identified. The H9c2 cells were exposed to 1 μM to establish a model of cardiac toxicity. The results revealed that water-extracted AA could block the expression of IGF-IIR/calcineurin signaling pathways induced by DOX. Notably, our results also showed that AA treatment markedly attenuated Akt phosphorylation and cleaved caspase 3, and the nuclear translocation markers NFATC3 and p-GATA4. Using actin staining for hypertrophy, we determined that AA can reduce the effect of mitochondrial reactive oxygen species and cell size. These findings suggest that water-extracted AA could be a suitable candidate for preventing DOX-induced cardiac damage.
Collapse
Affiliation(s)
- Jhong-Kuei Chen
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Samiraj Ramesh
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Research and Innovation, Institute of Biotechnology, Saveetha School of Engineering (SSE), Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Md Nazmul Islam
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | | | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
3
|
Somee LR, Barati A, Shahsavani MB, Hoshino M, Hong J, Kumar A, Moosavi-Movahedi AA, Amanlou M, Yousefi R. Understanding the structural and functional changes and biochemical pathomechanism of the cardiomyopathy-associated p.R123W mutation in human αB-crystallin. Biochim Biophys Acta Gen Subj 2024; 1868:130579. [PMID: 38307443 DOI: 10.1016/j.bbagen.2024.130579] [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: 12/15/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
αB-crystallin, a member of the small heat shock protein (sHSP) family, is expressed in diverse tissues, including the eyes, brain, muscles, and heart. This protein plays a crucial role in maintaining eye lens transparency and exhibits holdase chaperone and anti-apoptotic activities. Therefore, structural and functional changes caused by genetic mutations in this protein may contribute to the development of disorders like cataract and cardiomyopathy. Recently, the substitution of arginine 123 with tryptophan (p.R123W mutation) in human αB-crystallin has been reported to trigger cardiomyopathy. In this study, human αB-crystallin was expressed in Escherichia coli (E. coli), and the missense mutation p.R123W was created using site-directed mutagenesis. Following purification via anion exchange chromatography, the structural and functional properties of both proteins were investigated and compared using a wide range of spectroscopic and microscopic methods. The p.R123W mutation induced significant alterations in the secondary, tertiary, and quaternary structures of human αB-crystallin. This pathogenic mutation resulted in an increased β-sheet structure and formation of protein oligomers with larger sizes compared to the wild-type protein. The mutant protein also exhibited reduced chaperone activity and lower thermal stability. Atomic force microscopy (AFM) and transmission electron microscopy (TEM) demonstrated that the p.R123W mutant protein is more prone to forming amyloid aggregates. The structural and functional changes observed in the p.R123W mutant protein, along with its increased propensity for aggregation, could impact its proper functional interaction with the target proteins in the cardiac muscle, such as calcineurin. Our results provide an explanation for the pathogenic intervention of p.R123W mutant protein in the occurrence of hypertrophic cardiomyopathy (HCM).
Collapse
Affiliation(s)
- Leila Rezaei Somee
- Protein Chemistry Laboratory (PCL), Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Anis Barati
- Protein Chemistry Laboratory (PCL), Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | | | - Masaru Hoshino
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Jun Hong
- School of Life Sciences, Henan University, Kaifen, People's Republic of China
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India
| | | | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yousefi
- Protein Chemistry Laboratory (PCL), Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|
4
|
Curaj A, Vanholder R, Loscalzo J, Quach K, Wu Z, Jankowski V, Jankowski J. Cardiovascular Consequences of Uremic Metabolites: an Overview of the Involved Signaling Pathways. Circ Res 2024; 134:592-613. [PMID: 38422175 DOI: 10.1161/circresaha.123.324001] [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] [Indexed: 03/02/2024]
Abstract
The crosstalk of the heart with distant organs such as the lung, liver, gut, and kidney has been intensively approached lately. The kidney is involved in (1) the production of systemic relevant products, such as renin, as part of the most essential vasoregulatory system of the human body, and (2) in the clearance of metabolites with systemic and organ effects. Metabolic residue accumulation during kidney dysfunction is known to determine cardiovascular pathologies such as endothelial activation/dysfunction, atherosclerosis, cardiomyocyte apoptosis, cardiac fibrosis, and vascular and valvular calcification, leading to hypertension, arrhythmias, myocardial infarction, and cardiomyopathies. However, this review offers an overview of the uremic metabolites and details their signaling pathways involved in cardiorenal syndrome and the development of heart failure. A holistic view of the metabolites, but more importantly, an exhaustive crosstalk of their known signaling pathways, is important for depicting new therapeutic strategies in the cardiovascular field.
Collapse
Affiliation(s)
- Adelina Curaj
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
| | - Raymond Vanholder
- Department of Internal Medicine and Pediatrics, Nephrology Section, University Hospital, Ghent, Belgium (R.V.)
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.)
| | - Kaiseng Quach
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
| | - Zhuojun Wu
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
| | - Vera Jankowski
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
- Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht, University of Maastricht, the Netherlands (J.J.)
- Aachen-Maastricht Institute for Cardiorenal Disease, RWTH Aachen University, Aachen, Germany (J.J.)
| |
Collapse
|
5
|
Ren FF, Zhao L, Jiang XY, Zhang JJ, Gou JM, Yu XY, Wu SJ, Li L. Sphingosylphosphorylcholine alleviates pressure overload-induced myocardial remodeling in mice via inhibiting CaM-JNK/p38 signaling pathway. Acta Pharmacol Sin 2024; 45:312-326. [PMID: 37833535 PMCID: PMC10789762 DOI: 10.1038/s41401-023-01168-6] [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: 07/06/2023] [Accepted: 09/13/2023] [Indexed: 10/15/2023] Open
Abstract
Apoptosis plays a critical role in the development of heart failure, and sphingosylphosphorylcholine (SPC) is a bioactive sphingolipid naturally occurring in blood plasma. Some studies have shown that SPC inhibits hypoxia-induced apoptosis in myofibroblasts, the crucial non-muscle cells in the heart. Calmodulin (CaM) is a known SPC receptor. In this study we investigated the role of CaM in cardiomyocyte apoptosis in heart failure and the associated signaling pathways. Pressure overload was induced in mice by trans-aortic constriction (TAC) surgery. TAC mice were administered SPC (10 μM·kg-1·d-1) for 4 weeks post-surgery. We showed that SPC administration significantly improved survival rate and cardiac hypertrophy, and inhibited cardiac fibrosis in TAC mice. In neonatal mouse cardiomyocytes, treatment with SPC (10 μM) significantly inhibited Ang II-induced cardiomyocyte hypertrophy, fibroblast-to-myofibroblast transition and cell apoptosis accompanied by reduced Bax and phosphorylation levels of CaM, JNK and p38, as well as upregulated Bcl-2, a cardiomyocyte-protective protein. Thapsigargin (TG) could enhance CaM functions by increasing Ca2+ levels in cytoplasm. TG (3 μM) annulled the protective effect of SPC against Ang II-induced cardiomyocyte apoptosis. Furthermore, we demonstrated that SPC-mediated inhibition of cardiomyocyte apoptosis involved the regulation of p38 and JNK phosphorylation, which was downstream of CaM. These results offer new evidence for SPC regulation of cardiomyocyte apoptosis, potentially providing a new therapeutic target for cardiac remodeling following stress overload.
Collapse
Affiliation(s)
- Fang-Fang Ren
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lin Zhao
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xian-Yun Jiang
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jing-Jing Zhang
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jia-Min Gou
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiao-Yu Yu
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shu-Jin Wu
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lei Li
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
6
|
Ni M, Li Y, Wei J, Song Z, Wang H, Yao J, Chen YX, Belke D, Estillore JP, Wang R, Vallmitjana A, Benitez R, Hove-Madsen L, Feng W, Chen J, Roston TM, Sanatani S, Lehman A, Chen SRW. Increased Ca 2+ Transient Underlies RyR2-Related Left Ventricular Noncompaction. Circ Res 2023; 133:177-192. [PMID: 37325910 DOI: 10.1161/circresaha.123.322504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND A loss-of-function cardiac ryanodine receptor (RyR2) mutation, I4855M+/-, has recently been linked to a new cardiac disorder termed RyR2 Ca2+ release deficiency syndrome (CRDS) as well as left ventricular noncompaction (LVNC). The mechanism by which RyR2 loss-of-function causes CRDS has been extensively studied, but the mechanism underlying RyR2 loss-of-function-associated LVNC is unknown. Here, we determined the impact of a CRDS-LVNC-associated RyR2-I4855M+/- loss-of-function mutation on cardiac structure and function. METHODS We generated a mouse model expressing the CRDS-LVNC-associated RyR2-I4855M+/- mutation. Histological analysis, echocardiography, ECG recording, and intact heart Ca2+ imaging were performed to characterize the structural and functional consequences of the RyR2-I4855M+/- mutation. RESULTS As in humans, RyR2-I4855M+/- mice displayed LVNC characterized by cardiac hypertrabeculation and noncompaction. RyR2-I4855M+/- mice were highly susceptible to electrical stimulation-induced ventricular arrhythmias but protected from stress-induced ventricular arrhythmias. Unexpectedly, the RyR2-I4855M+/- mutation increased the peak Ca2+ transient but did not alter the L-type Ca2+ current, suggesting an increase in Ca2+-induced Ca2+ release gain. The RyR2-I4855M+/- mutation abolished sarcoplasmic reticulum store overload-induced Ca2+ release or Ca2+ leak, elevated sarcoplasmic reticulum Ca2+ load, prolonged Ca2+ transient decay, and elevated end-diastolic Ca2+ level upon rapid pacing. Immunoblotting revealed increased level of phosphorylated CaMKII (Ca2+-calmodulin dependent protein kinases II) but unchanged levels of CaMKII, calcineurin, and other Ca2+ handling proteins in the RyR2-I4855M+/- mutant compared with wild type. CONCLUSIONS The RyR2-I4855M+/- mutant mice represent the first RyR2-associated LVNC animal model that recapitulates the CRDS-LVNC overlapping phenotype in humans. The RyR2-I4855M+/- mutation increases the peak Ca2+ transient by increasing the Ca2+-induced Ca2+ release gain and the end-diastolic Ca2+ level by prolonging Ca2+ transient decay. Our data suggest that the increased peak-systolic and end-diastolic Ca2+ levels may underlie RyR2-associated LVNC.
Collapse
Affiliation(s)
- Mingke Ni
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Yanhui Li
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Jinhong Wei
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
- School of Medicine, Northwest University, Xi 'an, China (J.W.)
| | - Zhenpeng Song
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Hui Wang
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Yong-Xiang Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Darrell Belke
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - John Paul Estillore
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Ruiwu Wang
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Alexander Vallmitjana
- Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain (A.V., R.B.)
| | - Raul Benitez
- Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain (A.V., R.B.)
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain (R.B.)
| | - Leif Hove-Madsen
- Biomedical Research Institute Barcelona IIBB-CSIC, IIB Sant Pau and CIBERCV, Hospital de Sant Pau, Barcelona, Spain (L.H.-M.)
| | - Wei Feng
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla (W.F., J.C.)
| | - Ju Chen
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla (W.F., J.C.)
| | - Thomas M Roston
- Division of Pediatric Cardiology, Department of Pediatrics (T.M.R., S.S.), University of British Columbia, Vancouver, Canada
| | - Shubhayan Sanatani
- Division of Pediatric Cardiology, Department of Pediatrics (T.M.R., S.S.), University of British Columbia, Vancouver, Canada
| | - Anna Lehman
- Department of Medical Genetics (A.L.), University of British Columbia, Vancouver, Canada
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| |
Collapse
|
7
|
Watanabe H, Sucov HM. Cardiomyocyte proliferation by calcineurin inhibition. NATURE CARDIOVASCULAR RESEARCH 2022; 1:599-600. [PMID: 39196241 DOI: 10.1038/s44161-022-00100-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Hirofumi Watanabe
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
8
|
Kamareddine L, Ghantous CM, Allouch S, Al-Ashmar SA, Anlar G, Kannan S, Djouhri L, Korashy HM, Agouni A, Zeidan A. Between Inflammation and Autophagy: The Role of Leptin-Adiponectin Axis in Cardiac Remodeling. J Inflamm Res 2021; 14:5349-5365. [PMID: 34703273 PMCID: PMC8528546 DOI: 10.2147/jir.s322231] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/24/2021] [Indexed: 01/05/2023] Open
Abstract
Cardiac remodeling is the process by which the heart adapts to stressful stimuli, such as hypertension and ischemia/reperfusion; it ultimately leads to heart failure upon long-term exposure. Autophagy, a cellular catabolic process that was originally considered as a mechanism of cell death in response to detrimental stimuli, is thought to be one of the main mechanisms that controls cardiac remodeling and induces heart failure. Dysregulation of the adipokines leptin and adiponectin, which plays essential roles in lipid and glucose metabolism, and in the pathophysiology of the neuroendocrine and cardiovascular systems, has been shown to affect the autophagic response in the heart and to contribute to accelerate cardiac remodeling. The obesity-associated protein leptin is a pro-inflammatory, tumor-promoting adipocytokine whose elevated levels in obesity are associated with acute cardiovascular events, and obesity-related hypertension. Adiponectin exerts anti-inflammatory and anti-tumor effects, and its reduced levels in obesity correlate with the pathogenesis of obesity-associated cardiovascular diseases. Leptin- and adiponectin-induced changes in autophagic flux have been linked to cardiac remodeling and heart failure. In this review, we describe the different molecular mechanisms of hyperleptinemia- and hypoadiponectinemia-mediated pathogenesis of cardiac remodeling and the involvement of autophagy in this process. A better understanding of the roles of leptin, adiponectin, and autophagy in cardiac functions and remodeling, and the exact signal transduction pathways by which they contribute to cardiac diseases may well lead to discovery of new therapeutic agents for the treatment of cardiovascular remodeling.
Collapse
Affiliation(s)
- Layla Kamareddine
- Department Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Crystal M Ghantous
- Department of Nursing and Health Sciences, Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Keserwan, Lebanon
| | - Soumaya Allouch
- Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Sarah A Al-Ashmar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
- Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Gulsen Anlar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
- Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Surya Kannan
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
- Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Laiche Djouhri
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
- Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Hesham M Korashy
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Abdelali Agouni
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Asad Zeidan
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
- Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
9
|
Cai X, Tian Y, Wu Y, Bonner MY, Zhuo X, Yuan Z. An Optimized Model of Hypertrophic Preconditioning Confers Cardioprotection in the Mouse. J Surg Res 2021; 264:544-552. [PMID: 33864962 DOI: 10.1016/j.jss.2020.11.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/15/2020] [Accepted: 11/01/2020] [Indexed: 10/21/2022]
Abstract
BACKGROUND Conventional models of hypertrophic preconditioning (C-HP) can be established surgically through transverse aortic constriction (TAC) → deconstriction (De-TAC) → reconstriction (Re-TAC) characterized by dynamic afterload while it exerts technical difficulty on operators and poses high mortality during perioperative period in mice. We aimed to introduce an optimized method for obtaining a hypertrophic preconditioning (O-HP) model for further study on cardiac hypertrophy. METHODS Ninety mice were divided into four groups: sham, TAC, C-HP, and O-HP. The sham group was exerted on three-time thoracotomies. The TAC group experienced twice thoracotomies and one TAC operation. C-HP and O-HP groups were given TAC, De-TAC, and Re-TAC operation at day 0, day 3, and day 7 in conventional and optimized method, respectively. We optimized the operating procedure in O-HP mice compared with the C-HP group by (1) leaving a ∼3-cm suture fixed in the subcutaneous layer after aortic constriction in TAC surgery (2) using two small forceps to untie the constriction knot instead of cutting it in the De-TAC operation. Ultrasound biomicroscopy was used for hemodynamics and cardiac function detection. Four weeks after the third surgery, all mice were sacrificed and pathology was analyzed among four groups. RESULTS Four weeks after Re-TAC, the survival of O-HP mice was 63.3% while that of C-HP was 26.7%. Ultrasound biomicroscopy showed a successful establishment of HP models. C-HP and O-HP mice had improved cardiac structure and function indicated by left ventricular end-systolic diameter, left ventricular end-systolic posterior wall thickness, left ventricular ejection fraction, and left ventricular fractional shortening than the TAC group. Pathological analysis showed O-HP as well as C-HP had less hypertrophy than the TAC mice. CONCLUSIONS Our results provide a rapid, safe, efficient, and reproducible method for optimized establishment of the HP model, which will facilitate studies for early intervention and prevention of left ventricular hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xiaojie Cai
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an Shaanxi, China
| | - Yuling Tian
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an Shaanxi, China
| | - Yue Wu
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an Shaanxi, China
| | - Michael Y Bonner
- Division of Medical Inflammation Research, Department of Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Xiaozhen Zhuo
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an Shaanxi, China.
| | - Zuyi Yuan
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an Shaanxi, China.
| |
Collapse
|
10
|
Florczyk-Soluch U, Polak K, Dulak J. The multifaceted view of heart problem in Duchenne muscular dystrophy. Cell Mol Life Sci 2021; 78:5447-5468. [PMID: 34091693 PMCID: PMC8257522 DOI: 10.1007/s00018-021-03862-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/29/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022]
Abstract
Dystrophin is a large protein serving as local scaffolding repetitively bridging cytoskeleton and the outside of striated muscle cell. As such dystrophin is a critical brick primarily in dystrophin-associated protein complex (DAGC) and in a larger submembranous unit, costamere. Accordingly, the lack of functional dystrophin laying at the root of Duchenne muscular dystrophy (DMD) drives sarcolemma instability. From this point on, the cascade inevitably leading to the death of myocyte begins. In cardiomyocytes, intracellular calcium overload and related mitochondrial-mediated cell death mainly contribute to myocardial dysfunction and dilation while other protein dysregulation and/or mislocalization may affect electrical conduction system and favor arrhythmogenesis. Although clinically DMD manifests as progressive muscle weakness and skeletal muscle symptoms define characteristic of DMD, it is the heart problem the biggest challenge that most often develop in the form of dilated cardiomyopathy (DCM). Current standards of treatment and recent progress in respiratory care, introduced in most settings in the 1990s, have improved quality of life and median life expectancy to 4th decade of patient's age. At the same time, cardiac causes of death related to DMD increases. Despite preventive and palliative cardiac treatments available, the prognoses remain poor. Direct therapeutic targeting of dystrophin deficiency is critical, however, hindered by the large size of the dystrophin cDNA and/or stochastic, often extensive genetic changes in DMD gene. The correlation between cardiac involvement and mutations affecting specific dystrophin isoforms, may provide a mutation-specific cardiac management and novel therapeutic approaches for patients with CM. Nonetheless, the successful cardiac treatment poses a big challenge and may require combined therapy to combat dystrophin deficiency and its after-effects (critical in DMD pathogenesis). This review locates the multifaceted heart problem in the course of DMD, balancing the insights into basic science, translational efforts and clinical manifestation of dystrophic heart disease.
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
11
|
Wang EY, Kuzmanov U, Smith JB, Dou W, Rafatian N, Lai BFL, Lu RXZ, Wu Q, Yazbeck J, Zhang XO, Sun Y, Gramolini A, Radisic M. An organ-on-a-chip model for pre-clinical drug evaluation in progressive non-genetic cardiomyopathy. J Mol Cell Cardiol 2021; 160:97-110. [PMID: 34216608 DOI: 10.1016/j.yjmcc.2021.06.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 01/08/2023]
Abstract
Angiotensin II (Ang II) presents a critical mediator in various pathological conditions such as non-genetic cardiomyopathy. Osmotic pump infusion in rodents is a commonly used approach to model cardiomyopathy associated with Ang II. However, profound differences in electrophysiology and pharmacokinetics between rodent and human cardiomyocytes may limit predictability of animal-based experiments. This study investigates the application of an Organ-on-a-chip (OOC) system in modeling Ang II-induced progressive cardiomyopathy. The disease model is constructed to recapitulate myocardial response to Ang II in a temporal manner. The long-term tissue cultivation and non-invasive functional readouts enable monitoring of both acute and chronic cardiac responses to Ang II stimulation. Along with mapping of cytokine secretion and proteomic profiles, this model presents an opportunity to quantitatively measure the dynamic pathological changes that could not be otherwise identified in animals. Further, we present this model as a testbed to evaluate compounds that target Ang II-induced cardiac remodeling. Through assessing the effects of losartan, relaxin, and saracatinib, the drug screening data implicated multifaceted cardioprotective effects of relaxin in restoring contractile function and reducing fibrotic remodeling. Overall, this study provides a controllable platform where cardiac activities can be explicitly observed and tested over the pathological process. The facile and high-content screening can facilitate the evaluation of potential drug candidates in the pre-clinical stage.
Collapse
Affiliation(s)
- Erika Yan Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Uros Kuzmanov
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jacob B Smith
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Naimeh Rafatian
- Toronto General Research Institute, Toronto, Ontario M5G 2C4, Canada
| | - Benjamin Fook Lun Lai
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Rick Xing Ze Lu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Qinghua Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Joshua Yazbeck
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Xiao-Ou Zhang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Anthony Gramolini
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada; Toronto General Research Institute, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
12
|
Kolodziej F, O’Halloran KD. Re-Evaluating the Oxidative Phenotype: Can Endurance Exercise Save the Western World? Antioxidants (Basel) 2021; 10:609. [PMID: 33921022 PMCID: PMC8071436 DOI: 10.3390/antiox10040609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 01/16/2023] Open
Abstract
Mitochondria are popularly called the "powerhouses" of the cell. They promote energy metabolism through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation, which in contrast to cytosolic glycolysis are oxygen-dependent and significantly more substrate efficient. That is, mitochondrial metabolism provides substantially more cellular energy currency (ATP) per macronutrient metabolised. Enhancement of mitochondrial density and metabolism are associated with endurance training, which allows for the attainment of high relative VO2 max values. However, the sedentary lifestyle and diet currently predominant in the Western world lead to mitochondrial dysfunction. Underdeveloped mitochondrial metabolism leads to nutrient-induced reducing pressure caused by energy surplus, as reduced nicotinamide adenine dinucleotide (NADH)-mediated high electron flow at rest leads to "electron leak" and a chronic generation of superoxide radicals (O2-). Chronic overload of these reactive oxygen species (ROS) damages cell components such as DNA, cell membranes, and proteins. Counterintuitively, transiently generated ROS during exercise contributes to adaptive reduction-oxidation (REDOX) signalling through the process of cellular hormesis or "oxidative eustress" defined by Helmut Sies. However, the unaccustomed, chronic oxidative stress is central to the leading causes of mortality in the 21st century-metabolic syndrome and the associated cardiovascular comorbidities. The endurance exercise training that improves mitochondrial capacity and the protective antioxidant cellular system emerges as a universal intervention for mitochondrial dysfunction and resultant comorbidities. Furthermore, exercise might also be a solution to prevent ageing-related degenerative diseases, which are caused by impaired mitochondrial recycling. This review aims to break down the metabolic components of exercise and how they translate to athletic versus metabolically diseased phenotypes. We outline a reciprocal relationship between oxidative metabolism and inflammation, as well as hypoxia. We highlight the importance of oxidative stress for metabolic and antioxidant adaptation. We discuss the relevance of lactate as an indicator of critical exercise intensity, and inferring from its relationship with hypoxia, we suggest the most appropriate mode of exercise for the case of a lost oxidative identity in metabolically inflexible patients. Finally, we propose a reciprocal signalling model that establishes a healthy balance between the glycolytic/proliferative and oxidative/prolonged-ageing phenotypes. This model is malleable to adaptation with oxidative stress in exercise but is also susceptible to maladaptation associated with chronic oxidative stress in disease. Furthermore, mutations of components involved in the transcriptional regulatory mechanisms of mitochondrial metabolism may lead to the development of a cancerous phenotype, which progressively presents as one of the main causes of death, alongside the metabolic syndrome.
Collapse
Affiliation(s)
- Filip Kolodziej
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, T12 XF62 Cork, Ireland;
| | | |
Collapse
|
13
|
Shimoyama H, Yonezawa Y. Atomistic detailed free-energy landscape of intrinsically disordered protein studied by multi-scale divide-and-conquer molecular dynamics simulation. J Comput Chem 2021; 42:19-26. [PMID: 33030249 DOI: 10.1002/jcc.26429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/06/2020] [Accepted: 09/10/2020] [Indexed: 11/08/2022]
Abstract
Calcineurin (CaN) is a eukaryotic serine/threonine protein phosphatase activated by both Ca2+ and calmodulin (CaM), including intrinsically disordered region (IDR). The region undergoes folding into an α-helix form in the presence Ca2+ -loaded CaM. To sample the ordered structure of the IDR by conventional all atom model (AAM) molecular dynamics (MD) simulation, the IDR and Ca2+ -loaded CaM must be simultaneously treated. However, it is time-consuming task because the coupled folding and binding should include repeated binding and dissociation. Then, in this study, we propose novel multi-scale divide-and-conquer MD (MSDC-MD), which combines AAM-MD and coarse-grained model MD (CGM-MD). To speed up the conformation sampling, MSDC-MD simulation first treats the IDR by CGM to sample conformations from wide conformation space; then, multiple AAM-MD in a limited area is initiated using the resultant CGM conformation, which is reconstructed by homology modeling method. To investigate performance, we sampled the ordered conformation of the IDR using MSDC-MD; the root-mean-square distance (RMSD) with respect to the experimental structure was 2.23 Å.
Collapse
Affiliation(s)
| | - Yasushige Yonezawa
- High Pressure Protein Research Center, Institute of Advanced Technology, Kindai University, Wakayama, Japan
| |
Collapse
|
14
|
Martens MD, Field JT, Seshadri N, Day C, Chapman D, Keijzer R, Doucette CA, Hatch GM, West AR, Ivanco TL, Gordon JW. Misoprostol attenuates neonatal cardiomyocyte proliferation through Bnip3, perinuclear calcium signaling, and inhibition of glycolysis. J Mol Cell Cardiol 2020; 146:19-31. [DOI: 10.1016/j.yjmcc.2020.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/03/2020] [Accepted: 06/27/2020] [Indexed: 02/02/2023]
|
15
|
Valkov N, Das S. Y RNAs: Biogenesis, Function and Implications for the Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:327-342. [PMID: 32285422 DOI: 10.1007/978-981-15-1671-9_20] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In recent years, progress in the field of high-throughput sequencing technology and its application to a wide variety of biological specimens has greatly advanced the discovery and cataloging of a diverse set of non-coding RNAs (ncRNAs) that have been found to have unexpected biological functions. Y RNAs are an emerging class of highly conserved, small ncRNAs. There is a growing number of reports in the literature demonstrating that Y RNAs and their fragments are not just random degradation products but are themselves bioactive molecules. This review will outline what is currently known about Y RNA including biogenesis, structure and functional roles. In addition, we will provide an overview of studies reporting the presence and functions attributed to Y RNAs in the cardiovascular system.
Collapse
Affiliation(s)
- Nedyalka Valkov
- Cardiovascular Research Center of Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Saumya Das
- Cardiovascular Research Center of Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Pitoulis FG, Terracciano CM. Heart Plasticity in Response to Pressure- and Volume-Overload: A Review of Findings in Compensated and Decompensated Phenotypes. Front Physiol 2020; 11:92. [PMID: 32116796 PMCID: PMC7031419 DOI: 10.3389/fphys.2020.00092] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
The adult human heart has an exceptional ability to alter its phenotype to adapt to changes in environmental demand. This response involves metabolic, mechanical, electrical, and structural alterations, and is known as cardiac plasticity. Understanding the drivers of cardiac plasticity is essential for development of therapeutic agents. This is particularly important in contemporary cardiology, which uses treatments with peripheral effects (e.g., on kidneys, adrenal glands). This review focuses on the effects of different hemodynamic loads on myocardial phenotype. We examine mechanical scenarios of pressure- and volume overload, from the initial insult, to compensated, and ultimately decompensated stage. We discuss how different hemodynamic conditions occur and are underlined by distinct phenotypic and molecular changes. We complete the review by exploring how current basic cardiac research should leverage available cardiac models to study mechanical load in its different presentations.
Collapse
|
17
|
Chen LC, Shibu MA, Liu CJ, Han CK, Ju DT, Chen PY, Viswanadha VP, Lai CH, Kuo WW, Huang CY. ERK1/2 mediates the lipopolysaccharide-induced upregulation of FGF-2, uPA, MMP-2, MMP-9 and cellular migration in cardiac fibroblasts. Chem Biol Interact 2019; 306:62-69. [PMID: 30980805 DOI: 10.1016/j.cbi.2019.04.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 03/08/2019] [Accepted: 04/08/2019] [Indexed: 12/31/2022]
Abstract
Myocardial fibrosis is a critical event during septic shock. Upregulation in the fibrosis signaling cascade proteins such as fibroblast growth factor (FGF), urokinase plasminogen activator (uPA), tissue plasminogen activator (tPA) and activation of matrix metalloproteinases (MMPs) are widely associated with the development of myocardial infarction, dilated cardiomyopathy, cardiac fibrosis and heart failure. However, evidences suggest that the common upstream mediators of fibrosis cascade play little role in cardiac fibrosis induced by LPS; further, it is unknown if LPS directly triggers the expressions and/or activity of FGF-2, uPA, tPA, MMP-2 and MMP-9 in cardiac fibroblasts. In the present study, we treated primary cultures of cardiac fibroblasts with LPS to explore whether LPS upregulates FGF-2, uPA, tPA, MMP-2, MMP-9 and enhance cellular migration. Further the precise molecular and cellular mechanisms behind these LPS induced responses were identified. Inhibition assays on MAPKs using U0126 (ERK1/2 inhibitor), SB203580 (p38 MAPK inhibitor), SP600125 (JNK1/2 inhibitor), CsA (calcineurin inhibitor) and QNZ (NFκB inhibitor) show that LPS-induced upregulation of FGF-2, uPA, MMP-2 and MMP-9 in cardiac fibroblasts was mediated through ERK1/2 signaling. Collectively, our results provide a link between LPS-induced cardiac dysfunction and ERK1/2 signaling pathway and thereby implies ERK1/2 as a possible target to regulate LPS induced upregulation of FGF-2, uPA, MMP-2, MMP-9 and cellular migration in cardiac fibroblasts.
Collapse
Affiliation(s)
- Liang-Chi Chen
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Marthandam Asokan Shibu
- Medical Research Center for Exosome and Mitochondria Related Diseases, China Medical University and Hospital, Taichung, Taiwan
| | - Chung-Jung Liu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chien-Kuo Han
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Da-Tong Ju
- Department of Neurological Surgery,Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Pei-Yu Chen
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | | | - Chao-Hung Lai
- Division of Cardiology, Department of Internal Medicine, Taichung Armed Force General Hospital, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Department of Biotechnology, Asia University, Taichung, Taiwan; College of Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan; Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.
| |
Collapse
|
18
|
Embryonic programming of heart disease in response to obesity during pregnancy. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165402. [PMID: 30759362 DOI: 10.1016/j.bbadis.2019.01.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/19/2019] [Accepted: 01/28/2019] [Indexed: 12/20/2022]
Abstract
Obesity during pregnancy programs adult-onset heart disease in the offspring. Clinical studies indicate that exposure to an adverse environment in utero during early, as compared to late, gestation leads to a higher prevalence of adult-onset heart disease. This suggests that the early developing heart is particularly sensitive to an adverse environment. Accordingly, growing evidence from clinical studies and animal models demonstrates that obesity during pregnancy alters the function of the fetal heart, programming a higher risk of cardiovascular disease later in life. Moreover, gene expression patterns and signaling pathways that promote initiation and progression of cardiovascular disease are altered in the hearts in offspring born to obese mothers. However, the mechanisms mediating the long-term effects of an adverse environment in utero on the developing heart leading to adult-onset disease are not clear. Here, we review clinical and experimental evidence documenting the effects of maternal obesity during pregnancy on the fetal and post-natal heart and emphasize on the potential mechanisms of disease programming.
Collapse
|
19
|
Sharma RK, Parameswaran S. Calmodulin-binding proteins: A journey of 40 years. Cell Calcium 2018; 75:89-100. [PMID: 30205293 DOI: 10.1016/j.ceca.2018.09.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/02/2018] [Indexed: 01/04/2023]
Abstract
The proteins which bind to calmodulin in a Ca2+-dependent and reversible manner are known as calmodulin-binding proteins. These proteins are involved in a multitude of processes in which Ca2+ and calmodulin play crucial roles. Our group elucidated the mechanism and importance of these proteins in normal and diseased conditions. Various calmodulin-binding proteins were discovered and purified from bovine tissue including a heat stable calmodulin-binding protein 70, calmodulin-dependent protein kinase VI and a high molecular weight calmodulin-binding protein (HMWCaMBP). We observed a complex interplay occurs between these and other Ca2+ and calmodulin-binding proteins during cardiac ischemia and reperfusion. Purified cardiac HMWCaMBP is a homolog form of calpastatin and an inhibitor of the Ca2+-activated cysteine proteases, calpains and therefore can have cardioprotective role in ischemic conditions. Calcineurin is a Ca2+ and calmodulin-dependent serine/threonine protein phosphatase showed increased phosphatase activity in ischemic heart through its direct interaction with Hsp70 and expression of calcineurin following ischemia suggests self-repair and favorable survival outcomes. Calcineurin was also found to be present in other tissues including the eye; where its expression and calcineurin phosphatase activity varied. In neurons, calcineurin may play a key role in initiating apoptosis-related pathways especially in epilepsy. In colorectal cancer we demonstrated high calcineurin phosphatase activity and simultaneous overexpression of calcineurin. The impact of calcineurin signaling on neuronal apoptosis in epilepsy and its use as a diagnostic marker for colorectal cancer requires in-depth study.
Collapse
Affiliation(s)
- Rajendra K Sharma
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon S7N 5E5, Canada.
| | - Sreejit Parameswaran
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon S7N 5E5, Canada
| |
Collapse
|
20
|
NPY Impairs Cell Viability and Mitochondrial Membrane Potential Through Ca2+ and p38 Signaling Pathways in Neonatal Rat Cardiomyocytes. J Cardiovasc Pharmacol 2018; 70:52-59. [PMID: 28437279 DOI: 10.1097/fjc.0000000000000493] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
NPY is involved in stress cardiomyopathy. However, the associated mechanism for NPY-induced stress cardiomyopathy remains unclear. In this study, we aimed to explore potential cell signaling pathways that are related to NPY-mediated cell viability in neonatal rat cardiomyocytes. We found that NPY induced cell viability suppression in cultured cardiomyocytes in a dose-dependent manner. After NPY treatment, expression of CaN and p-CAMKII increased significantly, and phosphorylation of p38 but not ERK and JNK was changed. Moreover, NPY treatment significantly increased PGC-1α (the key factor of mitochondrial biogenesis and energy metabolism) expression but decreased mitochondrial membrane potential in cultured cardiomyocytes. More importantly, the blockage of CaN, CAMKII, and p38 signaling pathways by their inhibitors could rescue the reduced cell viability and mitochondrial membrane potential in NPY-treated cardiomyocytes. Collectively, our data demonstrated that NPY mediated cell viability and mitochondrial membrane potential in cardiomyocytes through CaN, CAMKII, and p38 signaling pathways.
Collapse
|
21
|
Abstract
Heart failure places an enormous burden on health and economic systems worldwide. It is a complex disease that is profoundly influenced by both genetic and environmental factors. Neither the molecular mechanisms underlying heart failure nor effective prevention strategies are fully understood. Fortunately, relevant aspects of human heart failure can be experimentally studied in tractable model animals, including the fruit fly, Drosophila, allowing the in vivo application of powerful and sophisticated molecular genetic and physiological approaches. Heart failure in Drosophila, as in humans, can be classified into dilated cardiomyopathies and hypertrophic cardiomyopathies. Critically, many genes and cellular pathways directing heart development and function are evolutionarily conserved from Drosophila to humans. Studies of molecular mechanisms linking aging with heart failure have revealed that genes involved in aging-associated energy homeostasis and oxidative stress resistance influence cardiac dysfunction through perturbation of IGF and TOR pathways. Importantly, ion channel proteins, cytoskeletal proteins, and integrins implicated in aging of the mammalian heart have been shown to play significant roles in heart failure. A number of genes previously described having roles in development of the Drosophila heart, such as genes involved in Wnt signaling pathways, have recently been shown to play important roles in the adult fly heart. Moreover, the fly model presents opportunities for innovative studies that cannot currently be pursued in the mammalian heart because of technical limitations. In this review, we discuss progress in our understanding of genes, proteins, and molecular mechanisms that affect the Drosophila adult heart and heart failure.
Collapse
Affiliation(s)
- Shasha Zhu
- The Center for Heart Development, Key Lab of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Zhe Han
- Center for Cancer and Immunology Research, Children's National Medical Center, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Yan Luo
- The Center for Heart Development, Key Lab of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Yulin Chen
- The Center for Heart Development, Key Lab of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Qun Zeng
- The Center for Heart Development, Key Lab of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Xiushan Wu
- The Center for Heart Development, Key Lab of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.
| | - Wuzhou Yuan
- The Center for Heart Development, Key Lab of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.
| |
Collapse
|
22
|
Liu T, Zhou HJ, Min W. ASK family in cardiovascular biology and medicine. Adv Biol Regul 2017; 66:54-62. [PMID: 29107568 PMCID: PMC5705453 DOI: 10.1016/j.jbior.2017.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 10/22/2017] [Accepted: 10/23/2017] [Indexed: 01/03/2023]
Abstract
Cardiovascular disease is a major cause of death worldwide. Mitogen-activated protein kinase (MAPK) signal cascades signaling pathways play crucial roles in cardiovascular pathophysiology. Apoptosis signal-regulating kinase (ASK) family members ASK1, ASK2 and ASK3 are the key molecules in MAPK signal cascades and are activated by various stresses. ASK1 is the most extensively studied MAPKKK and is involved in regulation of the cellular functions such as cell survival, proliferation, inflammation and apoptosis. The current review focuses on the relationship between ASK1 and cardiovascular disease, while exploring the novel therapeutic strategies for cardiovascular disease involved in the ASK1 signal pathway.
Collapse
Affiliation(s)
- Tingting Liu
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Huanjiao Jenny Zhou
- Department of Pathology and the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Wang Min
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Department of Pathology and the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
23
|
Zarndt R, Walls SM, Ocorr K, Bodmer R. Reduced Cardiac Calcineurin Expression Mimics Long-Term Hypoxia-Induced Heart Defects in Drosophila. CIRCULATION. CARDIOVASCULAR GENETICS 2017; 10:e001706. [PMID: 28986453 PMCID: PMC5669044 DOI: 10.1161/circgenetics.117.001706] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 07/28/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Hypoxia is often associated with cardiopulmonary diseases, which represent some of the leading causes of mortality worldwide. Long-term hypoxia exposures, whether from disease or environmental condition, can cause cardiomyopathy and lead to heart failure. Indeed, hypoxia-induced heart failure is a hallmark feature of chronic mountain sickness in maladapted populations living at high altitude. In a previously established Drosophila heart model for long-term hypoxia exposure, we found that hypoxia caused heart dysfunction. Calcineurin is known to be critical in cardiac hypertrophy under normoxia, but its role in the heart under hypoxia is poorly understood. METHODS AND RESULTS In the present study, we explore the function of calcineurin, a gene candidate we found downregulated in the Drosophila heart after lifetime and multigenerational hypoxia exposure. We examined the roles of 2 homologs of Calcineurin A, CanA14F, and Pp2B in the Drosophila cardiac response to long-term hypoxia. We found that knockdown of these calcineurin catalytic subunits caused cardiac restriction under normoxia that are further aggravated under hypoxia. Conversely, cardiac overexpression of Pp2B under hypoxia was lethal, suggesting that a hypertrophic signal in the presence of insufficient oxygen supply is deleterious. CONCLUSIONS Our results suggest a key role for calcineurin in cardiac remodeling during long-term hypoxia with implications for diseases of chronic hypoxia, and it likely contributes to mechanisms underlying these disease states.
Collapse
Affiliation(s)
- Rachel Zarndt
- From the Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute (R.Z., S.M.W., K.O., R.B.) and Biomedical Sciences Graduate Program, School of Medicine, University of California at San Diego (R.Z.), La Jolla, CA
| | - Stanley M Walls
- From the Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute (R.Z., S.M.W., K.O., R.B.) and Biomedical Sciences Graduate Program, School of Medicine, University of California at San Diego (R.Z.), La Jolla, CA
| | - Karen Ocorr
- From the Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute (R.Z., S.M.W., K.O., R.B.) and Biomedical Sciences Graduate Program, School of Medicine, University of California at San Diego (R.Z.), La Jolla, CA.
| | - Rolf Bodmer
- From the Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute (R.Z., S.M.W., K.O., R.B.) and Biomedical Sciences Graduate Program, School of Medicine, University of California at San Diego (R.Z.), La Jolla, CA.
| |
Collapse
|
24
|
Sukumaran A, Chang J, Han M, Mintri S, Khaw BA, Kim J. Iron overload exacerbates age-associated cardiac hypertrophy in a mouse model of hemochromatosis. Sci Rep 2017; 7:5756. [PMID: 28720890 PMCID: PMC5516030 DOI: 10.1038/s41598-017-05810-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/02/2017] [Indexed: 12/12/2022] Open
Abstract
Cardiac damage associated with iron overload is the most common cause of morbidity and mortality in patients with hereditary hemochromatosis, but the precise mechanisms leading to disease progression are largely unexplored. Here we investigated the effects of iron overload and age on cardiac hypertrophy using 1-, 5- and 12-month old Hfe-deficient mice, an animal model of hemochromatosis in humans. Cardiac iron levels increased progressively with age, which was exacerbated in Hfe-deficient mice. The heart/body weight ratios were greater in Hfe-deficient mice at 5- and 12-month old, compared with their age-matched wild-type controls. Cardiac hypertrophy in 12-month old Hfe-deficient mice was consistent with decreased alpha myosin and increased beta myosin heavy chains, suggesting an alpha-to-beta conversion with age. This was accompanied by cardiac fibrosis and up-regulation of NFAT-c2, reflecting increased calcineurin/NFAT signaling in myocyte hypertrophy. Moreover, there was an age-dependent increase in the cardiac isoprostane levels in Hfe-deficient mice, indicating elevated oxidative stress. Also, rats fed high-iron diet demonstrated increased heart-to-body weight ratios, alpha myosin heavy chain and cardiac isoprostane levels, suggesting that iron overload promotes oxidative stress and cardiac hypertrophy. Our findings provide a molecular basis for the progression of age-dependent cardiac stress exacerbated by iron overload hemochromatosis.
Collapse
Affiliation(s)
- Abitha Sukumaran
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - JuOae Chang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Murui Han
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Shrutika Mintri
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Ban-An Khaw
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA.
| |
Collapse
|
25
|
Li L, Li J, Drum BM, Chen Y, Yin H, Guo X, Luckey SW, Gilbert ML, McKnight GS, Scott JD, Santana LF, Liu Q. Loss of AKAP150 promotes pathological remodelling and heart failure propensity by disrupting calcium cycling and contractile reserve. Cardiovasc Res 2016; 113:147-159. [PMID: 27856611 DOI: 10.1093/cvr/cvw221] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/15/2016] [Accepted: 10/11/2016] [Indexed: 01/18/2023] Open
Abstract
AIMS Impaired Ca2 + cycling and myocyte contractility are a hallmark of heart failure triggered by pathological stress such as hemodynamic overload. The A-Kinase anchoring protein AKAP150 has been shown to coordinate key aspects of adrenergic regulation of Ca2+ cycling and excitation-contraction in cardiomyocytes. However, the role of the AKAP150 signalling complexes in the pathogenesis of heart failure has not been investigated. METHODS AND RESULTS Here we examined how AKAP150 signalling complexes impact Ca2+ cycling, myocyte contractility, and heart failure susceptibility following pathological stress. We detected a significant reduction of AKAP150 expression in the failing mouse heart induced by pressure overload. Importantly, cardiac-specific AKAP150 knockout mice were predisposed to develop dilated cardiomyopathy with severe cardiac dysfunction and fibrosis after pressure overload. Loss of AKAP150 also promoted pathological remodelling and heart failure progression following myocardial infarction. However, ablation of AKAP150 did not affect calcineurin-nuclear factor of activated T cells signalling in cardiomyocytes or pressure overload- or agonist-induced cardiac hypertrophy. Immunoprecipitation studies showed that AKAP150 was associated with SERCA2, phospholamban, and ryanodine receptor-2, providing a targeted control of sarcoplasmic reticulum Ca2+ regulatory proteins. Mechanistically, loss of AKAP150 led to impaired Ca2+ cycling and reduced myocyte contractility reserve following adrenergic stimulation or pressure overload. CONCLUSIONS These findings define a critical role for AKAP150 in regulating Ca2+ cycling and myocardial ionotropy following pathological stress, suggesting the AKAP150 signalling pathway may serve as a novel therapeutic target for heart failure.
Collapse
Affiliation(s)
- Lei Li
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Jing Li
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Benjamin M Drum
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Yi Chen
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Haifeng Yin
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Xiaoyun Guo
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Stephen W Luckey
- Department of Biology, Seattle University, 901 12th Ave., Seattle, WA 98122, USA
| | - Merle L Gilbert
- Department of Pharmacology, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - G Stanley McKnight
- Department of Pharmacology, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - John D Scott
- Department of Pharmacology, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA.,Howard Hughes Medical Institute, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - L Fernando Santana
- Deparment of Physiology & Membrane Biology, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Qinghang Liu
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA;
| |
Collapse
|
26
|
Deng W, Ednie AR, Qi J, Bennett ES. Aberrant sialylation causes dilated cardiomyopathy and stress-induced heart failure. Basic Res Cardiol 2016; 111:57. [PMID: 27506532 DOI: 10.1007/s00395-016-0574-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 07/05/2016] [Accepted: 07/25/2016] [Indexed: 12/19/2022]
Abstract
Dilated cardiomyopathy (DCM), the third most common cause of heart failure, is often associated with arrhythmias and sudden cardiac death if not controlled. The majority of DCM is of unknown etiology. Protein sialylation is altered in human DCM, with responsible mechanisms not yet described. Here we sought to investigate the impact of clinically relevant changes in sialylation on cardiac function using a novel model for altered glycoprotein sialylation that leads to DCM and to chronic stress-induced heart failure (HF), deletion of the sialyltransferase, ST3Gal4. We previously reported that 12- to 20-week-old ST3Gal4 (-/-) mice showed aberrant cardiac voltage-gated ion channel sialylation and gating that contribute to a pro-arrhythmogenic phenotype. Here, echocardiography supported by histology revealed modest dilated and thinner-walled left ventricles without increased fibrosis in ST3Gal4 (-/-) mice starting at 1 year of age. Cardiac calcineurin expression in younger (16-20 weeks old) ST3Gal4 (-/-) hearts was significantly reduced compared to WT. Transverse aortic constriction (TAC) was used as a chronic stressor on the younger mice to determine whether the ability to compensate against a pathologic insult is compromised in the ST3Gal4 (-/-) heart, as suggested by previous reports describing the functional implications of reduced cardiac calcineurin levels. TAC'd ST3Gal4 (-/-) mice presented with significantly reduced systolic function and ventricular dilation that deteriorated into congestive HF within 6 weeks post-surgery, while constricted WT hearts remained well-adapted throughout (ejection fraction, ST3Gal4 (-/-) = 34 ± 5.2 %; WT = 53.8 ± 7.4 %; p < 0.05). Thus, a novel, sialo-dependent model for DCM/HF is described in which clinically relevant reduced sialylation results in increased arrhythmogenicity and reduced cardiac calcineurin levels that precede cardiomyopathy and TAC-induced HF, suggesting a causal link among aberrant sialylation, chronic arrhythmia, reduced calcineurin levels, DCM in the absence of a pathologic stimulus, and stress-induced HF.
Collapse
Affiliation(s)
- Wei Deng
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, MDC 8, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612-4799, USA
| | - Andrew R Ednie
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, MDC 8, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612-4799, USA
| | - Jianyong Qi
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, MDC 8, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612-4799, USA.,Intensive Care Laboratory, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, People's Republic of China
| | - Eric S Bennett
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, MDC 8, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612-4799, USA.
| |
Collapse
|
27
|
Asadi F, Razmi A, Dehpour AR, Shafiei M. Tropisetron inhibits high glucose-induced calcineurin/NFAT hypertrophic pathway in H9c2 myocardial cells. ACTA ACUST UNITED AC 2016; 68:485-93. [PMID: 26945895 DOI: 10.1111/jphp.12522] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 01/17/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Cardiomyocyte hypertrophy is an important structural feature of diabetic cardiomyopathy. Calcineurin/nuclear factor of activated T-cell (NFAT) pathway plays a central role in the pathogenesis of cardiac hypertrophy. The purpose of this study was to investigate the effects of tropisetron, a novel calcineurin inhibitor, on high glucose (HG)-induced cardiomyocyte hypertrophy and its underlying mechanism. METHODS H9c2 myocardial cells were treated with tropisetron or cyclosporine A 1 h before exposure to HG for 48 h. KEY FINDINGS Exposure to HG resulted in enhanced cell size, protein content and atrial natriuretic peptide (ANP) protein expression. HG significantly increased Ca(2+) level, calcineurin expression and nuclear translocation of NFATc4. Both tropisetron and cyclosporine A markedly prevented the hypertrophic characteristic features, calcineurin overexpression and nuclear localization of NFATc4 while intracellular Ca(2+) was not affected. CONCLUSION Our results showed that tropisetron may have protective effects against HG-induced cardiomyocyte hypertrophy. The mechanism responsible for this beneficial effect seems to be, at least in part, blockade of calcineurin/NFAT signalling pathway.
Collapse
Affiliation(s)
- Firouzeh Asadi
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Razmi
- Medicinal Plants Research Center, Institute of Medicinal Plants ACECR, Karaj, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoumeh Shafiei
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Weng YS, Wang HF, Pai PY, Jong GP, Lai CH, Chung LC, Hsieh DJY, HsuanDay C, Kuo WW, Huang CY. Tanshinone IIA Prevents Leu27IGF-II-Induced Cardiomyocyte Hypertrophy Mediated by Estrogen Receptor and Subsequent Akt Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 43:1567-91. [PMID: 26621443 DOI: 10.1142/s0192415x15500895] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
IGF-IIR plays important roles as a key regulator in myocardial pathological hypertrophy and apoptosis, which subsequently lead to heart failure. Salvia miltiorrhiza Bunge (Danshen) is a traditional Chinese medicinal herb used to treat cardiovascular diseases. Tanshinone IIA is an active compound in Danshen and is structurally similar to 17[Formula: see text]-estradiol (E[Formula: see text]. However, whether tanshinone IIA improves cardiomyocyte survival in pathological hypertrophy through estrogen receptor (ER) regulation remains unclear. This study investigates the role of ER signaling in mediating the protective effects of tanshinone IIA on IGF-IIR-induced myocardial hypertrophy. Leu27IGF-II (IGF-II analog) was shown in this study to specifically activate IGF-IIR expression and ICI 182,780 (ICI), an ER antagonist used to investigate tanshinone IIA estrogenic activity. We demonstrated that tanshinone IIA significantly enhanced Akt phosphorylation through ER activation to inhibit Leu27IGF-II-induced calcineurin expression and subsequent NFATc3 nuclear translocation to suppress myocardial hypertrophy. Tanshinone IIA reduced the cell size and suppressed ANP and BNP, inhibiting antihypertrophic effects induced by Leu27IGF-II. The cardioprotective properties of tanshinone IIA that inhibit Leu27IGF-II-induced cell hypertrophy and promote cell survival were reversed by ICI. Furthermore, ICI significantly reduced phospho-Akt, Ly294002 (PI3K inhibitor), and PI3K siRNA significantly reduced the tanshinone IIA-induced protective effect. The above results suggest that tanshinone IIA inhibited cardiomyocyte hypertrophy, which was mediated through ER, by activating the PI3K/Akt pathway and inhibiting Leu27IGF-II-induced calcineurin and NFATC3. Tanshinone IIA exerted strong estrogenic activity and therefore represented a novel selective ER modulator that inhibits IGF-IIR signaling to block cardiac hypertrophy.
Collapse
Affiliation(s)
- Yueh-Shan Weng
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hsueh-Fang Wang
- Institute of Biomedical Nutrition, Hungkuang University, Taichung, Taiwan
| | - Pei-Ying Pai
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | - Gwo-Ping Jong
- Division of Cardiology, Armed Force Taichung General Hospital, Taichung, Taiwan
| | - Chao-Hung Lai
- Graduate Institute of Aging Medicine, China Medical University, Taichung, Taiwan
- Division of Cardiology, Armed Force Taichung General Hospital, Taichung, Taiwan
| | - Li-Chin Chung
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy & Science, Tainan County, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Cecilia HsuanDay
- Department of Nursing, Mei Ho University, Pingguang Road, Pingtung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
29
|
Viola HM, Hool LC. Role of the cytoskeleton in communication between L-type Ca(2+) channels and mitochondria. Clin Exp Pharmacol Physiol 2015; 40:295-304. [PMID: 23551128 DOI: 10.1111/1440-1681.12072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 02/25/2013] [Accepted: 02/26/2013] [Indexed: 12/15/2022]
Abstract
The L-type Ca(2+) channel is the main route for Ca(2+) entry into cardiac myocytes, which is essential for the maintenance of cardiac excitation and contraction. Alterations in L-type Ca(2+) channel activity and Ca(2+) homeostasis have been implicated in the development of cardiomyopathies. Cardiac excitation and contraction is fuelled by ATP, synthesized predominantly by the mitochondria via the Ca(2+)-dependent process oxidative phosphorylation. Mitochondrial reactive oxygen species (ROS) are by-products of oxidative phosphorylation and are associated with the development of cardiac pathology. The cytoskeleton plays a role in the communication of signals from the plasma membrane to intracellular organelles. There is good evidence that both L-type Ca(2+) channel activity and mitochondrial function can be modulated by changes in the cytoskeletal network. Activation of the L-type Ca(2+) channel can regulate mitochondrial function through cytoskeletal proteins as a result of transmission of movement from the β(2)-subunit of the channel that occurs during activation and inactivation of the channel. An association between cytoskeletal proteins and the mitochondrial voltage-dependent anion channel (VDAC) may play a role in this response. The L-type Ca(2+) channel is the initiator of contraction in cardiac muscle and the VDAC is responsible for regulating mitochondrial ATP/ADP trafficking. This article presents evidence that a functional coupling between L-type Ca(2+) channels and mitochondria may assist in meeting myocardial energy demand on a beat-to-beat basis.
Collapse
Affiliation(s)
- Helena M Viola
- Cardiovascular Electrophysiology Laboratory, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, WA, Australia
| | | |
Collapse
|
30
|
Breitkreuz M, Hamdani N. A change of heart: oxidative stress in governing muscle function? Biophys Rev 2015; 7:321-341. [PMID: 28510229 DOI: 10.1007/s12551-015-0175-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/08/2015] [Indexed: 02/07/2023] Open
Abstract
Redox/cysteine modification of proteins that regulate calcium cycling can affect contraction in striated muscles. Understanding the nature of these modifications would present the possibility of enhancing cardiac function through reversible cysteine modification of proteins, with potential therapeutic value in heart failure with diastolic dysfunction. Both heart failure and muscular dystrophy are characterized by abnormal redox balance and nitrosative stress. Recent evidence supports the synergistic role of oxidative stress and inflammation in the progression of heart failure with preserved ejection fraction, in concert with endothelial dysfunction and impaired nitric oxide-cyclic guanosine monophosphate-protein kinase G signalling via modification of the giant protein titin. Although antioxidant therapeutics in heart failure with diastolic dysfunction have no marked beneficial effects on the outcome of patients, it, however, remains critical to the understanding of the complex interactions of oxidative/nitrosative stress with pro-inflammatory mechanisms, metabolic dysfunction, and the redox modification of proteins characteristic of heart failure. These may highlight novel approaches to therapeutic strategies for heart failure with diastolic dysfunction. In this review, we provide an overview of oxidative stress and its effects on pathophysiological pathways. We describe the molecular mechanisms driving oxidative modification of proteins and subsequent effects on contractile function, and, finally, we discuss potential therapeutic opportunities for heart failure with diastolic dysfunction.
Collapse
Affiliation(s)
- Martin Breitkreuz
- Department of Cardiovascular Physiology, Ruhr University Bochum, MA 3/56, 44780, Bochum, Germany
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, MA 3/56, 44780, Bochum, Germany.
| |
Collapse
|
31
|
Wang KCW, Tosh DN, Zhang S, McMillen IC, Duffield JA, Brooks DA, Morrison JL. IGF-2R-Gαq signaling and cardiac hypertrophy in the low-birth-weight lamb. Am J Physiol Regul Integr Comp Physiol 2015; 308:R627-35. [PMID: 25632020 DOI: 10.1152/ajpregu.00346.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/29/2014] [Indexed: 01/19/2023]
Abstract
The cardiac insulin-like growth factor 2 receptor (IGF-2R) can induce cardiomyocyte hypertrophy in a heterotrimeric G protein receptor-coupled manner involving αq (Gαq) or αs (Gαs). We have previously shown increased left ventricular weight and cardiac IGF-2 and IGF-2R gene expression in low-birth-weight (LBW) compared with average-birth-weight (ABW) lambs. Here, we have investigated the cardiac expression of IGF-2 gene variants, the degree of histone acetylation, and the abundance of proteins in the IGF-2R downstream signaling pathway in ABW and LBW lambs. Samples from the left ventricle of ABW and LBW lambs were collected at 21 days of age. There was increased phospho-CaMKII protein with decreased HDAC 4 abundance in the LBW compared with ABW lambs. There was increased GATA 4 and decreased phospho-troponin I abundance in LBW compared with ABW lambs, which are markers of pathological cardiac hypertrophy and impaired or reduced contractility, respectively. There was increased histone acetylation of H3K9 at IGF-2R promoter and IGF-2R intron 2 differentially methylated region in the LBW lamb. In conclusion, histone acetylation of IGF-2R may lead to increased IGF-2R mRNA expression and subsequently mediate Gαq signaling early in life via CaMKII, resulting in an increased risk of left ventricular hypertrophy and cardiovascular disease in adult life.
Collapse
Affiliation(s)
- Kimberley C W Wang
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia; and
| | - Darran N Tosh
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia; and
| | - Song Zhang
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia; and
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia; and
| | - Jaime A Duffield
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia; and
| | - Doug A Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia; and
| |
Collapse
|
32
|
Aguiar CJ, Rocha-Franco JA, Sousa PA, Santos AK, Ladeira M, Rocha-Resende C, Ladeira LO, Resende RR, Botoni FA, Barrouin Melo M, Lima CX, Carballido JM, Cunha TM, Menezes GB, Guatimosim S, Leite MF. Succinate causes pathological cardiomyocyte hypertrophy through GPR91 activation. Cell Commun Signal 2014; 12:78. [PMID: 25539979 PMCID: PMC4296677 DOI: 10.1186/s12964-014-0078-2] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 11/28/2014] [Indexed: 12/28/2022] Open
Abstract
Background Succinate is an intermediate of the citric acid cycle as well as an extracellular circulating molecule, whose receptor, G protein-coupled receptor-91 (GPR91), was recently identified and characterized in several tissues, including heart. Because some pathological conditions such as ischemia increase succinate blood levels, we investigated the role of this metabolite during a heart ischemic event, using human and rodent models. Results We found that succinate causes cardiac hypertrophy in a GPR91 dependent manner. GPR91 activation triggers the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), the expression of calcium/calmodulin dependent protein kinase IIδ (CaMKIIδ) and the translocation of histone deacetylase 5 (HDAC5) into the cytoplasm, which are hypertrophic-signaling events. Furthermore, we found that serum levels of succinate are increased in patients with cardiac hypertrophy associated with acute and chronic ischemic diseases. Conclusions These results show for the first time that succinate plays an important role in cardiomyocyte hypertrophy through GPR91 activation, and extend our understanding of how ischemia can induce hypertrophic cardiomyopathy. Electronic supplementary material The online version of this article (doi:10.1186/s12964-014-0078-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carla J Aguiar
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - João A Rocha-Franco
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Pedro A Sousa
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Anderson K Santos
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Marina Ladeira
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Cibele Rocha-Resende
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Luiz O Ladeira
- Department of Physics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Fernando A Botoni
- Department of Medicine, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Marcos Barrouin Melo
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Cristiano X Lima
- Department of Medicine, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - José M Carballido
- Novartis Institutes for Biomedical Research, Basel, CH-4002, Switzerland.
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto, Medical School, University of São Paulo, São Paulo, Brazil.
| | - Gustavo B Menezes
- Department of Morphology, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - M Fatima Leite
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| |
Collapse
|
33
|
Tanaka A, Yuasa S, Mearini G, Egashira T, Seki T, Kodaira M, Kusumoto D, Kuroda Y, Okata S, Suzuki T, Inohara T, Arimura T, Makino S, Kimura K, Kimura A, Furukawa T, Carrier L, Node K, Fukuda K. Endothelin-1 induces myofibrillar disarray and contractile vector variability in hypertrophic cardiomyopathy-induced pluripotent stem cell-derived cardiomyocytes. J Am Heart Assoc 2014; 3:e001263. [PMID: 25389285 PMCID: PMC4338713 DOI: 10.1161/jaha.114.001263] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Despite the accumulating genetic and molecular investigations into hypertrophic cardiomyopathy (HCM), it remains unclear how this condition develops and worsens pathologically and clinically in terms of the genetic-environmental interactions. Establishing a human disease model for HCM would help to elucidate these disease mechanisms; however, cardiomyocytes from patients are not easily obtained for basic research. Patient-specific induced pluripotent stem cells (iPSCs) potentially hold much promise for deciphering the pathogenesis of HCM. The purpose of this study is to elucidate the interactions between genetic backgrounds and environmental factors involved in the disease progression of HCM. METHODS AND RESULTS We generated iPSCs from 3 patients with HCM and 3 healthy control subjects, and cardiomyocytes were differentiated. The HCM pathological phenotypes were characterized based on morphological properties and high-speed video imaging. The differences between control and HCM iPSC-derived cardiomyocytes were mild under baseline conditions in pathological features. To identify candidate disease-promoting environmental factors, the cardiomyocytes were stimulated by several cardiomyocyte hypertrophy-promoting factors. Interestingly, endothelin-1 strongly induced pathological phenotypes such as cardiomyocyte hypertrophy and intracellular myofibrillar disarray in the HCM iPSC-derived cardiomyocytes. We then reproduced these phenotypes in neonatal cardiomyocytes from the heterozygous Mybpc3-targeted knock in mice. High-speed video imaging with motion vector prediction depicted physiological contractile dynamics in the iPSC-derived cardiomyocytes, which revealed that self-beating HCM iPSC-derived single cardiomyocytes stimulated by endothelin-1 showed variable contractile directions. CONCLUSIONS Interactions between the patient's genetic backgrounds and the environmental factor endothelin-1 promote the HCM pathological phenotype and contractile variability in the HCM iPSC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.) Department of Cardiovascular Medicine, Saga University, Saga, Japan (A.T., K.N.)
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Giulia Mearini
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (G.M., L.C.) DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (G.M., L.C.)
| | - Toru Egashira
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Tomohisa Seki
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Masaki Kodaira
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Dai Kusumoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Yusuke Kuroda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Shinichiro Okata
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.) Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan (S.O., T.F.)
| | - Tomoyuki Suzuki
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Taku Inohara
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Takuro Arimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan (T.A., A.K.)
| | - Shinji Makino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Kensuke Kimura
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| | - Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan (T.A., A.K.)
| | - Tetsushi Furukawa
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan (S.O., T.F.)
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (G.M., L.C.) DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (G.M., L.C.)
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan (A.T., K.N.)
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (A.T., S.Y., T.E., T.S., M.K., D.K., Y.K., S.O., T.S., T.I., S.M., K.K., K.F.)
| |
Collapse
|
34
|
The cardiac ryanodine receptor luminal Ca2+ sensor governs Ca2+ waves, ventricular tachyarrhythmias and cardiac hypertrophy in calsequestrin-null mice. Biochem J 2014; 461:99-106. [PMID: 24758151 DOI: 10.1042/bj20140126] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
CASQ2 (cardiac calsequestrin) is commonly believed to serve as the SR (sarcoplasmic reticulum) luminal Ca2+ sensor. Ablation of CASQ2 promotes SCWs (spontaneous Ca2+ waves) and CPVT (catecholaminergic polymorphic ventricular tachycardia) upon stress but not at rest. How SCWs and CPVT are triggered by stress in the absence of the CASQ2-based luminal Ca2+ sensor is an important unresolved question. In the present study, we assessed the role of the newly identified RyR2 (ryanodine receptor 2)-resident luminal Ca2+ sensor in determining SCW propensity, CPVT susceptibility and cardiac hypertrophy in Casq2-KO (knockout) mice. We crossbred Casq2-KO mice with RyR2 mutant (E4872Q+/-) mice, which lack RyR2-resident SR luminal Ca2+ sensing, to generate animals with both deficiencies. Casq2+/- and Casq2-/- mice showed stress-induced VTs (ventricular tachyarrhythmias), whereas Casq2+/-/E4872Q+/- and Casq2-/-/E4872Q+/- mice displayed little or no stress-induced VTs. Confocal Ca2+ imaging revealed that Casq2-/- hearts frequently exhibited SCWs after extracellular Ca2+ elevation or adrenergic stimulation, whereas Casq2-/-/E4872Q+/- hearts had few or no SCWs under the same conditions. Cardiac hypertrophy developed and CPVT susceptibility increased with age in Casq2-/- mice, but not in Casq2-/-/E4872Q+/- mice. However, the amplitudes and dynamics of voltage-induced Ca2+ transients in Casq2-/- and Casq2-/-/E4872Q+/- hearts were not significantly different. Our results indicate that SCWs, CPVT and hypertrophy in Casq2-null cardiac muscle are governed by the RyR2-resident luminal Ca2+ sensor. This implies that defects in CASQ2-based lumi-nal Ca2+ sensing can be overridden by the RyR2-resident luminal Ca2+ sensor. This makes this RyR2-resident sensor a promising molecular target for the treatment of Ca2+-mediated arrhythmias.
Collapse
|
35
|
Abstract
Activated/uninhibited calcineurin is both necessary and sufficient to induce cardiac hypertrophy, a condition that often leads to dilated cardiomyopathy, heart failure, and sudden cardiac death. We expressed constitutively active calcineurin in the adult heart of Drosophila melanogaster and identified enlarged cardiac chamber dimensions and reduced cardiac contractility. In addition, expressing constitutively active calcineurin in the fly heart using the Gal4/UAS system induced an increase in heart wall thickness. We performed a targeted genetic screen for modifiers of calcineurin-induced cardiac enlargement based on previous calcineurin studies in the fly and identified galactokinase as a novel modifier of calcineurin-induced cardiomyopathy. Genomic deficiencies spanning the galactokinase locus, transposable elements that disrupt galactokinase, and cardiac-specific RNAi knockdown of galactokinase suppressed constitutively active calcineurin-induced cardiomyopathy. In addition, in flies expressing constitutively active calcineurin using the Gal4/UAS system, a transposable element in galactokinase suppressed the increase in heart wall thickness. Finally, genetic disruption of galactokinase suppressed calcineurin-induced wing vein abnormalities. Collectively, we generated a model for discovering novel modifiers of calcineurin-induced cardiac enlargement in the fly and identified galactokinase as a previously unknown regulator of calcineurin-induced cardiomyopathy in adult Drosophila.
Collapse
|
36
|
Lee SH, Choi J, Kim H, Lee DH, Roh GS, Kim HJ, Kang SS, Choi WS, Cho GJ. FK506 reduces calpain-regulated calcineurin activity in both the cytoplasm and the nucleus. Anat Cell Biol 2014; 47:91-100. [PMID: 24987545 PMCID: PMC4076426 DOI: 10.5115/acb.2014.47.2.91] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 05/30/2014] [Accepted: 06/03/2014] [Indexed: 12/25/2022] Open
Abstract
Excessive immune responses induced by ischemia-reperfusion injury (IRI) are known to lead to necrotic and apoptotic cell death, and calcineurin plays a major role in this process. Calcineurin dephosphorylates the nuclear factor of activated T-cells (NFAT), permitting its translocation into the nucleus. As a result, calcineurin promotes the release of pro-inflammatory cytokines, such as tumor necrosis factor-α. The overproduction of pro-inflammatory cytokines causes renal cell death. Calcineurin activity is regulated by calpain, a cysteine protease present in the nucleus. Calpain-mediated proteolysis increases the phosphatase activity of calcineurin, resulting in NFAT dephosphorylation. This process has been studied in cardiomyocytes but its role in renal IRI is unknown. Thus, we examined whether calpain regulates calcineurin in renal tubule nuclei. We established an in vivo renal IRI model in mice and identified the protective role of a calcineurin inhibitor, FK506, in this process. Calcineurin is expressed in the nucleus, where it is present in its calpain-cleaved form. FK506 reduced nuclear expression of calcineurin and prevented calcineurin-mediated NFAT activation. Our study shows clearly that FK506 reduces calpain-mediated calcineurin activity. Consequently, calcineurin could not maintain NFAT activation. FK506 reduced renal cell death by suppressing the transcription of pro-inflammatory cytokine genes. This study provides evidence that FK506 protects against inflammation in a renal IRI mouse model. We also provided a mechanism of calcineurin action in the nucleus. Therefore, FK506 could improve renal function by decreasing calcineurin activity in both the cytoplasm and the nucleus of renal tubule cells.
Collapse
Affiliation(s)
- Sun Hee Lee
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Jungil Choi
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Hwajin Kim
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Dong Hoon Lee
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Gu Seob Roh
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Hyun Joon Kim
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Sang Soo Kang
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Wan Sung Choi
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Gyeong Jae Cho
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| |
Collapse
|
37
|
Sequeira V, Nijenkamp LLAM, Regan JA, van der Velden J. The physiological role of cardiac cytoskeleton and its alterations in heart failure. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:700-22. [PMID: 23860255 DOI: 10.1016/j.bbamem.2013.07.011] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 12/11/2022]
Abstract
Cardiac muscle cells are equipped with specialized biochemical machineries for the rapid generation of force and movement central to the work generated by the heart. During each heart beat cardiac muscle cells perceive and experience changes in length and load, which reflect one of the fundamental principles of physiology known as the Frank-Starling law of the heart. Cardiac muscle cells are unique mechanical stretch sensors that allow the heart to increase cardiac output, and adjust it to new physiological and pathological situations. In the present review we discuss the mechano-sensory role of the cytoskeletal proteins with respect to their tight interaction with the sarcolemma and extracellular matrix. The role of contractile thick and thin filament proteins, the elastic protein titin, and their anchorage at the Z-disc and M-band, with associated proteins are reviewed in physiologic and pathologic conditions leading to heart failure. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé
Collapse
Affiliation(s)
- Vasco Sequeira
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Louise L A M Nijenkamp
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Jessica A Regan
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; Department of Physiology, Molecular Cardiovascular Research Program, Sarver Heart Center, University of Arizona, Tucson, AZ 85724, USA
| | - Jolanda van der Velden
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; ICIN-Netherlands Heart Institute, The Netherlands.
| |
Collapse
|
38
|
Reineke EL, York B, Stashi E, Chen X, Tsimelzon A, Xu J, Newgard CB, Taffet GE, Taegtmeyer H, Entman ML, O’Malley BW. SRC-2 coactivator deficiency decreases functional reserve in response to pressure overload of mouse heart. PLoS One 2012; 7:e53395. [PMID: 23300926 PMCID: PMC3534027 DOI: 10.1371/journal.pone.0053395] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 11/28/2012] [Indexed: 11/18/2022] Open
Abstract
A major component of the cardiac stress response is the simultaneous activation of several gene regulatory networks. Interestingly, the transcriptional regulator steroid receptor coactivator-2, SRC-2 is often decreased during cardiac failure in humans. We postulated that SRC-2 suppression plays a mechanistic role in the stress response and that SRC-2 activity is an important regulator of the adult heart gene expression profile. Genome-wide microarray analysis, confirmed with targeted gene expression analyses revealed that genetic ablation of SRC-2 activates the "fetal gene program" in adult mice as manifested by shifts in expression of a) metabolic and b) sarcomeric genes, as well as associated modulating transcription factors. While these gene expression changes were not accompanied by changes in left ventricular weight or cardiac function, imposition of transverse aortic constriction (TAC) predisposed SRC-2 knockout (KO) mice to stress-induced cardiac dysfunction. In addition, SRC-2 KO mice lacked the normal ventricular hypertrophic response as indicated through heart weight, left ventricular wall thickness, and blunted molecular signaling known to activate hypertrophy. Our results indicate that SRC-2 is involved in maintenance of the steady-state adult heart transcriptional profile, with its ablation inducing transcriptional changes that mimic a stressed heart. These results further suggest that SRC-2 deletion interferes with the timing and integration needed to respond efficiently to stress through disruption of metabolic and sarcomeric gene expression and hypertrophic signaling, the three key stress responsive pathways.
Collapse
Affiliation(s)
- Erin L. Reineke
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Erin Stashi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xian Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anna Tsimelzon
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Christopher B. Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical School, Durham, North Carolina, United States of America
| | - George E. Taffet
- Department of Medicine, Division of Cardiovascular Sciences, Baylor College of Medicine, Houston, Texas, United States of America
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Mark L. Entman
- Department of Medicine, Division of Cardiovascular Sciences, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bert W. O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
39
|
Sugiura R, Kita A, Tsutsui N, Muraoka O, Hagihara K, Umeda N, Kunoh T, Takada H, Hirose D. Acremomannolipin A, the potential calcium signal modulator with a characteristic glycolipid structure from the filamentous fungus Acremonium strictum. Bioorg Med Chem Lett 2012; 22:6735-9. [DOI: 10.1016/j.bmcl.2012.08.085] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 08/11/2012] [Accepted: 08/22/2012] [Indexed: 01/26/2023]
|
40
|
Zeidan A, Siam A, Al Kaabba A, Mohammad M, Khatib S. The ability of phosphodiesterase-5 inhibitors sildenafil and ordonafil to reverse L-NAME induced cardiac hypertrophy in the rabbit: possible role of calcineurin and p38. Can J Physiol Pharmacol 2012; 90:1247-55. [PMID: 22913522 DOI: 10.1139/y2012-098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Phosphodiesterase 5 inhibitors (PDE-5Is) can suppress and (or) reverse pressure overload induced myocardial hypertrophy. This study investigated the suppressive effect of 2 PDE-5Is (sildenafil and ordonafil) on N-nitro-l-arginine methyl ester (L-NAME)-induced cardiac hypertrophy in rabbit heart, and examined their possible mechanism of action. L-NAME increased left ventricular thickness to 6.1± 0.18 mm from 4.6 ± 0.13 mm (p < 0.05), which regressed after treatment with either sildenafil or ordonafil to 5.1 ± 0.1 mm and 4.8 ± 0.2 mm, respectively (p < 0.05). Phenylephrine increased neonatal rat ventricular myocyte cell surface area to 131% ± 3% of the control value, which was associated with significant increment in ERK1/2 to 143% ± 5% of the control value (p < 0.05). Ordonafil and sildenafil decreased cell surface area to 95% ± 3% and 90% ± 1% of the control value, respectively. Both drugs decreased ERK1/2 to 88% ± 4% of the control value. Calcineurin activity was significantly decreased after 1 h of treatment with 0.1 mg·L(-1) ordonafil (1.15 ± 0.05, p < 0.05). For sildenafil (0.1 mg·L(-1)), calcineurin activity significantly decreased only after 24 h of incubation (22%). Also p38 activation was attenuated by ordonafil and sildenafil (0.1 mg·L(-1)). It is suggested that both drugs have the ability to reverse L-NAME-induced cardiac hypertrophy and suppress phenylphrine-induced myocyte hypertrophy, and that these effects may be mediated through the attenuation of calcineurin and its downstream signaling pathways (p38) in neonatal rat ventricular myocytes.
Collapse
Affiliation(s)
- Asad Zeidan
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Lebanon
| | | | | | | | | |
Collapse
|
41
|
Mohamed BA, Barakat AZ, Zimmermann WH, Bittner RE, Mühlfeld C, Hünlich M, Engel W, Maier LS, Adham IM. Targeted disruption of Hspa4 gene leads to cardiac hypertrophy and fibrosis. J Mol Cell Cardiol 2012; 53:459-68. [PMID: 22884543 DOI: 10.1016/j.yjmcc.2012.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 07/12/2012] [Accepted: 07/24/2012] [Indexed: 01/14/2023]
Abstract
Failure of molecular chaperones to direct the correct folding of newly synthesized proteins leads to the accumulation of misfolded proteins in cells. HSPA4 is a member of the heat shock protein 110 family (HSP110) that acts as a nucleotide exchange factor of HSP70 chaperones. We found that the expression of HSPA4 is upregulated in murine hearts subjected to pressure overload and in failing human hearts. To investigate the cardiac function of HSPA4, Hspa4 knockout (KO) mice were generated and exhibited cardiac hypertrophy and fibrosis. Hspa4 KO hearts were characterized by a significant increase in heart weight/body weight ratio, elevated expression of hypertrophic and fibrotic gene markers, and concentric hypertrophy with preserved contractile function. In response to pressure overload, cardiac hypertrophy and remodeling were further aggravated in the Hspa4 KO compared to wild type (WT) mice. Cardiac hypertrophy in Hspa4 KO hearts was associated with enhanced activation of gp130-STAT3, CaMKII, and calcineurin-NFAT signaling. Protein blot and immunofluorescent analyses showed a significant accumulation of polyubiquitinated proteins in cardiac cells of Hspa4 KO mice. These results suggest that the myocardial remodeling of Hspa4 KO mice is due to accumulation of misfolded proteins resulting from impaired chaperone activity. Further analyses revealed a significant increase in cross sectional area of cardiomyocytes, and in expression levels of hypertrophic markers in cultured neonatal Hspa4 KO cardiomyocytes suggesting that the hypertrophy of mutant mice was a result of primary defects in cardiomyocytes. Gene expression profile in hearts of 3.5-week-old mice revealed a differentially expressed gene sets related to ion channels, muscle-specific contractile proteins and stress response. Taken together, our in vivo data demonstrate that Hspa4 gene ablation results in cardiac hypertrophy and fibrosis, possibly, through its role in protein quality control mechanism.
Collapse
Affiliation(s)
- Belal A Mohamed
- Institute of Human Genetics, University of Göttingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Merlet E, Lipskaia L, Marchand A, Hadri L, Mougenot N, Atassi F, Liang L, Hatem SN, Hajjar RJ, Lompré AM. A calcium-sensitive promoter construct for gene therapy. Gene Ther 2012; 20:248-54. [PMID: 22456325 DOI: 10.1038/gt.2012.30] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Targeting diseased cells is a challenging issue in both pharmacological and biological therapeutics. Gene therapy is emerging as a novel approach for treating rare diseases and for illnesses for which there is no other alternative. An important limitation of gene therapy has been the off-target effects and therefore efforts have been focused on increasing the specificity of gene transfer to the targeted organ. Here, we describe a promoter containing six nuclear factor of activated T cells (NFAT) consensus sequences, which is as efficient as the cytomegalovirus (CMV) promoter to drive expression in vascular smooth muscle cells both in vitro and in vivo. In contrast to the CMV promoter it is activated in a Ca(2+)-dependent manner after endoplasmic reticulum depletion and allows the transgene expression only in proliferative/diseased cells. Overexpression of sarco/endoplasmic reticulum (SR/ER) Ca(2+) ATPase 2a under the control of this NFAT promoter inhibits restenosis after angioplasty in rats. In conclusion, this promoter may be useful for gene therapy in vascular proliferative diseases and other diseases involving upregulation of the NFAT pathway.
Collapse
Affiliation(s)
- E Merlet
- Transatlantic Cardiovascular Research Center, INSERM UMRS 956, UPMC-Paris 6, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Liu Q, Chen Y, Auger-Messier M, Molkentin JD. Interaction between NFκB and NFAT coordinates cardiac hypertrophy and pathological remodeling. Circ Res 2012; 110:1077-86. [PMID: 22403241 DOI: 10.1161/circresaha.111.260729] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RATIONALE Both nuclear factors of activated T cells (NFAT) and nuclear factor-κB (NFκB) are Rel homology domain (RHD)-containing transcription factors whose independent activities are critically involved in regulating cardiac hypertrophy and failure. OBJECTIVE To determine the potential functional interaction between NFAT and NFκB signaling pathways in cardiomyocytes and its role in cardiac hypertrophy and remodeling. METHODS AND RESULTS We identified a novel transcriptional regulatory mechanism whereby NFκB and NFAT directly interact and synergistically promote transcriptional activation in cardiomyocytes. We show that the p65 subunit of NFκB coimmunoprecipitates with NFAT in cardiomyocytes, and this interaction maps to the RHD within p65. Overexpression of the p65-RHD disrupts the association between endogenous p65 and NFATc1, leading to reduced transcriptional activity. Overexpression of IκB kinase β (IKKβ) or p65-RHD causes nuclear translocation of NFATc1, and expression of a constitutively nuclear NFATc1-SA mutant similarly facilitated p65 nuclear translocation. Combined overexpression of p65 and NFATc1 promotes synergistic activation of NFAT transcriptional activity in cardiomyocytes, whereas inhibition of NFκB with IκBαM or dominant negative IKKβ reduces NFAT activity. Importantly, agonist-induced NFAT activation is reduced in p65 null mouse embryonic fibroblasts (MEFs) compared with wild-type MEFs. In vivo, cardiac-specific deletion of p65 using a Cre-loxP system causes a ≈50% reduction in NFAT activity in luciferase reporter mice. Moreover, ablation of p65 in the mouse heart decreases the hypertrophic response after pressure overload stimulation, reduces the degree of pathological remodeling, and preserves contractile function. CONCLUSIONS Our results suggest a direct interaction between NFAT and NFκB that effectively integrates 2 disparate signaling pathways in promoting cardiac hypertrophy and ventricular remodeling.
Collapse
Affiliation(s)
- Qinghang Liu
- Department of Physiology and Biophysics, University of Washington, Seattle, USA
| | | | | | | |
Collapse
|
44
|
Tang Y, Tian X, Wang R, Fill M, Chen SRW. Abnormal termination of Ca2+ release is a common defect of RyR2 mutations associated with cardiomyopathies. Circ Res 2012; 110:968-77. [PMID: 22374134 DOI: 10.1161/circresaha.111.256560] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Naturally occurring mutations in the cardiac ryanodine receptor (RyR2) have been associated with both cardiac arrhythmias and cardiomyopathies. It is clear that delayed afterdepolarization resulting from abnormal activation of sarcoplasmic reticulum Ca2+ release is the primary cause of RyR2-associated cardiac arrhythmias. However, the mechanism underlying RyR2-associated cardiomyopathies is completely unknown. OBJECTIVE In the present study, we investigate the role of the NH2-terminal region of RyR2 in and the impact of a number of cardiomyopathy-associated RyR2 mutations on the termination of Ca2+ release. METHODS AND RESULTS The 35-residue exon-3 region of RyR2 is associated with dilated cardiomyopathy. Single-cell luminal Ca2+ imaging revealed that the deletion of the first 305 NH2-terminal residues encompassing exon-3 or the deletion of exon-3 itself markedly reduced the luminal Ca2+ threshold at which Ca2+ release terminates and increased the fractional Ca2+ release. Single-cell cytosolic Ca2+ imaging also showed that both RyR2 deletions enhanced the amplitude of store overload-induced Ca2+ transients in HEK293 cells or HL-1 cardiac cells. Furthermore, the RyR2 NH2-terminal mutations, A77V, R176Q/T2504M, R420W, and L433P, which are associated with arrhythmogenic right ventricular displasia type 2, also reduced the threshold for Ca2+ release termination and increased fractional release. The RyR2 A1107M mutation associated with hypertrophic cardiomyopathy had the opposite action (i.e., increased the threshold for Ca2+ release termination and reduced fractional release). CONCLUSIONS These results provide the first evidence that the NH2-terminal region of RyR2 is an important determinant of Ca2+ release termination, and that abnormal fractional Ca2+ release attributable to aberrant termination of Ca2+ release is a common defect in RyR2-associated cardiomyopathies.
Collapse
Affiliation(s)
- Yijun Tang
- Department of Physiology and Pharmacology, the Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | | | | | | | | |
Collapse
|
45
|
Tacrolimus Attenuates Myocardium Damage to the Total Hepatic Ischemia-Reperfusion Via Regulation of the Mitochondrial Function. J Surg Res 2012; 172:e47-54. [DOI: 10.1016/j.jss.2010.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/30/2010] [Accepted: 10/15/2010] [Indexed: 11/18/2022]
|
46
|
HE ZIHONG, HU YANG, LI YANCHUN, YVERT THOMAS, SANTIAGO CATALINA, GÓMEZ-GALLEGO FÉLIX, RUIZ JONATANR, LUCIA ALEJANDRO. Are Calcineurin Genes Associated with Athletic Status? A Function, Replication Study. Med Sci Sports Exerc 2011; 43:1433-40. [DOI: 10.1249/mss.0b013e31820e7f38] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
47
|
Involvement of calcineurin in ischemic myocardial damage. Int J Angiol 2011. [DOI: 10.1007/s00547-005-2005-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
48
|
Abstract
Oxidative stress is common in many clinically important cardiac disorders, including ischemia/reperfusion, diabetes, and hypertensive heart disease. Oxidative stress leads to derangements in pump function due to changes in the expression or function of proteins that regulate intracellular Ca(2+) homeostasis. There is growing evidence that the cardiodepressant actions of reactive oxygen species (ROS) also are attributable to ROS-dependent signaling events in the sarcomere. This minireview focuses on myofilament protein post-translational modifications induced by ROS or ROS-activated signaling enzymes that regulate cardiac contractility.
Collapse
Affiliation(s)
- Marius P Sumandea
- Department of Physiology, Center for Muscle Biology, University of Kentucky, Lexington, Kentucky 40536, USA.
| | | |
Collapse
|
49
|
Abstract
Cardiac hypertrophy is classically considered as an adaptive and compensatory response enabling cardiomyocytes to increase their work output and thus cardiac function. Biomechanical stress and neurohumoral activation are the most important triggers of pathological hypertrophy and the transition of cardiac hypertrophy to heart failure. Several novel regulators and putative drug targets of cardiac hypertrophy have been found by using gene-modified and acquired models of cardiac hypertrophy. Recent studies have also revealed distinct patterns of cardiac substrate utilization in cardiac hypertrophy and heart failure. The use of novel systems biology techniques such as metabolomics may therefore in future provide insights into the metabolic processes and cardiovascular biology related to cardiac hypertrophy and also extend the ability to discover circulating biomarkers for cardiovascular diseases. The present review discusses current knowledge on molecular mechanisms of cardiac hypertrophy, with special emphasis on novel regulators and putative drug targets of cardiac hypertrophy such as the tissue renin-angiotensin-aldosterone system, calcineurin/nuclear factor of activated T cells pathway, phosphatidylinositol 3-kinase/growth promoting protein kinase B, mammalian target of rapamycin, histone deacetylases, AMPkinases, microRNAs and angiogenetic factors.
Collapse
|
50
|
Escobar M, Cardenas C, Colavita K, Petrenko NB, Franzini-Armstrong C. Structural evidence for perinuclear calcium microdomains in cardiac myocytes. J Mol Cell Cardiol 2010; 50:451-9. [PMID: 21147122 DOI: 10.1016/j.yjmcc.2010.11.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 11/24/2010] [Accepted: 11/29/2010] [Indexed: 10/18/2022]
Abstract
At each heartbeat, cardiac myocytes are activated by a cytoplasmic Ca(2+) transient in great part due to Ca(2+) release from the sarcoplasmic reticulum via ryanodine receptors (RyRs) clustered within calcium release units (peripheral couplings/dyads). A Ca(2+) transient also occurs in the nucleoplasm, following the cytoplasmic transient with some delay. Under conditions where the InsP3 production is stimulated, these Ca(2+) transients are regulated actively, presumably by an additional release of Ca(2+) via InsP3 receptors (InsP3Rs). This raises the question whether InsP3Rs are appropriately located for this effect and whether sources of InsP3 and Ca(2+) are available for their activation. We have defined the structural basis for InsP3R activity at the nucleus, using immunolabeling for confocal microscopy and freeze-drying/shadowing, T tubule "staining" and thin sectioning for electron microscopy. By these means we establish the presence of InsP3R at the outer nuclear envelope and show a close spatial relationship between the nuclear envelope, T tubules (a likely source of InsP3) and dyads (the known source of Ca(2+)). The frequency, distribution and distance from the nucleus of T tubules and dyads appropriately establish local perinuclear Ca(2+) microdomains in cardiac myocytes.
Collapse
Affiliation(s)
- Matias Escobar
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | | | | | | | | |
Collapse
|