1
|
Parikh VN, Day SM, Lakdawala NK, Adler ED, Olivotto I, Seidman CE, Ho CY. Advances in the study and treatment of genetic cardiomyopathies. Cell 2025; 188:901-918. [PMID: 39983674 DOI: 10.1016/j.cell.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 02/23/2025]
Abstract
Cardiomyopathies are primary disorders of the heart muscle. Three key phenotypes have been defined, based on morphology and arrhythmia burden: hypertrophic cardiomyopathy (HCM), with thickened heart muscle and diastolic dysfunction; dilated cardiomyopathy (DCM), with left ventricular enlargement and systolic dysfunction; and arrhythmogenic cardiomyopathy (ACM), with right, left, or biventricular involvement and arrhythmias out of proportion to systolic dysfunction. Genetic discoveries of the molecular basis of disease are paving the way for greater precision in diagnosis and management and revealing mechanisms that account for distinguishing clinical features. This deeper understanding has propelled the development of new treatments for cardiomyopathies: disease-specific, mechanistically based medicines that counteract pathophysiology, and emergent gene therapies that aim to intercept disease progression and restore cardiac physiology. Together, these discoveries have advanced fundamental insights into cardiac biology and herald a new era for patients with cardiomyopathy.
Collapse
Affiliation(s)
- Victoria N Parikh
- Stanford Center for Inherited Cardiovascular Disease, Stanford School of Medicine, Stanford, CA, USA
| | - Sharlene M Day
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Neal K Lakdawala
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Eric D Adler
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | | | - Christine E Seidman
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Carolyn Y Ho
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
2
|
Sadayappan S. Proteomic Profiling: The Key to Unlocking the Complexities of Hypertrophic Cardiomyopathy. J Am Coll Cardiol 2024; 84:2012-2013. [PMID: 39365225 DOI: 10.1016/j.jacc.2024.08.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024]
Affiliation(s)
- Sakthivel Sadayappan
- Department of Cellular and Molecular Medicine, Sarver Heart Center, Molecular Cardiovascular Research Program, University of Arizona College of Medicine, Tucson, Arizona, USA.
| |
Collapse
|
3
|
Jean-Charles PY, Roy B, Yu SMW, Pironti G, Nagi K, Mao L, Kaur S, Abraham DM, Maudsley S, Rockman HA, Shenoy SK. USP20 deletion promotes eccentric cardiac remodeling in response to pressure overload and increases mortality. Am J Physiol Heart Circ Physiol 2024; 327:H1257-H1271. [PMID: 39365672 PMCID: PMC11559650 DOI: 10.1152/ajpheart.00329.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024]
Abstract
Left ventricular hypertrophy (LVH) caused by chronic pressure overload with subsequent pathological remodeling is a major cardiovascular risk factor for heart failure and mortality. The role of deubiquitinases in LVH has not been well characterized. To define whether the deubiquitinase ubiquitin-specific peptidase 20 (USP20) regulates LVH, we subjected USP20 knockout (KO) and cognate wild-type (WT) mice to chronic pressure overload by transverse aortic constriction (TAC) and measured changes in cardiac function by serial echocardiography followed by histological and biochemical evaluations. USP20-KO mice showed severe deterioration of systolic function within 4 wk of TAC compared with WT cohorts. Both USP20-KO TAC and WT-TAC cohorts presented cardiac hypertrophy following pressure overload. However, USP20-KO-TAC mice showed an increase in cardiomyocyte length and developed maladaptive eccentric hypertrophy, a phenotype generally observed with volume overload states and decompensated heart failure. In contrast, WT-TAC mice displayed an increase in cardiomyocyte width, producing concentric remodeling that is characteristic of pressure overload. In addition, cardiomyocyte apoptosis, interstitial fibrosis, and mouse mortality were augmented in USP20-KO-TAC compared with WT-TAC mice. Quantitative mass spectrometry of LV tissue revealed that the expression of sarcomeric myosin heavy chain 7 (MYH7), a fetal gene normally upregulated during cardiac remodeling, was significantly reduced in USP20-KO after TAC. Mechanistically, we identified increased degradative lysine-48 polyubiquitination of MYH7 in USP20-KO hearts, indicating that USP20-mediated deubiquitination likely prevents protein degradation of MYH7 during pressure overload. Our findings suggest that USP20-dependent signaling pathways regulate the layering pattern of sarcomeres to suppress maladaptive remodeling during chronic pressure overload and prevent cardiac failure.NEW & NOTEWORTHY We identify ubiquitin-specific peptidase 20 (USP20) as an important enzyme that is required for cardiac homeostasis and function, particularly during myocardial pressure overload. USP20 regulates protein stability of cardiac MYH7, an essential molecular motor protein expressed in sarcomeres; loss-of-function mutations of MYH7 are associated with human hypertrophic cardiomyopathy, cardiac failure, and sudden death. Enhancing USP20 activity could be a potential therapeutic approach to prevent the development of maladaptive state of eccentric hypertrophy and heart failure.
Collapse
MESH Headings
- Animals
- Ventricular Remodeling
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Mice, Knockout
- Ubiquitin Thiolesterase/metabolism
- Ubiquitin Thiolesterase/genetics
- Apoptosis
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Mice
- Mice, Inbred C57BL
- Male
- Heart Failure/physiopathology
- Heart Failure/metabolism
- Heart Failure/genetics
- Heart Failure/pathology
- Fibrosis
- Ventricular Function, Left
- Disease Models, Animal
- Ubiquitination
- Myosin Heavy Chains/metabolism
- Myosin Heavy Chains/genetics
Collapse
Affiliation(s)
- Pierre-Yves Jean-Charles
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
| | - Bipradas Roy
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
| | - Samuel Mon-Wei Yu
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
| | - Gianluigi Pironti
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
| | - Karim Nagi
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
| | - Lan Mao
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
| | - Suneet Kaur
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
| | - Dennis M Abraham
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
| | - Stuart Maudsley
- Receptor Biology Laboratory, Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
| | - Howard A Rockman
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States
| | - Sudha K Shenoy
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
4
|
Van Theemsche KM, Frans L, Van de Sande DV, Martinez-Morales E, Snyders DJ, Labro AJ. Evaluating the proarrhythmic risk of delayed-action compounds in serum free cell culture conditions; serum-starvation accelerates/amplifies the effect of probucol on the KCNQ1 + KCNE1 channel. J Pharmacol Toxicol Methods 2024; 130:107566. [PMID: 39357805 DOI: 10.1016/j.vascn.2024.107566] [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: 06/26/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
In vitro testing procedures for evaluating acute effects of compound on ion channels, utilizing heterologous expression systems (HES), are well-established, while slowly manifesting delayed effects remain challenging to detect. For this, immortalized HES are exposed to the compounds for a longer time, in general 24 h. As these cells proliferate every 12-20 h, we evaluated if the proliferation status, and by extension cell metabolism, influences the delayed compound response. The intervention of halting cell proliferation by excluding serum from the culturing medium was evaluated on CHO cells, stably expressing the KCNQ1 + KCNE1 channel complex that mediates the slow delayed rectifier potassium current (Iks). No abnormal changes in KCNQ1 + KCNE1 current were observed upon serum-starvation, except for a negative shift in the voltage dependence of channel activation (GV-curve) after 72 h. The delayed effect of probucol, a compound reported to interfere with Iks expression, was evaluated after 24 and 72 h of incubation. In serum-free conditions the inhibitory effect of probucol was increased fourfold after 24 h, compared to serum supplemented conditions. After 72 h, the current inhibition was similar between both culture conditions. Besides decreasing current expression, probucol shifted the GV-curve more positive combined with a shallower voltage response, changes that were more pronounced in serum-depleted conditions. The results indicated that serum-starvation had no substantial effect on the KCNQ1 + KCNE1 current in the tested CHO cells, but it amplified or accelerated the response to probucol, suggesting that halting cell proliferation is a method for enhancing the detection of delayed compound effects in HES.
Collapse
Affiliation(s)
- Kenny M Van Theemsche
- Department of Basic and Applied Medical Sciences, Faculty of Medicine, Ghent University, 9000 Ghent, Belgium; Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Lisse Frans
- Department of Basic and Applied Medical Sciences, Faculty of Medicine, Ghent University, 9000 Ghent, Belgium
| | - Dieter V Van de Sande
- Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Evelyn Martinez-Morales
- Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Dirk J Snyders
- Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Alain J Labro
- Department of Basic and Applied Medical Sciences, Faculty of Medicine, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
5
|
Ott C. Mapping the interplay of immunoproteasome and autophagy in different heart failure phenotypes. Free Radic Biol Med 2024; 218:149-165. [PMID: 38570171 DOI: 10.1016/j.freeradbiomed.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
Proper protein degradation is required for cellular protein homeostasis and organ function. Particularly, in post-mitotic cells, such as cardiomyocytes, unbalanced proteolysis due to inflammatory stimuli and oxidative stress contributes to organ dysfunction. To ensure appropriate protein turnover, eukaryotic cells exert two main degradation systems, the ubiquitin-proteasome-system and the autophagy-lysosome-pathway. It has been shown that proteasome activity affects the development of cardiac dysfunction differently, depending on the type of heart failure. Studies analyzing the inducible subtype of the proteasome, the immunoproteasome (i20S), demonstrated that the i20S plays a double role in diseased hearts. While i20S subunits are increased in cardiac hypertrophy, atrial fibrillation and partly in myocarditis, the opposite applies to diabetic cardiomyopathy and ischemia/reperfusion injury. In addition, the i20S appears to play a role in autophagy modulation depending on heart failure phenotype. This review summarizes the current literature on the i20S in different heart failure phenotypes, emphasizing the two faces of i20S in injured hearts. A selection of established i20S inhibitors is introduced and signaling pathways linking the i20S to autophagy are highlighted. Mapping the interplay of the i20S and autophagy in different types of heart failure offers potential approaches for developing treatment strategies against heart failure.
Collapse
Affiliation(s)
- Christiane Ott
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
6
|
Cao H, Zhou X, Xu B, Hu H, Guo J, Ma Y, Wang M, Li N, Jun Z. Advances in the study of protein folding and endoplasmic reticulum-associated degradation in mammal cells. J Zhejiang Univ Sci B 2024; 25:212-232. [PMID: 38453636 PMCID: PMC10918413 DOI: 10.1631/jzus.b2300403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/03/2023] [Indexed: 03/09/2024]
Abstract
The endoplasmic reticulum is a key site for protein production and quality control. More than one-third of proteins are synthesized and folded into the correct three-dimensional conformation in the endoplasmic reticulum. However, during protein folding, unfolded and/or misfolded proteins are prone to occur, which may lead to endoplasmic reticulum stress. Organisms can monitor the quality of the proteins produced by endoplasmic reticulum quality control (ERQC) and endoplasmic reticulum-associated degradation (ERAD), which maintain endoplasmic reticulum protein homeostasis by degrading abnormally folded proteins. The underlying mechanisms of protein folding and ERAD in mammals have not yet been fully explored. Therefore, this paper reviews the process and function of protein folding and ERAD in mammalian cells, in order to help clinicians better understand the mechanism of ERAD and to provide a scientific reference for the treatment of diseases caused by abnormal ERAD.
Collapse
Affiliation(s)
- Hong Cao
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Xuchang Zhou
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Bowen Xu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Han Hu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Jianming Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Yuwei Ma
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Miao Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Nan Li
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China.
| | - Zou Jun
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
7
|
Meyer-Jens M, Wenzel K, Grube K, Rüdebusch J, Krämer E, Bahls M, Müller K, Voß H, Schlüter H, Felix SB, Carrier L, Könemann S, Schlossarek S. Sacubitril/valsartan reduces proteasome activation and cardiomyocyte area in an experimental mouse model of hypertrophy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 7:100059. [PMID: 39802437 PMCID: PMC11708427 DOI: 10.1016/j.jmccpl.2023.100059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/16/2025]
Abstract
Sacubitril/valsartan (Sac/Val) belongs to the group of angiotensin receptor-neprilysin inhibitors and has been used for the treatment of heart failure (HF) for several years. The mechanisms that mediate the beneficial effects of Sac/Val are not yet fully understood. In this study we investigated whether Sac/Val influences the two proteolytic systems, the ubiquitin-proteasome system (UPS) and the autophagy-lysosomal pathway (ALP), in a mouse model of pressure overload induced by transverse aortic constriction (TAC) and in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) treated with endothelin-1 (ET1) serving as a human cellular model of hypertrophy. TAC mice showed a continuous decline in cardiac function starting from day 14 after surgery. Administration of Sac/Val for 6 weeks counteracted the deterioration of cardiac function and attenuated hypertrophy and fibrosis in TAC mice. The expression of ALP key markers did not differ between the groups. Proteasome activity was higher in TAC mice and normalized by Sac/Val. In hiPSC-CMs, all treatments (Sac, Val or Sac/Val) normalized mean cell area. However, Sac alone or in combination with Val, but not Val alone prevented ET1-induced hypertrophic gene program and proteomic changes. In conclusion, Sac/Val normalized proteasome activity, improved cardiac function and reduced fibrosis and hypertrophy in TAC mice. Molecular analysis in hiPSC-CMs suggests that a major part of the beneficial effects of Sac/Val is derived from the Sac action rather than from Val.
Collapse
Affiliation(s)
- Moritz Meyer-Jens
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Kristin Wenzel
- Department of Internal Medicine B, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany
| | - Karina Grube
- Department of Internal Medicine B, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany
| | - Julia Rüdebusch
- Department of Internal Medicine B, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany
| | - Elisabeth Krämer
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Martin Bahls
- Department of Internal Medicine B, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany
| | - Kilian Müller
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Hannah Voß
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Stephan B. Felix
- Department of Internal Medicine B, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Stephanie Könemann
- Department of Internal Medicine B, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
8
|
Yang Z, Cao Y, Kong L, Xi J, Liu S, Zhang J, Cheng W. Small molecules as modulators of the proteostasis machinery: Implication in cardiovascular diseases. Eur J Med Chem 2024; 264:116030. [PMID: 38071793 DOI: 10.1016/j.ejmech.2023.116030] [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: 10/05/2023] [Revised: 11/25/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023]
Abstract
With the escalating prevalence of cardiovascular diseases, the substantial socioeconomic burden on healthcare systems is intensifying. Accumulating empirical evidence underscores the pivotal role of the proteostasis network in regulating cardiac homeostasis and function. Disruptions in proteostasis may contribute to the loss of protein function or the acquisition of toxic functions, which are intricately linked to the development of cardiovascular ailments such as atrial fibrillation, heart failure, atherosclerosis, and cardiac aging. It is widely acknowledged that the proteostasis network encompasses molecular chaperones, autophagy, and the ubiquitin proteasome system (UPS). Consequently, the proteostasis network emerges as an appealing target for therapeutic interventions in cardiovascular diseases. Numerous small molecules, acting as modulators of the proteostasis machinery, have exhibited therapeutic efficacy in managing cardiovascular diseases. This review centers on elucidating the role of the proteostasis network in various cardiovascular diseases and explores the potential of small molecules as therapeutic agents.
Collapse
Affiliation(s)
- Zhiheng Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yu Cao
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Limin Kong
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Jianjun Xi
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Shourong Liu
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China.
| | - Jiankang Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Weiyan Cheng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
9
|
Madè A, Bibi A, Garcia-Manteiga JM, Tascini AS, Piella SN, Tikhomirov R, Voellenkle C, Gaetano C, Leszek P, Castelvecchio S, Menicanti L, Martelli F, Greco S. circRNA-miRNA-mRNA Deregulated Network in Ischemic Heart Failure Patients. Cells 2023; 12:2578. [PMID: 37947656 PMCID: PMC10648415 DOI: 10.3390/cells12212578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Noncoding RNAs (ncRNAs), which include circular RNAs (circRNAs) and microRNAs (miRNAs), regulate the development of cardiovascular diseases (CVD). Notably, circRNAs can interact with miRNAs, influencing their specific mRNA targets' levels and shaping a competing endogenous RNAs (ceRNA) network. However, these interactions and their respective functions remain largely unexplored in ischemic heart failure (IHF). This study is aimed at identifying circRNA-centered ceRNA networks in non-end-stage IHF. Approximately 662 circRNA-miRNA-mRNA interactions were identified in the heart by combining state-of-the-art bioinformatics tools with experimental data. Importantly, KEGG terms of the enriched mRNA indicated CVD-related signaling pathways. A specific network centered on circBPTF was validated experimentally. The levels of let-7a-5p, miR-18a-3p, miR-146b-5p, and miR-196b-5p were enriched in circBPTF pull-down experiments, and circBPTF silencing inhibited the expression of HDAC9 and LRRC17, which are targets of miR-196b-5p. Furthermore, as suggested by the enriched pathway terms of the circBPTF ceRNA network, circBPTF inhibition elicited endothelial cell cycle arrest. circBPTF expression increased in endothelial cells exposed to hypoxia, and its upregulation was confirmed in cardiac samples of 36 end-stage IHF patients compared to healthy controls. In conclusion, circRNAs act as miRNA sponges, regulating the functions of multiple mRNA targets, thus providing a novel vision of HF pathogenesis and laying the theoretical foundation for further experimental studies.
Collapse
Affiliation(s)
- Alisia Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Alessia Bibi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jose Manuel Garcia-Manteiga
- Center for Omics Sciences COSR, BioInformatics Laboratory, San Raffaele Scientific Institute, 20132 Milan, Italy; (J.M.G.-M.); (A.S.T.)
| | - Anna Sofia Tascini
- Center for Omics Sciences COSR, BioInformatics Laboratory, San Raffaele Scientific Institute, 20132 Milan, Italy; (J.M.G.-M.); (A.S.T.)
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Santiago Nicolas Piella
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Roman Tikhomirov
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, National Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Serenella Castelvecchio
- Department of Adult Cardiac Surgery, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (S.C.); (L.M.)
| | - Lorenzo Menicanti
- Department of Adult Cardiac Surgery, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (S.C.); (L.M.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| |
Collapse
|
10
|
Yang J, Yan B, Zhang H, Lu Q, Yang L, Liu P, Bai L. Estimating the causal effects of genetically predicted plasma proteome on heart failure. Front Cardiovasc Med 2023; 10:978918. [PMID: 36860279 PMCID: PMC9968807 DOI: 10.3389/fcvm.2023.978918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 01/27/2023] [Indexed: 02/16/2023] Open
Abstract
Background Heart Failure (HF) is the end-stage cardiovascular syndrome with poor prognosis. Proteomics holds great promise in the discovery of novel biomarkers and therapeutic targets for HF. The aim of this study is to investigate the causal effects of genetically predicted plasma proteome on HF using the Mendelian randomization (MR) approach. Methods Summary-level data for the plasma proteome (3,301 healthy individuals) and HF (47,309 cases; 930,014 controls) were extracted from genome-wide association studies (GWASs) of European descent. MR associations were obtained using the inverse variance-weighted (IVW) method, sensitivity analyses, and multivariable MR analyses. Results Using single-nucleotide polymorphisms as instrumental variables, 1-SD increase in MET level was associated with an approximately 10% decreased risk of HF (odds ratio [OR]: 0.92; 95% confidence interval [CI]: 0.89 to 0.95; p = 1.42 × 10-6), whereas increases in the levels of CD209 (OR: 1.04; 95% CI: 1.02-1.06; p = 6.67 × 10-6) and USP25 (OR: 1.06; 95% CI: 1.03-1.08; p = 7.83 × 10-6) were associated with an increased risk of HF. The causal associations were robust in sensitivity analyses, and no evidence of pleiotropy was observed. Conclusion The study findings suggest that the hepatocyte growth factor/c-MET signaling pathway, dendritic cells-mediated immune processes, and ubiquitin-proteasome system pathway are involved in the pathogenesis of HF. Moreover, the identified proteins have potential to uncover novel therapies for cardiovascular diseases.
Collapse
Affiliation(s)
- Jian Yang
- Clinical Research Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Bin Yan
- Clinical Research Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Haoxuan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Qun Lu
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Lihong Yang
- Clinical Research Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Ping Liu
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Ling Bai
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China,*Correspondence: Ling Bai,
| |
Collapse
|
11
|
Myostatin/AKT/FOXO Signaling Is Altered in Human Non-Ischemic Dilated Cardiomyopathy. Life (Basel) 2022; 12:life12091418. [PMID: 36143454 PMCID: PMC9506454 DOI: 10.3390/life12091418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Disturbances in the ubiquitin proteasome system, and especially changes of the E3 ligases, are subjects of interest when searching for causes and therapies for cardiomyopathies. The aim of this study was to clarify whether the myostatin/AKT/forkhead box O (FOXO) pathway, which regulates the expression of the E3 ligases muscle atrophy F-box gene (MAFbx) and muscle ring-finger protein-1 (MuRF1), is changed in dilated cardiomyopathy of ischemic origin (IDCM) and dilated cardiomyopathy of non-ischemic origin (NIDCM). The mRNA and protein expression of myostatin, AKT, FOXO1, FOXO3, MAFbx and MuRF1 were quantified by real-time polymerase chain reaction and ELISA, respectively, in myocardial tissue from 26 IDCM and 23 NIDCM patients. Septal tissue from 17 patients undergoing Morrow resection served as a control. MAFbx and FOXO1 mRNA and protein expression (all p < 0.05), AKT mRNA (p < 0.01) and myostatin protein expression (p = 0.02) were decreased in NIDCM patients compared to the control group. Apart from decreases of AKT and MAFbx mRNA expression (both p < 0.01), no significant differences were detected in IDCM patients compared to the control group. Our results demonstrate that the myostatin/AKT/FOXO pathway is altered in NIDCM but not in IDCM patients. FOXO1 seems to be an important drug target for regulating the expression of MAFbx in NIDCM patients.
Collapse
|
12
|
Halawa S, Latif N, Tseng YT, Ibrahim AM, Chester AH, Moustafa A, Aguib Y, Yacoub MH. Profiling Genome-Wide DNA Methylation Patterns in Human Aortic and Mitral Valves. Front Cardiovasc Med 2022; 9:840647. [PMID: 35463757 PMCID: PMC9019152 DOI: 10.3389/fcvm.2022.840647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/11/2022] [Indexed: 12/05/2022] Open
Abstract
Cardiac valves exhibit highly complex structures and specialized functions that include dynamic interactions between cells, extracellular matrix (ECM) and their hemodynamic environment. Valvular gene expression is tightly regulated by a variety of mechanisms including epigenetic factors such as histone modifications, RNA-based mechanisms and DNA methylation. To date, methylation fingerprints of non-diseased human aortic and mitral valves have not been studied. In this work we analyzed the differential methylation profiles of 12 non-diseased aortic and mitral valve tissue samples (in matched pairs). Analysis of methylation data [reduced representation bisulfite sequencing (RRBS)] of 16,101 promoters genome-wide revealed 584 differentially methylated (DM) promoters, of which 13 were reported in endothelial mesenchymal trans-differentiation (EMT), 37 in aortic and mitral valve disease and 7 in ECM remodeling. Both functional classification as well as network analysis showed that the genes associated with the DM promoters were enriched for WNT-, Cadherin-, Endothelin-, PDGF-, HIF-1 and VEGF- signaling implicated in valvular physiology and pathophysiology. Additional enrichment was detected for TGFB-, NOTCH- and Integrin- signaling involved in EMT as well as ECM remodeling. This data provides the first insight into differential regulation of human aortic and mitral valve tissue and identifies candidate genes linked to DM promoters. Our work will improve the understanding of valve biology, valve tissue engineering approaches and contributes to the identification of relevant drug targets.
Collapse
Affiliation(s)
- Sarah Halawa
- Aswan Heart Centre, Aswan, Egypt
- Biotechnology Graduate Program, American University in Cairo, New Cairo, Egypt
- Sarah Halawa
| | - Najma Latif
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
| | - Yuan-Tsan Tseng
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
| | - Ayman M. Ibrahim
- Aswan Heart Centre, Aswan, Egypt
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Adrian H. Chester
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
| | - Ahmed Moustafa
- Biotechnology Graduate Program, American University in Cairo, New Cairo, Egypt
- Department of Biology, American University in Cairo, New Cairo, Egypt
| | - Yasmine Aguib
- Aswan Heart Centre, Aswan, Egypt
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
- Yasmine Aguib
| | - Magdi H. Yacoub
- Aswan Heart Centre, Aswan, Egypt
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
- *Correspondence: Magdi H. Yacoub
| |
Collapse
|
13
|
Shared genetic architecture between neuroticism, coronary artery disease and cardiovascular risk factors. Transl Psychiatry 2021; 11:368. [PMID: 34226488 PMCID: PMC8257646 DOI: 10.1038/s41398-021-01466-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 11/08/2022] Open
Abstract
Neuroticism is associated with poor health, cardiovascular disease (CVD) risk factors and coronary artery disease (CAD). The conditional/conjunctional false discovery rate method (cond/conjFDR) was applied to genome wide association study (GWAS) summary statistics on neuroticism (n = 432,109), CAD (n = 184,305) and 12 CVD risk factors (n = 188,577-339,224) to investigate genetic overlap between neuroticism and CAD and CVD risk factors. CondFDR analyses identified 729 genomic loci associated with neuroticism after conditioning on CAD and CVD risk factors. The conjFDR analyses revealed 345 loci jointly associated with neuroticism and CAD (n = 30), body mass index (BMI) (n = 96) or another CVD risk factor (n = 1-60). Several loci were jointly associated with neuroticism and multiple CVD risk factors. Seventeen of the shared loci with CAD and 61 of the shared loci with BMI are novel for neuroticism. 21 of 30 (70%) neuroticism risk alleles were associated with higher CAD risk. Functional analyses of the genes mapped to the shared loci implicated cell division, nuclear receptor, elastic fiber formation as well as starch and sucrose metabolism pathways. Our results indicate polygenic overlap between neuroticism and CAD and CVD risk factors, suggesting that genetic factors may partly cause the comorbidity. This gives new insight into the shared molecular genetic basis of these conditions.
Collapse
|
14
|
Wenzel K, Krämer E, Geertz B, Carrier L, Felix SB, Könemann S, Schlossarek S. A Transgenic Mouse Model of Eccentric Left Ventricular Hypertrophy With Preserved Ejection Fraction Exhibits Alterations in the Autophagy-Lysosomal Pathway. Front Physiol 2021; 12:614878. [PMID: 33995116 PMCID: PMC8121148 DOI: 10.3389/fphys.2021.614878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/29/2021] [Indexed: 01/02/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) and the autophagy-lysosomal pathway (ALP) are the main proteolytic systems involved in cellular homeostasis. Since cardiomyocytes, as terminally differentiated cells, lack the ability to share damaged proteins with their daughter cells, they are especially reliant on these protein degradation systems for their proper function. Alterations of the UPS and ALP have been reported in a wide range of cardiac diseases, including cardiomyopathies. In this study, we determined whether the UPS and ALP are altered in a mouse model of eccentric left ventricular (LV) hypertrophy expressing both cyclin T1 and Gαq under the control of the cardiac-specific α-myosin heavy chain promoter (double transgenic; DTG). Compared to wild-type (WT) littermates, DTG mice showed higher end-diastolic (ED) LV wall thicknesses and diameter with preserved ejection fraction (EF). The cardiomyopathic phenotype was further confirmed by an upregulation of the fetal gene program and genes associated with fibrosis as well as a downregulation of genes involved in Ca2+ handling. Likewise, higher NT-proBNP levels were detected in DTG mice. Investigation of the UPS showed elevated steady-state levels of (poly)ubiquitinated proteins without alterations of all proteasomal activities in DTG mice. Evaluation of ALP key marker revealed a mixed pattern with higher protein levels of microtubule-associated protein 1 light chain 3 beta (LC3)-I and lysosomal-associated membrane protein-2, lower protein levels of beclin-1 and FYVE and coiled-coil domain-containing protein 1 (FYCO1) and unchanged protein levels of p62/SQSTM1 in DTG mice when compared to WT. At transcriptional level, a > 1.2-fold expression was observed for Erbb2, Hdac6, Lamp2, Nrg1, and Sqstm1, while a < 0.8-fold expression was revealed for Fyco1 in DTG mice. The results related to the ALP suggested overall a repression of the ALP during the initiation process, but an induction of the ALP at the level of autophagosome-lysosome fusion and the delivery of ubiquitinated cargo to the ALP for degradation.
Collapse
Affiliation(s)
- Kristin Wenzel
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Elisabeth Krämer
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Birgit Geertz
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Stephan B Felix
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Stephanie Könemann
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
15
|
Perez-Bermejo JA, Kang S, Rockwood SJ, Simoneau CR, Joy DA, Silva AC, Ramadoss GN, Flanigan WR, Fozouni P, Li H, Chen PY, Nakamura K, Whitman JD, Hanson PJ, McManus BM, Ott M, Conklin BR, McDevitt TC. SARS-CoV-2 infection of human iPSC-derived cardiac cells reflects cytopathic features in hearts of patients with COVID-19. Sci Transl Med 2021; 13:eabf7872. [PMID: 33723017 PMCID: PMC8128284 DOI: 10.1126/scitranslmed.abf7872] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/23/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022]
Abstract
Although coronavirus disease 2019 (COVID-19) causes cardiac dysfunction in up to 25% of patients, its pathogenesis remains unclear. Exposure of human induced pluripotent stem cell (iPSC)-derived heart cells to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) revealed productive infection and robust transcriptomic and morphological signatures of damage, particularly in cardiomyocytes. Transcriptomic disruption of structural genes corroborates adverse morphologic features, which included a distinct pattern of myofibrillar fragmentation and nuclear disruption. Human autopsy specimens from patients with COVID-19 reflected similar alterations, particularly sarcomeric fragmentation. These notable cytopathic features in cardiomyocytes provide insights into SARS-CoV-2-induced cardiac damage, offer a platform for discovery of potential therapeutics, and raise concerns about the long-term consequences of COVID-19 in asymptomatic and severe cases.
Collapse
Affiliation(s)
| | - Serah Kang
- Gladstone Institutes, San Francisco, CA 94158, USA
| | | | - Camille R Simoneau
- Gladstone Institutes, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David A Joy
- Gladstone Institutes, San Francisco, CA 94158, USA
- UC Berkeley-UCSF Joint Program in Bioengineering, Berkeley, CA 94720, USA
| | - Ana C Silva
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Gokul N Ramadoss
- Gladstone Institutes, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Will R Flanigan
- Gladstone Institutes, San Francisco, CA 94158, USA
- UC Berkeley-UCSF Joint Program in Bioengineering, Berkeley, CA 94720, USA
| | - Parinaz Fozouni
- Gladstone Institutes, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Huihui Li
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Pei-Yi Chen
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ken Nakamura
- Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Neurology, UCSF, San Francisco, CA 94143, USA
| | - Jeffrey D Whitman
- Department of Laboratory Medicine, UCSF, San Francisco, CA 94143, USA
| | - Paul J Hanson
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Bruce M McManus
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Melanie Ott
- Gladstone Institutes, San Francisco, CA 94158, USA.
- Department of Medicine, UCSF, San Francisco, CA 94143, USA
| | - Bruce R Conklin
- Gladstone Institutes, San Francisco, CA 94158, USA.
- Department of Medicine, UCSF, San Francisco, CA 94143, USA
- Innovative Genomics Institute, Berkeley, CA 94704, USA
- Department of Ophthalmology, UCSF, San Francisco, CA 94158, USA
| | - Todd C McDevitt
- Gladstone Institutes, San Francisco, CA 94158, USA.
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA 94158, USA
| |
Collapse
|
16
|
Greenberg MJ, Tardiff JC. Complexity in genetic cardiomyopathies and new approaches for mechanism-based precision medicine. J Gen Physiol 2021; 153:e202012662. [PMID: 33512404 PMCID: PMC7852459 DOI: 10.1085/jgp.202012662] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Genetic cardiomyopathies have been studied for decades, and it has become increasingly clear that these progressive diseases are more complex than originally thought. These complexities can be seen both in the molecular etiologies of these disorders and in the clinical phenotypes observed in patients. While these disorders can be caused by mutations in cardiac genes, including ones encoding sarcomeric proteins, the disease presentation varies depending on the patient mutation, where mutations even within the same gene can cause divergent phenotypes. Moreover, it is challenging to connect the mutation-induced molecular insult that drives the disease pathogenesis with the various compensatory and maladaptive pathways that are activated during the course of the subsequent progressive, pathogenic cardiac remodeling. These inherent complexities have frustrated our ability to understand and develop broadly effective treatments for these disorders. It has been proposed that it might be possible to improve patient outcomes by adopting a precision medicine approach. Here, we lay out a practical framework for such an approach, where patient subpopulations are binned based on common underlying biophysical mechanisms that drive the molecular disease pathogenesis, and we propose that this function-based approach will enable the development of targeted therapeutics that ameliorate these effects. We highlight several mutations to illustrate the need for mechanistic molecular experiments that span organizational and temporal scales, and we describe recent advances in the development of novel therapeutics based on functional targets. Finally, we describe many of the outstanding questions for the field and how fundamental mechanistic studies, informed by our more nuanced understanding of the clinical disorders, will play a central role in realizing the potential of precision medicine for genetic cardiomyopathies.
Collapse
Affiliation(s)
- Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
| | - Jil C. Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ
- Department of Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
17
|
Mishra S, Dunkerly-Eyring BL, Keceli G, Ranek MJ. Phosphorylation Modifications Regulating Cardiac Protein Quality Control Mechanisms. Front Physiol 2020; 11:593585. [PMID: 33281625 PMCID: PMC7689282 DOI: 10.3389/fphys.2020.593585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Many forms of cardiac disease, including heart failure, present with inadequate protein quality control (PQC). Pathological conditions often involve impaired removal of terminally misfolded proteins. This results in the formation of large protein aggregates, which further reduce cellular viability and cardiac function. Cardiomyocytes have an intricately collaborative PQC system to minimize cellular proteotoxicity. Increased expression of chaperones or enhanced clearance of misfolded proteins either by the proteasome or lysosome has been demonstrated to attenuate disease pathogenesis, whereas reduced PQC exacerbates pathogenesis. Recent studies have revealed that phosphorylation of key proteins has a potent regulatory role, both promoting and hindering the PQC machinery. This review highlights the recent advances in phosphorylations regulating PQC, the impact in cardiac pathology, and the therapeutic opportunities presented by harnessing these modifications.
Collapse
Affiliation(s)
- Sumita Mishra
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brittany L Dunkerly-Eyring
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
18
|
Mei ZL, Wang HB, Hu YH, Xiong L. CSN6 aggravates Ang II-induced cardiomyocyte hypertrophy via inhibiting SIRT2. Exp Cell Res 2020; 396:112245. [DOI: 10.1016/j.yexcr.2020.112245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022]
|
19
|
Zhang K, Chen L, Zhang Z, Cao J, He L, Li L. Ubiquitin-like protein FAT10: A potential cardioprotective factor and novel therapeutic target in cancer. Clin Chim Acta 2020; 510:802-811. [DOI: 10.1016/j.cca.2020.09.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
|
20
|
Kaur N, Raja R, Ruiz-Velasco A, Liu W. Cellular Protein Quality Control in Diabetic Cardiomyopathy: From Bench to Bedside. Front Cardiovasc Med 2020; 7:585309. [PMID: 33195472 PMCID: PMC7593653 DOI: 10.3389/fcvm.2020.585309] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
Heart failure is a serious comorbidity and the most common cause of mortality in diabetes patients. Diabetic cardiomyopathy (DCM) features impaired cellular structure and function, culminating in heart failure; however, there is a dearth of specific clinical therapy for treating DCM. Protein homeostasis is pivotal for the maintenance of cellular viability under physiological and pathological conditions, particularly in the irreplaceable cardiomyocytes; therefore, it is tightly regulated by a protein quality control (PQC) system. Three evolutionarily conserved molecular processes, the unfolded protein response (UPR), the ubiquitin-proteasome system (UPS), and autophagy, enhance protein turnover and preserve protein homeostasis by suppressing protein translation, degrading misfolded or unfolded proteins in cytosol or organelles, disposing of damaged and toxic proteins, recycling essential amino acids, and eliminating insoluble protein aggregates. In response to increased cellular protein demand under pathological insults, including the diabetic condition, a coordinated PQC system retains cardiac protein homeostasis and heart performance, on the contrary, inappropriate PQC function exaggerates cardiac proteotoxicity with subsequent heart dysfunction. Further investigation of the PQC mechanisms in diabetes propels a more comprehensive understanding of the molecular pathogenesis of DCM and opens new prospective treatment strategies for heart disease and heart failure in diabetes patients. In this review, the function and regulation of cardiac PQC machinery in diabetes mellitus, and the therapeutic potential for the diabetic heart are discussed.
Collapse
Affiliation(s)
- Namrita Kaur
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Rida Raja
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Andrea Ruiz-Velasco
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Wei Liu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
21
|
Oeing CU, Mishra S, Dunkerly-Eyring BL, Ranek MJ. Targeting Protein Kinase G to Treat Cardiac Proteotoxicity. Front Physiol 2020; 11:858. [PMID: 32848832 PMCID: PMC7399205 DOI: 10.3389/fphys.2020.00858] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
Impaired or insufficient protein kinase G (PKG) signaling and protein quality control (PQC) are hallmarks of most forms of cardiac disease, including heart failure. Their dysregulation has been shown to contribute to and exacerbate cardiac hypertrophy and remodeling, reduced cell survival and disease pathogenesis. Enhancement of PKG signaling and PQC are associated with improved cardiac function and survival in many pre-clinical models of heart disease. While many clinically used pharmacological approaches exist to stimulate PKG, there are no FDA-approved therapies to safely enhance cardiomyocyte PQC. The latter is predominantly due to our lack of knowledge and identification of proteins regulating cardiomyocyte PQC. Recently, multiple studies have demonstrated that PKG regulates PQC in the heart, both during physiological and pathological states. These studies tested already FDA-approved pharmacological therapies to activate PKG, which enhanced cardiomyocyte PQC and alleviated cardiac disease. This review examines the roles of PKG and PQC during disease pathogenesis and summarizes the experimental and clinical data supporting the utility of stimulating PKG to target cardiac proteotoxicity.
Collapse
Affiliation(s)
- Christian U Oeing
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, United States.,Department of Cardiology, Charité - University Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Sumita Mishra
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Brittany L Dunkerly-Eyring
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, United States
| |
Collapse
|
22
|
Wang X, Wang H. Priming the Proteasome to Protect against Proteotoxicity. Trends Mol Med 2020; 26:639-648. [PMID: 32589934 PMCID: PMC7321925 DOI: 10.1016/j.molmed.2020.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Increased proteotoxic stress (IPTS) resulting from the increased production or decreased removal of abnormally folded proteins is recognized as an important pathogenic factor for a large group of highly disabling and life-threatening human diseases, such as neurodegenerative disorders and many heart diseases. The proteasome is pivotal to the timely removal of abnormal proteins but its functional capacity often becomes inadequate in the disease conditions; consequently, proteasome functional insufficiency in return exacerbates IPTS. Recent research in proteasome biology reveals that the proteasome can be activated by endogenous protein kinases, making it possible to pharmacologically prime the proteasome for treating diseases with IPTS.
Collapse
Affiliation(s)
- Xuejun Wang
- University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA.
| | - Hongmin Wang
- University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| |
Collapse
|
23
|
Pan B, Li J, Parajuli N, Tian Z, Wu P, Lewno MT, Zou J, Wang W, Bedford L, Mayer RJ, Fang J, Liu J, Cui T, Su H, Wang X. The Calcineurin-TFEB-p62 Pathway Mediates the Activation of Cardiac Macroautophagy by Proteasomal Malfunction. Circ Res 2020; 127:502-518. [PMID: 32366200 DOI: 10.1161/circresaha.119.316007] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE The ubiquitin-proteasome system (UPS) and the autophagic-lysosomal pathway are pivotal to proteostasis. Targeting these pathways is emerging as an attractive strategy for treating cancer. However, a significant proportion of patients who receive a proteasome inhibitor-containing regime show cardiotoxicity. Moreover, UPS and autophagic-lysosomal pathway defects are implicated in cardiac pathogenesis. Hence, a better understanding of the cross-talk between the 2 catabolic pathways will help advance cardiac pathophysiology and medicine. OBJECTIVE Systemic proteasome inhibition (PSMI) was shown to increase p62/SQSTM1 expression and induce myocardial macroautophagy. Here we investigate how proteasome malfunction activates cardiac autophagic-lysosomal pathway. METHODS AND RESULTS Myocardial macroautophagy, TFEB (transcription factor EB) expression and activity, and p62 expression were markedly increased in mice with either cardiomyocyte-restricted ablation of Psmc1 (an essential proteasome subunit gene) or pharmacological PSMI. In cultured cardiomyocytes, PSMI-induced increases in TFEB activation and p62 expression were blunted by pharmacological and genetic calcineurin inhibition and by siRNA-mediated Molcn1 silencing. PSMI induced remarkable increases in myocardial autophagic flux in wild type mice but not p62 null (p62-KO) mice. Bortezomib-induced left ventricular wall thickening and diastolic malfunction was exacerbated by p62 deficiency. In cultured cardiomyocytes from wild type mice but not p62-KO mice, PSMI induced increases in LC3-II flux and the lysosomal removal of ubiquitinated proteins. Myocardial TFEB activation by PSMI as reflected by TFEB nuclear localization and target gene expression was strikingly less in p62-KO mice compared with wild type mice. CONCLUSIONS (1) The activation of cardiac macroautophagy by proteasomal malfunction is mediated by the Mocln1-calcineurin-TFEB-p62 pathway; (2) p62 unexpectedly exerts a feed-forward effect on TFEB activation by proteasome malfunction; and (3) targeting the Mcoln1 (mucolipin1)-calcineurin-TFEB-p62 pathway may provide new means to intervene cardiac autophagic-lysosomal pathway activation during proteasome malfunction.
Collapse
Affiliation(s)
- Bo Pan
- From the Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion (B.P., J. Li, N.P., Z.T., P.W., M.T.L., H.S., X.W.)
| | - Jie Li
- From the Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion (B.P., J. Li, N.P., Z.T., P.W., M.T.L., H.S., X.W.).,Vascular Biology Center and Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University (J. Li, J.Z., W.W., H.S.)
| | - Nirmal Parajuli
- From the Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion (B.P., J. Li, N.P., Z.T., P.W., M.T.L., H.S., X.W.)
| | - Zongwen Tian
- From the Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion (B.P., J. Li, N.P., Z.T., P.W., M.T.L., H.S., X.W.).,Department of Anatomy, Wuhan University College of Basic Medical Sciences, Hubei, China (Z.T.)
| | - Penglong Wu
- From the Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion (B.P., J. Li, N.P., Z.T., P.W., M.T.L., H.S., X.W.).,Guangzhou Institute of Oncology, Tumor Hospital, Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangdong, China (P.W., W.W., J. Liu)
| | - Megan T Lewno
- From the Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion (B.P., J. Li, N.P., Z.T., P.W., M.T.L., H.S., X.W.)
| | - Jianqiu Zou
- Vascular Biology Center and Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University (J. Li, J.Z., W.W., H.S.)
| | - Wenjuan Wang
- Vascular Biology Center and Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University (J. Li, J.Z., W.W., H.S.).,Guangzhou Institute of Oncology, Tumor Hospital, Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangdong, China (P.W., W.W., J. Liu)
| | - Lynn Bedford
- School of Life Sciences, University of Nottingham, United Kingdom (L.B.)
| | - R John Mayer
- The University of Nottingham Medical School, Queen's Medical Centre, United Kingdom (R.J.M.)
| | - Jing Fang
- Department of Drug Discovery and Biomedical Sciences (J.F.), University of South Carolina College of Pharmacy, Columbia
| | - Jinbao Liu
- Guangzhou Institute of Oncology, Tumor Hospital, Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangdong, China (P.W., W.W., J. Liu)
| | - Taixing Cui
- Department of Anatomy and Cell Biology (T.C.), University of South Carolina College of Pharmacy, Columbia
| | - Huabo Su
- From the Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion (B.P., J. Li, N.P., Z.T., P.W., M.T.L., H.S., X.W.).,Vascular Biology Center and Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University (J. Li, J.Z., W.W., H.S.)
| | - Xuejun Wang
- From the Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion (B.P., J. Li, N.P., Z.T., P.W., M.T.L., H.S., X.W.)
| |
Collapse
|
24
|
The Ubiquitin Proteasome System in Ischemic and Dilated Cardiomyopathy. Int J Mol Sci 2019; 20:ijms20246354. [PMID: 31861129 PMCID: PMC6940920 DOI: 10.3390/ijms20246354] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/21/2022] Open
Abstract
Dilated (DCM) and ischemic cardiomyopathies (ICM) are associated with cardiac remodeling, where the ubiquitin–proteasome system (UPS) holds a central role. Little is known about the UPS and its alterations in patients suffering from DCM or ICM. The aim of this study is to characterize the UPS activity in human heart tissue from cardiomyopathy patients. Myocardial tissue from ICM (n = 23), DCM (n = 28), and control (n = 14) patients were used to quantify ubiquitinylated proteins, E3-ubiquitin-ligases muscle-atrophy-F-box (MAFbx)/atrogin-1, muscle-RING-finger-1 (MuRF1), and eukaryotic-translation-initiation-factor-4E (eIF4E), by Western blot. Furthermore, the proteasomal chymotrypsin-like and trypsin-like peptidase activities were determined fluorometrically. Enzyme activity of NAD(P)H oxidase was assessed as an index of reactive oxygen species production. The chymotrypsin- (p = 0.71) and caspase-like proteasomal activity (p = 0.93) was similar between the groups. Trypsin-like proteasomal activity was lower in ICM (0.78 ± 0.11 µU/mg) compared to DCM (1.06 ± 0.08 µU/mg) and control (1.00 ± 0.06 µU/mg; p = 0.06) samples. Decreased ubiquitin expression in both cardiomyopathy groups (ICM vs. control: p < 0.001; DCM vs. control: p < 0.001), as well as less ubiquitin-positive deposits in ICM-damaged tissue (ICM: 4.19% ± 0.60%, control: 6.28% ± 0.40%, p = 0.022), were detected. E3-ligase MuRF1 protein expression (p = 0.62), NADPH-oxidase activity (p = 0.63), and AIF-positive cells (p = 0.50). Statistical trends were detected for reduced MAFbx protein expression in the DCM-group (p = 0.07). Different levels of UPS components, E3 ligases, and UPS activation markers were observed in myocardial tissue from patients affected by DCM and ICM, suggesting differential involvement of the UPS in the underlying pathologies.
Collapse
|
25
|
Wei L, Zhang Y, Qi X, Sun X, Li Y, Xu Y. Ubiquitin‑proteasomes are the dominant mediators of the regulatory effect of microRNA‑1 on cardiac remodeling after myocardial infarction. Int J Mol Med 2019; 44:1899-1907. [PMID: 31485642 PMCID: PMC6777676 DOI: 10.3892/ijmm.2019.4330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 08/20/2019] [Indexed: 12/25/2022] Open
Abstract
Patients with ischemic hearts who have refused coronary vascular reconstruction may exhibit dynamic myocardial remodeling and cardiac dysfunction. In the present study, the role of miRNA-1 and its association with the ubiquitin-proteasome system (UPS) in regulating myocardial remodeling was investigated. A myocardial infarction (MI) model was constructed and the hearts were treated with miRNA-1 antagomir, miRNA-1 lentiviral vectors and the UPS proteasome blocker bortezomib. The expression levels of miRNA-1 were evaluated using reverse transcription PCR and the abundance of the ubiquitin-proteasome protein and caspase-3 were evaluated via western blot analysis. Furthermore, the collagen volume fraction was calculated using Masson's trichrome staining, and the apoptosis index was detected via terminal deoxynucleotidyl transferase dUTP-biotin nick end labeling staining. Transforming growth factor (TGF)-β expression was assessed via immunohistochemical staining. Echocardiographic characteristics and myocardial infarct size were analyzed. miRNA-1 expression levels were found to be increased following MI. miRNA-1 antagomir administration clearly inhibited miRNA-1 expression, whereas the miRNA-1 lentiviral vector exerted the opposite effect. The levels of 19s proteasome, 20S proteasome and ubiquitin ligase E3 were decreased in the miRNA-1 antagomir group, but were significantly increased in the miRNA-1 lentiviral group; however, only 20S proteasome expression was decreased in the bortezomib group. Collagen hyperplasia and TGF-β expression were decreased in both the miRNA-1 antagomir and bortezomib groups, although the effects of the miRNA-1 antagomir were more noticeable. The miRNA-1 antagomir and the UPS proteasome blocker both alleviated the ultrastructural impairments, demonstrated by a decreased left ventricular (LV) end-diastolic diameter and LV mass, but the miRNA-1 antagomir was also able to increase LV ejection fraction and LV fractional shortening. miRNA-1 regulated UPS-associated mRNA expression and affected the majority of the UPS components in the myocardium, thereby leading to increased myocardial cell apoptosis, myocardial fibrosis and remodeling. The miRNA-1 antagomir exerted a more prominent cardioprotective effect compared with the UPS proteasome blocker bortezomib.
Collapse
Affiliation(s)
- Liping Wei
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin 300121, P.R. China
| | - Yufan Zhang
- School of Graduate Studies, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xin Qi
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin 300121, P.R. China
| | - Xuseng Sun
- School of Graduate Studies, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yuanyang Li
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Yue Xu
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| |
Collapse
|
26
|
Turkieh A, Porouchani S, Beseme O, Chwastyniak M, Amouyel P, Lamblin N, Balligand JL, Bauters C, Pinet F. Increased clusterin levels after myocardial infarction is due to a defect in protein degradation systems activity. Cell Death Dis 2019; 10:608. [PMID: 31406108 PMCID: PMC6691115 DOI: 10.1038/s41419-019-1857-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/22/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022]
Abstract
Clusterin (CLU) is induced in many organs after tissue injury or remodeling. Recently, we show that CLU levels are increased in plasma and left ventricle (LV) after MI, however, the mechanisms involved are not yet elucidated. On the other hand, it has been shown that the activity of the protein degradation systems (PDS) is affected after MI with a decrease in ubiquitin proteasome system (UPS) and an increase in macroautophagy. The aim of this study was to decipher if the increased CLU levels after MI are in part due to the alteration of PDS activity. Rat neonate cardiomyocytes (NCM) were treated with different modulators of UPS and macroautophagy in order to decipher their role in CLU expression, secretion, and degradation. We observed that inhibition of UPS activity in NCM increased CLU mRNA levels, its intracellular protein levels (p-CLU and m-CLU) and its secreted form (s-CLU). Macroautophagy was also induced after MG132 treatment but is not active. The inhibition of macroautophagy induction in MG132-treated NCM increased CLU mRNA and m-CLU levels, but not s-CLU compared to NCM only treated by MG132. We also demonstrate that CLU can be degraded in NCM through proteasome and lysosome by a macroautophagy independent pathway. In another hand, CLU silencing in NCM has no effect either on macroautophagy or apoptosis induced by MG132. However, the overexpression of CLU secreted isoform in H9c2 cells, but not in NCM decreased apoptosis after MG132 treatment. Finally, we observed that increased CLU levels in hypertrophied NCM and in failing human hearts are associated with proteasome inhibition and macroautophagy alteration. All these data suggest that increased CLU expression and secretion after MI is, in part, due to a defect of UPS and macroautophagy activities in the heart and may have a protective effect by decreasing apoptosis induced by proteasome inhibition.
Collapse
Affiliation(s)
- Annie Turkieh
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France.,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France
| | - Sina Porouchani
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France.,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France
| | - Olivia Beseme
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France
| | - Maggy Chwastyniak
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France
| | - Philippe Amouyel
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France
| | - Nicolas Lamblin
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France.,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France
| | - Jean-Luc Balligand
- Institut de Recherche Experimentale et Clinique, Pole of Pharmacology and Therapeutics and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Christophe Bauters
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France.,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France
| | - Florence Pinet
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France. .,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France.
| |
Collapse
|
27
|
Lino CA, Demasi M, Barreto-Chaves ML. Ubiquitin proteasome system (UPS) activation in the cardiac hypertrophy of hyperthyroidism. Mol Cell Endocrinol 2019; 493:110451. [PMID: 31112742 DOI: 10.1016/j.mce.2019.110451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/13/2019] [Accepted: 05/17/2019] [Indexed: 11/16/2022]
Abstract
Ubiquitin proteasome system (UPS) is the main proteolytic pathway in eukaryotic cells. Changes in proteasome expression and activity have been associated to cardiovascular diseases as cardiac hypertrophy. Considering that cardiac hypertrophy is commonly associated to hyperthyroidism condition, the present study aimed to investigate the contribution of UPS in cardiac hypertrophy induced by thyroid hormones. Hyperthyroidism was induced in male Wistar rats by intraperitoneal injections of triiodothyronine (T3; 7 μg/100 g of body weight) for 7 days and confirmed by raised levels of total T3 and decreased levels of total T4. In addition, systolic blood pressure and heart rate were significantly increased in hyperthyroid group. Cardiac hypertrophy was confirmed in hyperthyroid group by increased heart weight/tibia length ratio and by increased α-MHC/β-MHC relative expression. Both catalytic (20SPT) and regulatory subunits (19SPT) of the constitutive proteasome were upregulated in hyperthyroid hearts. In addition, the transcripts that encode immunoproteasome subunits were also elevated. Furthermore, ATP-dependent chymotrypsin-like activity (26SPT) was significantly increased in hyperthyroid group. Despite the upregulation and activation of UPS in hyperthyroid hearts, the content of polyubiquitinated proteins was unaltered in relation to control. Together, these results evidence the activation of cardiac proteasome by thyroid hormones, which possibly contribute to the maintenance of protein quality control and regulation of cardiac hypertrophy in response to thyroid hormones.
Collapse
Affiliation(s)
- Caroline Antunes Lino
- Laboratory of Cellular Biology and Functional Anatomy, Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Marilene Demasi
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Sao Paulo, Brazil
| | - Maria Luiza Barreto-Chaves
- Laboratory of Cellular Biology and Functional Anatomy, Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
28
|
Dorsch LM, Schuldt M, dos Remedios CG, Schinkel AFL, de Jong PL, Michels M, Kuster DWD, Brundel BJJM, van der Velden J. Protein Quality Control Activation and Microtubule Remodeling in Hypertrophic Cardiomyopathy. Cells 2019; 8:E741. [PMID: 31323898 PMCID: PMC6678711 DOI: 10.3390/cells8070741] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 12/14/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disorder. It is mainly caused by mutations in genes encoding sarcomere proteins. Mutant forms of these highly abundant proteins likely stress the protein quality control (PQC) system of cardiomyocytes. The PQC system, together with a functional microtubule network, maintains proteostasis. We compared left ventricular (LV) tissue of nine donors (controls) with 38 sarcomere mutation-positive (HCMSMP) and 14 sarcomere mutation-negative (HCMSMN) patients to define HCM and mutation-specific changes in PQC. Mutations in HCMSMP result in poison polypeptides or reduced protein levels (haploinsufficiency, HI). The main findings were 1) several key PQC players were more abundant in HCM compared to controls, 2) after correction for sex and age, stabilizing heat shock protein (HSP)B1, and refolding, HSPD1 and HSPA2 were increased in HCMSMP compared to controls, 3) α-tubulin and acetylated α-tubulin levels were higher in HCM compared to controls, especially in HCMHI, 4) myosin-binding protein-C (cMyBP-C) levels were inversely correlated with α-tubulin, and 5) α-tubulin levels correlated with acetylated α-tubulin and HSPs. Overall, carrying a mutation affects PQC and α-tubulin acetylation. The haploinsufficiency of cMyBP-C may trigger HSPs and α-tubulin acetylation. Our study indicates that proliferation of the microtubular network may represent a novel pathomechanism in cMyBP-C haploinsufficiency-mediated HCM.
Collapse
Affiliation(s)
- Larissa M Dorsch
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| | - Maike Schuldt
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Cristobal G dos Remedios
- Sydney Heart Bank, Discipline of Anatomy, Bosch Institute, University of Sydney, Sydney 2006, Australia
| | - Arend F L Schinkel
- Department of Cardiology, Thoraxcenter, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Peter L de Jong
- Department of Cardiothoracic Surgery, Thoraxcenter, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Michelle Michels
- Department of Cardiology, Thoraxcenter, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Diederik W D Kuster
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
- Netherlands Heart Institute, 3511 EP Utrecht, The Netherlands
| |
Collapse
|
29
|
Zhang H, Pan B, Wu P, Parajuli N, Rekhter MD, Goldberg AL, Wang X. PDE1 inhibition facilitates proteasomal degradation of misfolded proteins and protects against cardiac proteinopathy. SCIENCE ADVANCES 2019; 5:eaaw5870. [PMID: 31131329 PMCID: PMC6531002 DOI: 10.1126/sciadv.aaw5870] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/11/2019] [Indexed: 06/09/2023]
Abstract
No current treatment targets cardiac proteotoxicity or can reduce mortality of heart failure (HF) with preserved ejection fraction (HFpEF). Selective degradation of misfolded proteins by the ubiquitin-proteasome system (UPS) is vital to the cell. Proteasome impairment contributes to HF. Activation of cAMP-dependent protein kinase (PKA) or cGMP-dependent protein kinase (PKG) facilitates proteasome functioning. Phosphodiesterase 1 (PDE1) hydrolyzes both cyclic nucleotides and accounts for most PDE activities in human myocardium. We report that PDE1 inhibition (IC86430) increases myocardial 26S proteasome activities and UPS proteolytic function in mice. Mice with CryABR120G-based proteinopathy develop HFpEF and show increased myocardial PDE1A expression. PDE1 inhibition markedly attenuates HFpEF, improves mouse survival, increases PKA-mediated proteasome phosphorylation, and reduces myocardial misfolded CryAB. Therefore, PDE1 inhibition induces PKA- and PKG-mediated promotion of proteasomal degradation of misfolded proteins and treats HFpEF caused by CryABR120G, representing a potentially new therapeutic strategy for HFpEF and heart disease with increased proteotoxic stress.
Collapse
Affiliation(s)
- Hanming Zhang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Bo Pan
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Penglong Wu
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
- Department of Pathophysiology, Guangzhou Medical University College of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| | - Nirmal Parajuli
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Mark D. Rekhter
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Alfred L. Goldberg
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| |
Collapse
|
30
|
Shukla SK, Rafiq K. Proteasome biology and therapeutics in cardiac diseases. Transl Res 2019; 205:64-76. [PMID: 30342797 PMCID: PMC6372329 DOI: 10.1016/j.trsl.2018.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 08/30/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
Abstract
The ubiquitin proteasome system (UPS) is the major pathway for intracellular protein degradation in most organs, including the heart. UPS controls many fundamental biological processes such as cell cycle, cell division, immune responses, antigen presentation, apoptosis, and cell signaling. The UPS not only degrades substrates but also regulates activity of gene transcription at the post-transcription level. Emerging evidence suggests that impairment of UPS function is sufficient to cause a number of cardiac diseases, including heart failure, cardiomyopathies, hypertrophy, atrophy, ischemia-reperfusion, and atherosclerosis. Alterations in the expression of UPS components, changes in proteasomal peptidase activities and increased ubiquitinated and oxidized proteins have also been detected in diabetic cardiomyopathy (DCM). However, the pathophysiological role of the UPS in DCM has not been examined. Recently, in vitro and in vivo studies have proven highly valuable in assessing effects of various stressors on the UPS and, in some cases, suggesting a causal link between defective protein clearance and disease phenotypes in different cardiac diseases, including DCM. Translation of these findings to human disease can be greatly strengthened by corroboration of discoveries from experimental model systems using human heart tissue from well-defined patient populations. This review will summarize the general role of the UPS in different cardiac diseases, with major focus on DCM, and on recent advances in therapeutic development.
Collapse
Affiliation(s)
- Sanket Kumar Shukla
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Khadija Rafiq
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
31
|
Heckmann MB, Doroudgar S, Katus HA, Lehmann LH. Cardiovascular adverse events in multiple myeloma patients. J Thorac Dis 2018; 10:S4296-S4305. [PMID: 30701098 PMCID: PMC6328391 DOI: 10.21037/jtd.2018.09.87] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022]
Abstract
Multiple myeloma is a malignant disease, caused by an uncontrolled clonal proliferation of a specific group of white blood cells, the plasma cells. Clinical manifestations include bone pain due to osteolysis, hypercalcemia, anemia, and renal insufficiency. Proteasome inhibitors have substantially improved survival of patients suffering from multiple myeloma, providing an efficient treatment option mainly for relapsed and refractory multiple myeloma. Although constituting one substance class, bortezomib, carfilzomib, and ixazomib differ greatly regarding their non-hematologic side effects. This article reviews the clinical and preclinical data on approved proteasome inhibitors in an attempt to decipher the underlying pathomechanisms related to cardiovascular adverse events seen in clinical trials.
Collapse
Affiliation(s)
- Markus B. Heckmann
- Department of Cardiology, Angiology, and Pneumology, Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Shirin Doroudgar
- Department of Cardiology, Angiology, and Pneumology, Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Hugo A. Katus
- Department of Cardiology, Angiology, and Pneumology, Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Lorenz H. Lehmann
- Department of Cardiology, Angiology, and Pneumology, Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
32
|
Affiliation(s)
| | - Rosa Barrio
- CIC bioGUNE, Bizkaia Technology Park, Derio, Spain
| |
Collapse
|
33
|
Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding Key Mechanisms of Exercise-Induced Cardiac Protection to Mitigate Disease: Current Knowledge and Emerging Concepts. Physiol Rev 2018; 98:419-475. [PMID: 29351515 DOI: 10.1152/physrev.00043.2016] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The benefits of exercise on the heart are well recognized, and clinical studies have demonstrated that exercise is an intervention that can improve cardiac function in heart failure patients. This has led to significant research into understanding the key mechanisms responsible for exercise-induced cardiac protection. Here, we summarize molecular mechanisms that regulate exercise-induced cardiac myocyte growth and proliferation. We discuss in detail the effects of exercise on other cardiac cells, organelles, and systems that have received less or little attention and require further investigation. This includes cardiac excitation and contraction, mitochondrial adaptations, cellular stress responses to promote survival (heat shock response, ubiquitin-proteasome system, autophagy-lysosomal system, endoplasmic reticulum unfolded protein response, DNA damage response), extracellular matrix, inflammatory response, and organ-to-organ crosstalk. We summarize therapeutic strategies targeting known regulators of exercise-induced protection and the challenges translating findings from bench to bedside. We conclude that technological advancements that allow for in-depth profiling of the genome, transcriptome, proteome and metabolome, combined with animal and human studies, provide new opportunities for comprehensively defining the signaling and regulatory aspects of cell/organelle functions that underpin the protective properties of exercise. This is likely to lead to the identification of novel biomarkers and therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Jenny Y Y Ooi
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Natalie L Patterson
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| |
Collapse
|
34
|
Glazier AA, Hafeez N, Mellacheruvu D, Basrur V, Nesvizhskii AI, Lee LM, Shao H, Tang V, Yob JM, Gestwicki JE, Helms AS, Day SM. HSC70 is a chaperone for wild-type and mutant cardiac myosin binding protein C. JCI Insight 2018; 3:99319. [PMID: 29875314 PMCID: PMC6124431 DOI: 10.1172/jci.insight.99319] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/01/2018] [Indexed: 11/17/2022] Open
Abstract
Cardiac myosin binding protein C (MYBPC3) is the most commonly mutated gene associated with hypertrophic cardiomyopathy (HCM). Haploinsufficiency of full-length MYBPC3 and disruption of proteostasis have both been proposed as central to HCM disease pathogenesis. Discriminating the relative contributions of these 2 mechanisms requires fundamental knowledge of how turnover of WT and mutant MYBPC3 proteins is regulated. We expressed several disease-causing mutations in MYBPC3 in primary neonatal rat ventricular cardiomyocytes. In contrast to WT MYBPC3, mutant proteins showed reduced expression and failed to localize to the sarcomere. In an unbiased coimmunoprecipitation/mass spectrometry screen, we identified HSP70-family chaperones as interactors of both WT and mutant MYBPC3. Heat shock cognate 70 kDa (HSC70) was the most abundant chaperone interactor. Knockdown of HSC70 significantly slowed degradation of both WT and mutant MYBPC3, while pharmacologic activation of HSC70 and HSP70 accelerated degradation. HSC70 was expressed in discrete striations in the sarcomere. Expression of mutant MYBPC3 did not affect HSC70 localization, nor did it induce a protein folding stress response or ubiquitin proteasome dysfunction. Together these data suggest that WT and mutant MYBPC3 proteins are clients for HSC70, and that the HSC70 chaperone system plays a major role in regulating MYBPC3 protein turnover.
Collapse
Affiliation(s)
| | | | | | | | | | - Lap Man Lee
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Hao Shao
- Institute for Neurodegenerative Diseases, UCSF, San Francisco, California, USA
| | - Vi Tang
- Department of Molecular and Integrative Physiology
| | | | - Jason E. Gestwicki
- Institute for Neurodegenerative Diseases, UCSF, San Francisco, California, USA
| | | | - Sharlene M. Day
- Department of Molecular and Integrative Physiology
- Department of Internal Medicine
| |
Collapse
|
35
|
Byrne CD, Perseghin G. Non-alcoholic fatty liver disease: A risk factor for myocardial dysfunction? J Hepatol 2018; 68:640-642. [PMID: 29237547 DOI: 10.1016/j.jhep.2017.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 12/04/2017] [Indexed: 01/31/2023]
Affiliation(s)
- Christopher D Byrne
- Nutrition and Metabolism, Faculty of Medicine, Institute of Developmental Sciences (IDS), MP887, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; Southampton National Institute for Health Research Biomedical Research Centre, University Hospital Southampton, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK.
| | - Gianluca Perseghin
- Department of Medicine and Surgery, Università degli Studi di Milano Bicocca, Milan, Italy; Department of Medicine and Rehabilitation, Unit of Metabolic Medicine, Policlinico di Monza, Monza, Italy
| |
Collapse
|
36
|
Lee YH, Kim KJ, Yoo ME, Kim G, Yoon HJ, Jo K, Youn JC, Yun M, Park JY, Shim CY, Lee BW, Kang SM, Ha JW, Cha BS, Kang ES. Association of non-alcoholic steatohepatitis with subclinical myocardial dysfunction in non-cirrhotic patients. J Hepatol 2018; 68:764-772. [PMID: 29175242 DOI: 10.1016/j.jhep.2017.11.023] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/24/2017] [Accepted: 11/13/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) is associated with increased cardiovascular risk. Among categories of NAFLD, hepatic fibrosis is most likely to affect mortality. Myocardial function and its energy metabolism are tightly linked, which might be altered by an insulin resistant condition such as NAFLD. We investigated whether hepatic steatosis and fibrosis were associated with myocardial dysfunction relative to myocardial glucose uptake. METHODS A total of 308 patients (190 without NAFLD, 118 with NAFLD) were studied in a tertiary care hospital. Myocardial glucose uptake was evaluated at fasted state using [18F]-fluorodeoxyglucose-positron emission tomography (18FDG-PET). Hepatic steatosis and fibrosis were assessed by transient liver elastography (Fibroscan®) with controlled attenuation parameter, which quantifies hepatic fat and by surrogate indices (fatty liver index and NAFLD fibrosis score). Cardiac structure and function were examined by echocardiogram. RESULTS Compared to those without NAFLD, patients with NAFLD had alterations in cardiac remodeling, manifested by increased left ventricular mass index, left ventricular end-diastolic diameter, and left atrial volume index (all p <0.05). Hepatic steatosis was significantly associated with left ventricular filling pressure (E/e' ratio), which reflects diastolic dysfunction (p for trend <0.05). Those without NAFLD were more likely to have higher myocardial glucose uptake compared to those with NAFLD. Significant hepatic fibrosis was also correlated with diastolic dysfunction and impaired myocardial glucose uptake. Using multivariable linear regression, E/e' ratio was independently associated with hepatic fibrosis (standardized β = 0.12 to 0.27; all p <0.05). Association between hepatic steatosis and E/e' ratio was also significant (standardized β = 0.10 to 0.15; all p <0.05 excluding the model adjusted for adiposity). CONCLUSIONS Hepatic steatosis and fibrosis are significantly associated with diastolic heart dysfunction. This association is linked with myocardial glucose uptake evaluated by 18FDG-PET. LAY SUMMARY Non-alcoholic fatty liver disease is associated with an increased risk of cardiovascular disease. More severe forms of non-alcoholic fatty liver disease, where hepatic fibrosis occurs, are linked to increased mortality. In this study, we have shown that hepatic steatosis and fibrosis are associated with subclinical myocardial dysfunction. This association is linked to altered myocardial glucose uptake.
Collapse
Affiliation(s)
- Yong-Ho Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kwang Joon Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myung Eun Yoo
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gyuri Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye-Jin Yoon
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kwanhyeong Jo
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong-Chan Youn
- Division of Cardiology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea; Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chi Young Shim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung-Wan Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seok-Min Kang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong-Won Ha
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bong-Soo Cha
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Seok Kang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
37
|
Wang ZG, Li H, Huang Y, Li R, Wang XF, Yu LX, Guang XQ, Li L, Zhang HY, Zhao YZ, Zhang C, Li XK, Wu RZ, Chu MP, Xiao J. Nerve growth factor-induced Akt/mTOR activation protects the ischemic heart via restoring autophagic flux and attenuating ubiquitinated protein accumulation. Oncotarget 2018; 8:5400-5413. [PMID: 28036273 PMCID: PMC5354918 DOI: 10.18632/oncotarget.14284] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 12/06/2016] [Indexed: 01/06/2023] Open
Abstract
The dysregulation of autophagy is related to a variety of cardiovascular diseases, such as myocardial ischemia/reperfusion (I/R). Nerve growth factor (NGF) has been shown to have therapeutic potential in ischaemic heart injury. In this study, we demonstrate that NGF administration can accelerate autophagic flux and attenuate protein ubiquitination in myocardial I/R heart. Our results showed that NGF could restored heart function and decreased the apoptosis of cardiomyocytes which induced by myocardial I/R injury. The protective effect of NGF is associated with the inhibition of autophagy related proteins. On another hand, NGF enhances the clearance of ubiquitinated protein and increases the survival of myocardial cell in vivo and in vitro. Additionally, NGF could activate the PI3K/AKT and mTOR signaling pathways. These results suggested that the cardioprotective effect of NGF is related to the restoration of autophagic flux and attenuation of protein ubiquitination via the activation of PI3K/AKT and mTOR pathway.
Collapse
Affiliation(s)
- Zhou-Guang Wang
- Institute of Cardiovascular Development and Translational Medicine, Children's Heart Center, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China.,Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China.,Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Jilin University, Changchun, 130012, China
| | - Hao Li
- Institute of Cardiovascular Development and Translational Medicine, Children's Heart Center, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yan Huang
- Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China
| | - Rui Li
- Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiao-Fan Wang
- Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China
| | - Li-Xia Yu
- Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China
| | - Xue-Qiang Guang
- Institute of Cardiovascular Development and Translational Medicine, Children's Heart Center, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Lei Li
- Institute of Cardiovascular Development and Translational Medicine, Children's Heart Center, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hong-Yu Zhang
- Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China
| | - Ying-Zheng Zhao
- Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China
| | - Chunxiang Zhang
- Institute of Cardiovascular Development and Translational Medicine, Children's Heart Center, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xiao-Kun Li
- Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China.,Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Jilin University, Changchun, 130012, China
| | - Rong-Zhou Wu
- Institute of Cardiovascular Development and Translational Medicine, Children's Heart Center, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Mao-Ping Chu
- Institute of Cardiovascular Development and Translational Medicine, Children's Heart Center, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jian Xiao
- Institute of Cardiovascular Development and Translational Medicine, Children's Heart Center, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China.,Molecular Pharmacology Research Center, School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
38
|
Hauck L, Stanley-Hasnain S, Fung A, Grothe D, Rao V, Mak TW, Billia F. Cardiac-specific ablation of the E3 ubiquitin ligase Mdm2 leads to oxidative stress, broad mitochondrial deficiency and early death. PLoS One 2017; 12:e0189861. [PMID: 29267372 PMCID: PMC5739440 DOI: 10.1371/journal.pone.0189861] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 12/04/2017] [Indexed: 12/15/2022] Open
Abstract
The maintenance of normal heart function requires proper control of protein turnover. The ubiquitin-proteasome system is a principal regulator of protein degradation. Mdm2 is the main E3 ubiquitin ligase for p53 in mitotic cells thereby regulating cellular growth, DNA repair, oxidative stress and apoptosis. However, which of these Mdm2-related activities are preserved in differentiated cardiomyocytes has yet to be determined. We sought to elucidate the role of Mdm2 in the control of normal heart function. We observed markedly reduced Mdm2 mRNA levels accompanied by highly elevated p53 protein expression in the hearts of wild type mice subjected to myocardial infarction or trans-aortic banding. Accordingly, we generated conditional cardiac-specific Mdm2 gene knockout (Mdm2f/f;mcm) mice. In adulthood, Mdm2f/f;mcm mice developed spontaneous cardiac hypertrophy, left ventricular dysfunction with early mortality post-tamoxifen. A decreased polyubiquitination of myocardial p53 was observed, leading to its stabilization and activation, in the absence of acute stress. In addition, transcriptomic analysis of Mdm2-deficient hearts revealed that there is an induction of E2f1 and c-Myc mRNA levels with reduced expression of the Pgc-1a/Ppara/Esrrb/g axis and Pink1. This was associated with a significant degree of cardiomyocyte apoptosis, and an inhibition of redox homeostasis and mitochondrial bioenergetics. All these processes are early, Mdm2-associated events and contribute to the development of pathological hypertrophy. Our genetic and biochemical data support a role for Mdm2 in cardiac growth control through the regulation of p53, the Pgc-1 family of transcriptional coactivators and the pivotal antioxidant Pink1.
Collapse
Affiliation(s)
- Ludger Hauck
- Toronto General Research Institute, Toronto, Ontario, Canada
| | | | - Amelia Fung
- Toronto General Research Institute, Toronto, Ontario, Canada
| | - Daniela Grothe
- Toronto General Research Institute, Toronto, Ontario, Canada
| | - Vivek Rao
- Division of Cardiovascular Surgery, UHN, Toronto, Ontario, Canada
| | - Tak W. Mak
- Campbell Family Cancer Research Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Filio Billia
- Toronto General Research Institute, Toronto, Ontario, Canada
- Division of Cardiology, University Health Network (UHN), Toronto, Ontario, Canada
- Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario Canada
- * E-mail:
| |
Collapse
|
39
|
Yokoe S, Asahi M. Phospholamban Is Downregulated by pVHL-Mediated Degradation through Oxidative Stress in Failing Heart. Int J Mol Sci 2017; 18:ijms18112232. [PMID: 29068413 PMCID: PMC5713202 DOI: 10.3390/ijms18112232] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/20/2017] [Accepted: 10/20/2017] [Indexed: 12/04/2022] Open
Abstract
The E3 ubiquitin ligase, von Hippel–Lindau (VHL), regulates protein expression by polyubiquitination. Although the protein VHL (pVHL) was reported to be involved in the heart function, the underlying mechanism is unclear. Here, we show that pVHL was upregulated in hearts from two types of genetically dilated cardiomyopathy (DCM) mice models. In comparison with the wild-type mouse, both DCM mice models showed a significant reduction in the expression of phospholamban (PLN), a potent inhibitor of sarco(endo)plasmic reticulum Ca2+-ATPase, and enhanced interaction between pVHL and PLN. To clarify whether pVHL is involved in PLN degradation in failing hearts, we used carbonylcyanide m-chlorophenylhydrazone (CCCP), a mitochondrial membrane potential (MMP)-lowering reagent, to mimic the heart failure condition in PLN-expressing HEK293 cells and found that CCCP treatment resulted in PLN degradation and increased interaction between PLN and pVHL. However, these effects were reversed with the addition of N-acetyl-l-cysteine. Furthermore, the co-transfection of VHL and PLN in HEK293 cells decreased PLN expression under oxidative stress, whereas knockdown of VHL increased PLN expression both under normal and oxidative stress conditions. Together, we propose that oxidative stress upregulates pVHL expression to induce PLN degradation in failing hearts.
Collapse
Affiliation(s)
- Shunichi Yokoe
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| |
Collapse
|
40
|
Abstract
The incidence and prevalence of cardiac diseases, which are the main cause of death worldwide, are likely to increase because of population ageing. Prevailing theories about the mechanisms of ageing feature the gradual derailment of cellular protein homeostasis (proteostasis) and loss of protein quality control as central factors. In the heart, loss of protein patency, owing to flaws in genetically-determined design or because of environmentally-induced 'wear and tear', can overwhelm protein quality control, thereby triggering derailment of proteostasis and contributing to cardiac ageing. Failure of protein quality control involves impairment of chaperones, ubiquitin-proteosomal systems, autophagy, and loss of sarcomeric and cytoskeletal proteins, all of which relate to induction of cardiomyocyte senescence. Targeting protein quality control to maintain cardiac proteostasis offers a novel therapeutic strategy to promote cardiac health and combat cardiac disease. Currently marketed drugs are available to explore this concept in the clinical setting.
Collapse
Affiliation(s)
- Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, De Boelelaan 1117, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
41
|
Abstract
In most patients with chronic heart failure (HF), levels of circulating cytokines are elevated and the elevated cytokine levels correlate with the severity of HF and prognosis. Various stresses induce subcellular component abnormalities, such as mitochondrial damage. Damaged mitochondria induce accumulation of reactive oxygen species and apoptogenic proteins, and subcellular inflammation. The vicious cycle of subcellular component abnormalities, inflammatory cell infiltration and neurohumoral activation induces cardiomyocyte injury and death, and cardiac fibrosis, resulting in cardiac dysfunction and HF. Quality control mechanisms at both the protein and organelle levels, such as elimination of apoptogenic proteins and damaged mitochondria, maintain cellular homeostasis. An imbalance between protein synthesis and degradation is likely to result in cellular dysfunction and disease. Three major protein degradation systems have been identified, namely the cysteine protease system, autophagy, and the ubiquitin proteasome system. Autophagy was initially believed to be a non-selective process. However, recent studies have described the process of selective mitochondrial autophagy, known as mitophagy. Elimination of damaged mitochondria by autophagy is important for maintenance of cellular homeostasis. DNA and RNA degradation systems also play a critical role in regulating inflammation and maintaining cellular homeostasis mediated by damaged DNA clearance and post-transcriptional regulation, respectively. This review discusses some recent advances in understanding the role of sterile inflammation and degradation systems in HF.
Collapse
Affiliation(s)
- Kazuhiko Nishida
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence
| | - Kinya Otsu
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence
| |
Collapse
|
42
|
Gilda JE, Gomes AV. Proteasome dysfunction in cardiomyopathies. J Physiol 2017; 595:4051-4071. [PMID: 28181243 DOI: 10.1113/jp273607] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a critical role in removing unwanted intracellular proteins and is involved in protein quality control, signalling and cell death. Because the heart is subject to continuous metabolic and mechanical stress, the proteasome plays a particularly important role in the heart, and proteasome dysfunction has been suggested as a causative factor in cardiac dysfunction. Proteasome impairment has been detected in cardiomyopathies, heart failure, myocardial ischaemia, and hypertrophy. Proteasome inhibition is also sufficient to cause cardiac dysfunction in healthy pigs, and patients using a proteasome inhibitor for cancer therapy have a higher incidence of heart failure. In this Topical Review we discuss the experimental data which suggest UPS dysfunction is a common feature of cardiomyopathies, with an emphasis on hypertrophic cardiomyopathy caused by sarcomeric mutations. We also propose potential mechanisms by which cardiomyopathy-causing mutations may lead to proteasome impairment, such as altered calcium handling and increased oxidative stress due to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jennifer E Gilda
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA.,Department of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| |
Collapse
|
43
|
Mohamed IA, Krishnamoorthy NT, Nasrallah GK, Da'as SI. The Role of Cardiac Myosin Binding Protein C3 in Hypertrophic Cardiomyopathy-Progress and Novel Therapeutic Opportunities. J Cell Physiol 2017; 232:1650-1659. [PMID: 27731493 DOI: 10.1002/jcp.25639] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/07/2016] [Indexed: 11/11/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a common autosomal dominant genetic cardiovascular disorder marked by genetic and phenotypic heterogeneity. Mutations in the gene encodes the cardiac myosin-binding protein C, cMYBPC3 is amongst the various sarcomeric genes that are associated with HCM. These mutations produce mutated mRNAs and truncated cMyBP-C proteins. In this review, we will discuss the implications and molecular mechanisms involved in MYBPC3 different mutations. Further, we will highlight the novel targets that can be developed into potential therapeutics for the treatment of HMC. J. Cell. Physiol. 232: 1650-1659, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Iman A Mohamed
- Department of Biomedical Science, Zewail City of Science and Technology, Giza, Egypt
| | - Navaneethakrishnan T Krishnamoorthy
- Division of Experimental Genetics, Sidra Medical and Research Center, Doha, Qatar.,Heart Science Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Gheyath K Nasrallah
- Department of Biomedical Science, College of Health Science, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| | - Sahar I Da'as
- Division of Experimental Genetics, Sidra Medical and Research Center, Doha, Qatar.,Department of Biomedical and Biological Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
44
|
Association of LIM Domain 7 Gene Polymorphisms and Plasma Levels of LIM Domain 7 with Dilated Cardiomyopathy in a Chinese Population. Appl Biochem Biotechnol 2016; 182:885-897. [PMID: 27988857 DOI: 10.1007/s12010-016-2368-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/12/2016] [Indexed: 02/05/2023]
Abstract
The aim of our study was to investigate the potential association of mRNA expression and plasma levels of the LIM domain 7 (LMO7) gene with the pathogenesis of dilated cardiomyopathy (DCM). Two SNPs of the LMO7 gene were genotyped in 310 patients with DCM and 415 controls. Our results showed that SNP rs7986131 (p = 0.002, OR = 1.38, 95% CI = 1.12-1.71), but not SNP rs4884021, was associated with DCM in the Han Chinese population. Haplotype analysis showed that the haplotype GT was associated with increased DCM susceptibility while AC was a protective haplotype. The Cox multivariate survival analysis indicated that the rs7986131 TT genotype (HR 1.659, 95% CI = 1.122-2.454, p = 0.011) was an independent multivariate predictor for shorter overall survival in patients with DCM. LMO7 mRNA expression and plasma LMO7 levels were significantly decreased in DCM (p < 0.0001). Spearman correlation test revealed that the plasma LMO7 level was negatively associated with left ventricular end-diastolic diameter (r = -0.384, p = 0.01), left ventricular end-diastolic volume (r = -0.375, p = 0.012), and brain natriuretic peptide (r = -0.482, p = 0.001). Our study suggested that the LMO7 gene may play an important role in the pathogenesis of DCM in the Han Chinese population.
Collapse
|
45
|
HSP70: therapeutic potential in acute and chronic cardiac disease settings. Future Med Chem 2016; 8:2177-2183. [DOI: 10.4155/fmc-2016-0192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Heat shock proteins are a family of proteins that are produced by cells in response to exposure to stressful conditions. The best studied heat shock protein is HSP70, which is known to act as a molecular chaperone to maintain cellular homeostasis and inhibit protein aggregation in response to stress. While early animal studies suggested that increasing HSP70 in the heart (using a transgenic, gene transfer or pharmacological approach) provided cardiac protection against acute cardiac stress, recent studies have found no benefit of increasing HSP70 in mouse models of chronic cardiac stress. As HSP70 has been considered a potential therapeutic target, it is important to comprehensively assess HSP70 therapies in preclinical models of acute and chronic cardiac disease.
Collapse
|
46
|
Zhang Y, Aguilar OA, Storey KB. Transcriptional activation of muscle atrophy promotes cardiac muscle remodeling during mammalian hibernation. PeerJ 2016; 4:e2317. [PMID: 27602284 PMCID: PMC4991874 DOI: 10.7717/peerj.2317] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/12/2016] [Indexed: 12/12/2022] Open
Abstract
Background. Mammalian hibernation in thirteen-lined ground squirrels (Ictidomys tridecemlineatus) is characterized by dramatic changes on a physiological and molecular level. During hibernation, mammalian hearts show a propensity to hypertrophy due to the need for increasing contractility to pump colder and more viscous blood. While cardiac hypertrophy is quite often a process characterized by decompensation, the ground squirrel studied is an excellent model of cardiac plasticity and cardioprotection under conditions of hypothermia and ischemia. The forkhead box O (Foxo) family of proteins and myogenin (MyoG) are transcription factors that control protein degradation and muscle atrophy by regulating the expression of the E3 ubiquitin ligases, MAFbx and MuRF1. These ligases are part of the ubiquitin proteasome system by transferring ubiquitin to proteins and targeting these proteins for degradation. Regulation of Foxo1 and 3a occurs through phosphorylation at different residues. The threonine-24 (Thr-24) and serine-319 (Ser-319) residues on Foxo1, and the Thr-32 residue on Foxo3a are phosphorylated by Akt, leading to cytoplasmic localization of Foxo. We propose that the described mechanism contributes to the changes taking place in cardiac muscle throughout hibernation. Methods. Total and phosphorylated protein levels of Foxo1 and Foxo3a, as well as total protein levels of MyoG, MAFbx, and MuRF1, were studied using immunoblotting. Results. Immunoblotting results demonstrated upregulations in Foxo1 and Foxo3a total protein levels (1.3- and 4.5-fold increases relative to euthermic control, for Foxo1 and 3a respectively) during late torpor, and protein levels remained elevated throughout the rest of torpor and at interbout arousal. We also observed decreases in inactive, phosphorylated Foxo1 and 3a proteins during throughout torpor, where levels of p-Foxo1 Ser319 and Thr24, as well as p-Foxo3a Thr32 decreased by at least 45% throughout torpor. MyoG was upregulated only during late torpor by 2.4-fold. Protein levels of MAFbx and MuRF1 increased in late torpor as well as during early arousal by as much as 2.8-fold, and MAFbx levels remained elevated during interbout arousal, whereas MuRF1 levels returned to control levels. Discussion. The present results indicate that upregulation and activation of Foxo1 and 3a, in addition to the increase in MyoG levels at late torpor, may be upregulating the expression of MAFbx and MuRF1. These findings suggest that there is activation of the ubiquitin proteasome system (UPS) as ground squirrels arouse from torpor. Therefore, the signalling pathway involving MyoG, and the E3 ligases MAFbx and MuRF1, plays a significant role in cardiac muscle remodelling during hibernation. These findings provide insights into the regulation of protein degradation and turnover in the cardiac muscle of a hibernator model.
Collapse
Affiliation(s)
- Yichi Zhang
- Institute of Biochemistry and Department of Biology, Carleton University , Ottawa , ON , Canada
| | - Oscar A Aguilar
- Institute of Biochemistry and Department of Biology, Carleton University , Ottawa , ON , Canada
| | - Kenneth B Storey
- Institute of Biochemistry and Department of Biology, Carleton University , Ottawa , ON , Canada
| |
Collapse
|
47
|
Zhang M, Li L, Xie W, Wu JF, Yao F, Tan YL, Xia XD, Liu XY, Liu D, Lan G, Zeng MY, Gong D, Cheng HP, Huang C, Zhao ZW, Zheng XL, Tang CK. Apolipoprotein A-1 binding protein promotes macrophage cholesterol efflux by facilitating apolipoprotein A-1 binding to ABCA1 and preventing ABCA1 degradation. Atherosclerosis 2016; 248:149-59. [DOI: 10.1016/j.atherosclerosis.2016.03.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 01/29/2016] [Accepted: 03/04/2016] [Indexed: 01/27/2023]
|
48
|
Mohamed RMSM, Morimoto S, Ibrahim IAAEH, Zhan DY, Du CK, Arioka M, Yoshihara T, Takahashi-Yanaga F, Sasaguri T. GSK-3β heterozygous knockout is cardioprotective in a knockin mouse model of familial dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2016; 310:H1808-15. [PMID: 27106044 DOI: 10.1152/ajpheart.00771.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/18/2016] [Indexed: 11/22/2022]
Abstract
Glycogen synthase kinase-3β (GSK-3β) plays a central role in both cardiac physiology and pathology. Herein we want to clarify the role of GSK-3β in familial dilated cardiomyopathy. We generated a mouse model carrying a heterozygous knockout mutation of GSK-3β (GSK-3β(+/-) KO), together with a ΔK210 knockin mutation in cardiac troponin T (ΔK210 cTnT KI), which was proved to be one of the genetic causes of familial dilated cardiomyopathy (DCM). GSK-3β(+/-) KO prevented the slow and rapid deterioration in left ventricular systolic function accompanying heart failure (HF) in DCM mice with heterozygous and homozygous ΔK210 cTnT KI mutations, respectively. GSK-3β(+/-) KO also prevented cardiac enlargement, myocardial fibrosis, and cardiomyocyte apoptosis and markedly reduced the expression of cardiac β-myosin heavy chain isoform, indicative of HF, in DCM mice with homozygous ΔK210 cTnT KI mutation. GSK-3β(+/-) KO also extended the life span of these DCM mice. This study suggests that the inhibition of GSK-3β is cardioprotective in familial DCM associated with ΔK210 cTnT mutation.
Collapse
Affiliation(s)
- Rasha M S M Mohamed
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sachio Morimoto
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan;
| | - Islam A A E-H Ibrahim
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt; and
| | - Dong-Yun Zhan
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Cheng-Kun Du
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Masaki Arioka
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuya Yoshihara
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumi Takahashi-Yanaga
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiyuki Sasaguri
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
49
|
Gilda JE, Lai X, Witzmann FA, Gomes AV. Delineation of Molecular Pathways Involved in Cardiomyopathies Caused by Troponin T Mutations. Mol Cell Proteomics 2016; 15:1962-81. [PMID: 27022107 DOI: 10.1074/mcp.m115.057380] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Indexed: 11/06/2022] Open
Abstract
Familial hypertrophic cardiomyopathy (FHC) is associated with mild to severe cardiac problems and is the leading cause of sudden death in young people and athletes. Although the genetic basis for FHC is well-established, the molecular mechanisms that ultimately lead to cardiac dysfunction are not well understood. To obtain important insights into the molecular mechanism(s) involved in FHC, hearts from two FHC troponin T models (Ile79Asn [I79N] and Arg278Cys [R278C]) were investigated using label-free proteomics and metabolomics. Mutations in troponin T are the third most common cause of FHC, and the I79N mutation is associated with a high risk of sudden cardiac death. Most FHC-causing mutations, including I79N, increase the Ca(2+) sensitivity of the myofilament; however, the R278C mutation does not alter Ca(2+) sensitivity and is associated with a better prognosis than most FHC mutations. Out of more than 1200 identified proteins, 53 and 76 proteins were differentially expressed in I79N and R278C hearts, respectively, when compared with wild-type hearts. Interestingly, more than 400 proteins were differentially expressed when the I79N and R278C hearts were directly compared. The three major pathways affected in I79N hearts relative to R278C and wild-type hearts were the ubiquitin-proteasome system, antioxidant systems, and energy production pathways. Further investigation of the proteasome system using Western blotting and activity assays showed that proteasome dysfunction occurs in I79N hearts. Metabolomic results corroborate the proteomic data and suggest the glycolytic, citric acid, and electron transport chain pathways are important pathways that are altered in I79N hearts relative to R278C or wild-type hearts. Our findings suggest that impaired energy production and protein degradation dysfunction are important mechanisms in FHCs associated with poor prognosis and that cardiac hypertrophy is not likely needed for a switch from fatty acid to glucose metabolism.
Collapse
Affiliation(s)
| | - Xianyin Lai
- ¶Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Frank A Witzmann
- ¶Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Aldrin V Gomes
- From the ‡Department of Neurobiology, Physiology, and Behavior, §Department of Physiology and Membrane Biology, University of California, Davis, California 95616;
| |
Collapse
|
50
|
Lau E, Cao Q, Ng DCM, Bleakley BJ, Dincer TU, Bot BM, Wang D, Liem DA, Lam MPY, Ge J, Ping P. A large dataset of protein dynamics in the mammalian heart proteome. Sci Data 2016; 3:160015. [PMID: 26977904 PMCID: PMC4792174 DOI: 10.1038/sdata.2016.15] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/12/2016] [Indexed: 01/03/2023] Open
Abstract
Protein stability is a major regulatory principle of protein function and cellular homeostasis. Despite limited understanding on mechanisms, disruption of protein turnover is widely implicated in diverse pathologies from heart failure to neurodegenerations. Information on global protein dynamics therefore has the potential to expand the depth and scope of disease phenotyping and therapeutic strategies. Using an integrated platform of metabolic labeling, high-resolution mass spectrometry and computational analysis, we report here a comprehensive dataset of the in vivo half-life of 3,228 and the expression of 8,064 cardiac proteins, quantified under healthy and hypertrophic conditions across six mouse genetic strains commonly employed in biomedical research. We anticipate these data will aid in understanding key mitochondrial and metabolic pathways in heart diseases, and further serve as a reference for methodology development in dynamics studies in multiple organ systems.
Collapse
Affiliation(s)
- Edward Lau
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Quan Cao
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Dominic C M Ng
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Brian J Bleakley
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - T Umut Dincer
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Bioinformatics, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Brian M Bot
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Sage Bionetworks, Seattle, Washignton 98109, USA
| | - Ding Wang
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - David A Liem
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Maggie P Y Lam
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Bioinformatics, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Junbo Ge
- Department of Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Peipei Ping
- The NIH Big Data to Knowledge (BD2K) Center of Excellence in Biomedical Computing at UCLA, Los Angeles, California 90095, USA.,Department of Physiology, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Bioinformatics, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Medicine,University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|