1
|
Sakamoto T, Kelly DP. Cardiac maturation. J Mol Cell Cardiol 2024; 187:38-50. [PMID: 38160640 PMCID: PMC10923079 DOI: 10.1016/j.yjmcc.2023.12.008] [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: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The heart undergoes a dynamic maturation process following birth, in response to a wide range of stimuli, including both physiological and pathological cues. This process entails substantial re-programming of mitochondrial energy metabolism coincident with the emergence of specialized structural and contractile machinery to meet the demands of the adult heart. Many components of this program revert to a more "fetal" format during development of pathological cardiac hypertrophy and heart failure. In this review, emphasis is placed on recent progress in our understanding of the transcriptional control of cardiac maturation, encompassing the results of studies spanning from in vivo models to cardiomyocytes derived from human stem cells. The potential applications of this current state of knowledge to new translational avenues aimed at the treatment of heart failure is also addressed.
Collapse
Affiliation(s)
- Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Xu W, Billon C, Li H, Wilderman A, Qi L, Graves A, Rideb JRDC, Zhao Y, Hayes M, Yu K, Losby M, Hampton CS, Adeyemi CM, Hong SJ, Nasiotis E, Fu C, Oh TG, Fan W, Downes M, Welch RD, Evans RM, Milosavljevic A, Walker JK, Jensen BC, Pei L, Burris T, Zhang L. Novel Pan-ERR Agonists Ameliorate Heart Failure Through Enhancing Cardiac Fatty Acid Metabolism and Mitochondrial Function. Circulation 2024; 149:227-250. [PMID: 37961903 PMCID: PMC10842599 DOI: 10.1161/circulationaha.123.066542] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Cardiac metabolic dysfunction is a hallmark of heart failure (HF). Estrogen-related receptors ERRα and ERRγ are essential regulators of cardiac metabolism. Therefore, activation of ERR could be a potential therapeutic intervention for HF. However, in vivo studies demonstrating the potential usefulness of ERR agonist for HF treatment are lacking, because compounds with pharmacokinetics appropriate for in vivo use have not been available. METHODS Using a structure-based design approach, we designed and synthesized 2 structurally distinct pan-ERR agonists, SLU-PP-332 and SLU-PP-915. We investigated the effect of ERR agonist on cardiac function in a pressure overload-induced HF model in vivo. We conducted comprehensive functional, multi-omics (RNA sequencing and metabolomics studies), and genetic dependency studies both in vivo and in vitro to dissect the molecular mechanism, ERR isoform dependency, and target specificity. RESULTS Both SLU-PP-332 and SLU-PP-915 significantly improved ejection fraction, ameliorated fibrosis, and increased survival associated with pressure overload-induced HF without affecting cardiac hypertrophy. A broad spectrum of metabolic genes was transcriptionally activated by ERR agonists, particularly genes involved in fatty acid metabolism and mitochondrial function. Metabolomics analysis showed substantial normalization of metabolic profiles in fatty acid/lipid and tricarboxylic acid/oxidative phosphorylation metabolites in the mouse heart with 6-week pressure overload. ERR agonists increase mitochondria oxidative capacity and fatty acid use in vitro and in vivo. Using both in vitro and in vivo genetic dependency experiments, we show that ERRγ is the main mediator of ERR agonism-induced transcriptional regulation and cardioprotection and definitively demonstrated target specificity. ERR agonism also led to downregulation of cell cycle and development pathways, which was partially mediated by E2F1 in cardiomyocytes. CONCLUSIONS ERR agonists maintain oxidative metabolism, which confers cardiac protection against pressure overload-induced HF in vivo. Our results provide direct pharmacologic evidence supporting the further development of ERR agonists as novel HF therapeutics.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Cyrielle Billon
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St Louis, MO (C.B., M.H., T.B.)
- Center for Clinical Pharmacology, St Louis College of Pharmacy, Washington University School of Medicine, St Louis, MO (C.B., M.H., T.B.)
| | - Hui Li
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Andrea Wilderman
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Lei Qi
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Andrea Graves
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Jernie Rae Dela Cruz Rideb
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Yuanbiao Zhao
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Matthew Hayes
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St Louis, MO (C.B., M.H., T.B.)
- Center for Clinical Pharmacology, St Louis College of Pharmacy, Washington University School of Medicine, St Louis, MO (C.B., M.H., T.B.)
| | - Keyang Yu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - McKenna Losby
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Carissa S Hampton
- Department of Pharmacology and Physiology, St Louis University School of Medicine, MO (C.S.H., C.M.A., J.K.W.)
| | - Christiana M Adeyemi
- Department of Pharmacology and Physiology, St Louis University School of Medicine, MO (C.S.H., C.M.A., J.K.W.)
| | - Seok Jae Hong
- McAllister Heart Institute (S.J.H., B.C.J.), University of North Carolina, Chapel Hill
| | - Eleni Nasiotis
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Chen Fu
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA (C.F.)
- University Hospitals Cleveland Medical Center, OH (C.F.)
| | - Tae Gyu Oh
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA (T.G.O., W.F., M.D., R.M.E.)
| | - Weiwei Fan
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA (T.G.O., W.F., M.D., R.M.E.)
| | - Michael Downes
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA (T.G.O., W.F., M.D., R.M.E.)
| | - Ryan D Welch
- Biology and Chemistry Department, Blackburn College, Carlinville, IL (R.D.W.)
| | - Ronald M Evans
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA (T.G.O., W.F., M.D., R.M.E.)
| | - Aleksandar Milosavljevic
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - John K Walker
- Department of Pharmacology and Physiology, St Louis University School of Medicine, MO (C.S.H., C.M.A., J.K.W.)
| | - Brian C Jensen
- McAllister Heart Institute (S.J.H., B.C.J.), University of North Carolina, Chapel Hill
- Department of Medicine, Division of Cardiology (B.C.J.), University of North Carolina, Chapel Hill
| | - Liming Pei
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, and University of Pennsylvania, Philadelphia (L.P.)
| | - Thomas Burris
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St Louis, MO (C.B., M.H., T.B.)
- Center for Clinical Pharmacology, St Louis College of Pharmacy, Washington University School of Medicine, St Louis, MO (C.B., M.H., T.B.)
| | - Lilei Zhang
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| |
Collapse
|
3
|
Shiheido-Watanabe Y, Sadoshima J. Nobiletin, a polymethoxylated flavonoid, regulates cell survival via the nuclear receptor RORα in cardiomyocytes. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:6. [PMID: 39119356 PMCID: PMC11309434 DOI: 10.20517/jca.2023.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Affiliation(s)
- Yuka Shiheido-Watanabe
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Junichi Sadoshima
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
4
|
Ravingerova T, Adameova A, Lonek L, Farkasova V, Ferko M, Andelova N, Kura B, Slezak J, Galatou E, Lazou A, Zohdi V, Dhalla NS. Is Intrinsic Cardioprotection a Laboratory Phenomenon or a Clinically Relevant Tool to Salvage the Failing Heart? Int J Mol Sci 2023; 24:16497. [PMID: 38003687 PMCID: PMC10671596 DOI: 10.3390/ijms242216497] [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: 10/24/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiovascular diseases, especially ischemic heart disease, as a leading cause of heart failure (HF) and mortality, will not reduce over the coming decades despite the progress in pharmacotherapy, interventional cardiology, and surgery. Although patients surviving acute myocardial infarction live longer, alteration of heart function will later lead to HF. Its rising incidence represents a danger, especially among the elderly, with data showing more unfavorable results among females than among males. Experiments revealed an infarct-sparing effect of ischemic "preconditioning" (IPC) as the most robust form of innate cardioprotection based on the heart's adaptation to moderate stress, increasing its resistance to severe insults. However, translation to clinical practice is limited by technical requirements and limited time. Novel forms of adaptive interventions, such as "remote" IPC, have already been applied in patients, albeit with different effectiveness. Cardiac ischemic tolerance can also be increased by other noninvasive approaches, such as adaptation to hypoxia- or exercise-induced preconditioning. Although their molecular mechanisms are not yet fully understood, some noninvasive modalities appear to be promising novel strategies for fighting HF through targeting its numerous mechanisms. In this review, we will discuss the molecular mechanisms of heart injury and repair, as well as interventions that have potential to be used in the treatment of patients.
Collapse
Affiliation(s)
- Tanya Ravingerova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Adriana Adameova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 10 Odbojárov St., 832 32 Bratislava, Slovakia
| | - Lubomir Lonek
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Veronika Farkasova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Miroslav Ferko
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Natalia Andelova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Branislav Kura
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Jan Slezak
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Eleftheria Galatou
- School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (E.G.); (A.L.)
- Department of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (E.G.); (A.L.)
| | - Vladislava Zohdi
- Department of Anatomy, Faculty of Medicine, Comenius University in Bratislava, 24 Špitalska, 813 72 Bratislava, Slovakia;
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, 19 Innovation Walk, Clayton, VIC 3800, Australia
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada;
| |
Collapse
|
5
|
Hu P, Li K, Peng X, Kan Y, Li H, Zhu Y, Wang Z, Li Z, Liu HY, Cai D. Nuclear Receptor PPARα as a Therapeutic Target in Diseases Associated with Lipid Metabolism Disorders. Nutrients 2023; 15:4772. [PMID: 38004166 PMCID: PMC10674366 DOI: 10.3390/nu15224772] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Lipid metabolic diseases have substantial morbidity and mortality rates, posing a significant threat to human health. PPARα, a member of the peroxisome proliferator-activated receptors (PPARs), plays a crucial role in lipid metabolism and immune regulation. Recent studies have increasingly recognized the pivotal involvement of PPARα in diverse pathological conditions. This comprehensive review aims to elucidate the multifaceted role of PPARα in metabolic diseases including liver diseases, diabetes-related diseases, age-related diseases, and cancers, shedding light on the underlying molecular mechanisms and some regulatory effects of natural/synthetic ligands of PPARα. By summarizing the latest research findings on PPARα, we aim to provide a foundation for the possible therapeutic exploitation of PPARα in lipid metabolic diseases.
Collapse
Affiliation(s)
- Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Kaiqi Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Xiaoxu Peng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Yufei Kan
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Hao Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Yanli Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Ziyu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Zhaojian Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, China
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, China
| |
Collapse
|
6
|
Palioura D, Mellidis K, Ioannidou-Kabouri K, Galatou E, Mouchtouri ET, Stamatiou R, Mavrommatis-Parasidis P, Panteris E, Varela A, Davos C, Drosatos K, Mavroidis M, Lazou A. PPARδ activation improves cardiac mitochondrial homeostasis in desmin deficient mice but does not alleviate systolic dysfunction. J Mol Cell Cardiol 2023; 183:27-41. [PMID: 37603971 DOI: 10.1016/j.yjmcc.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/22/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) δ is a major transcriptional regulator of cardiac energy metabolism with pleiotropic properties, including anti-inflammatory, anti-oxidative and cardioprotective action. In this study, we sought to investigate whether pharmacological activation of PPARδ via intraperitoneal administration of the selective ligand GW0742 could ameliorate heart failure and mitochondrial dysfunction that have been previously reported in a characterized genetic model of heart failure, the desmin null mice (Des-/-). Our studies demonstrate that treatment of Des-/- mice with the PPARδ agonist attenuated cardiac inflammation, fibrosis and cardiac remodeling. In addition, PPARδ activation alleviated oxidative stress in the failing myocardium as evidenced by decreased ROS levels. Importantly, PPARδ activation stimulated mitochondrial biogenesis, prevented mitochondrial and sarcoplasmic reticulum vacuolar degeneration and improved the mitochondrial intracellular distribution. Finally, PPARδ activation alleviated the mitochondrial respiratory dysfunction, prevented energy depletion and alleviated excessive autophagy and mitophagy in Des-/- hearts. Nevertheless, improvement of all these parameters did not suffice to overcome the significant structural deficiencies that desmin deletion incurs in cardiomyocytes and cardiac function did not improve significantly. In conclusion, pharmacological PPARδ activation in Des-/- hearts exerts protective effects during myocardial degeneration and heart failure by preserving the function and quality of the mitochondrial network. These findings implicate PPARδ agonists as a supplemental constituent of heart failure medications.
Collapse
Affiliation(s)
- Dimitra Palioura
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Kyriakos Mellidis
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Konstantina Ioannidou-Kabouri
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Eleftheria Galatou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | - Rodopi Stamatiou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | - Emmanuel Panteris
- Department of Botany, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Aimilia Varela
- Clinical, Experimental Surgery & Translational Research Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Constantinos Davos
- Clinical, Experimental Surgery & Translational Research Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece.
| |
Collapse
|
7
|
Yamamoto T, Maurya SK, Pruzinsky E, Batmanov K, Xiao Y, Sulon SM, Sakamoto T, Wang Y, Lai L, McDaid KS, Shewale SV, Leone TC, Koves TR, Muoio DM, Dierickx P, Lazar MA, Lewandowski ED, Kelly DP. RIP140 deficiency enhances cardiac fuel metabolism and protects mice from heart failure. J Clin Invest 2023; 133:e162309. [PMID: 36927960 PMCID: PMC10145947 DOI: 10.1172/jci162309] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
During the development of heart failure (HF), the capacity for cardiomyocyte (CM) fatty acid oxidation (FAO) and ATP production is progressively diminished, contributing to pathologic cardiac hypertrophy and contractile dysfunction. Receptor-interacting protein 140 (RIP140, encoded by Nrip1) has been shown to function as a transcriptional corepressor of oxidative metabolism. We found that mice with striated muscle deficiency of RIP140 (strNrip1-/-) exhibited increased expression of a broad array of genes involved in mitochondrial energy metabolism and contractile function in heart and skeletal muscle. strNrip1-/- mice were resistant to the development of pressure overload-induced cardiac hypertrophy, and CM-specific RIP140-deficient (csNrip1-/-) mice were protected against the development of HF caused by pressure overload combined with myocardial infarction. Genomic enhancers activated by RIP140 deficiency in CMs were enriched in binding motifs for transcriptional regulators of mitochondrial function (estrogen-related receptor) and cardiac contractile proteins (myocyte enhancer factor 2). Consistent with a role in the control of cardiac fatty acid oxidation, loss of RIP140 in heart resulted in augmented triacylglyceride turnover and fatty acid utilization. We conclude that RIP140 functions as a suppressor of a transcriptional regulatory network that controls cardiac fuel metabolism and contractile function, representing a potential therapeutic target for the treatment of HF.
Collapse
Affiliation(s)
- Tsunehisa Yamamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Santosh K. Maurya
- Davis Heart and Lung Research Institute and Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Elizabeth Pruzinsky
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kirill Batmanov
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Diabetes, Obesity and Metabolism, and Division of Endocrinology, Diabetes, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yang Xiao
- Institute for Diabetes, Obesity and Metabolism, and Division of Endocrinology, Diabetes, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sarah M. Sulon
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yang Wang
- Davis Heart and Lung Research Institute and Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Ling Lai
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kendra S. McDaid
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Swapnil V. Shewale
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa C. Leone
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy R. Koves
- Departments of Medicine and Pharmacology and Cancer Biology, and Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Deborah M. Muoio
- Departments of Medicine and Pharmacology and Cancer Biology, and Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Pieterjan Dierickx
- Institute for Diabetes, Obesity and Metabolism, and Division of Endocrinology, Diabetes, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mitchell A. Lazar
- Institute for Diabetes, Obesity and Metabolism, and Division of Endocrinology, Diabetes, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - E. Douglas Lewandowski
- Davis Heart and Lung Research Institute and Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Daniel P. Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Abstract
The ketone bodies beta-hydroxybutyrate and acetoacetate are hepatically produced metabolites catabolized in extrahepatic organs. Ketone bodies are a critical cardiac fuel and have diverse roles in the regulation of cellular processes such as metabolism, inflammation, and cellular crosstalk in multiple organs that mediate disease. This review focuses on the role of cardiac ketone metabolism in health and disease with an emphasis on the therapeutic potential of ketosis as a treatment for heart failure (HF). Cardiac metabolic reprogramming, characterized by diminished mitochondrial oxidative metabolism, contributes to cardiac dysfunction and pathologic remodeling during the development of HF. Growing evidence supports an adaptive role for ketone metabolism in HF to promote normal cardiac function and attenuate disease progression. Enhanced cardiac ketone utilization during HF is mediated by increased availability due to systemic ketosis and a cardiac autonomous upregulation of ketolytic enzymes. Therapeutic strategies designed to restore high-capacity fuel metabolism in the heart show promise to address fuel metabolic deficits that underpin the progression of HF. However, the mechanisms involved in the beneficial effects of ketone bodies in HF have yet to be defined and represent important future lines of inquiry. In addition to use as an energy substrate for cardiac mitochondrial oxidation, ketone bodies modulate myocardial utilization of glucose and fatty acids, two vital energy substrates that regulate cardiac function and hypertrophy. The salutary effects of ketone bodies during HF may also include extra-cardiac roles in modulating immune responses, reducing fibrosis, and promoting angiogenesis and vasodilation. Additional pleotropic signaling properties of beta-hydroxybutyrate and AcAc are discussed including epigenetic regulation and protection against oxidative stress. Evidence for the benefit and feasibility of therapeutic ketosis is examined in preclinical and clinical studies. Finally, ongoing clinical trials are reviewed for perspective on translation of ketone therapeutics for the treatment of HF.
Collapse
Affiliation(s)
- Timothy R. Matsuura
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Patrycja Puchalska
- Department of Medicine, Division of Molecular Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Peter A. Crawford
- Department of Medicine, Division of Molecular Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Daniel P. Kelly
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
9
|
Cao Y, Zhang X, Akerberg BN, Yuan H, Sakamoto T, Xiao F, VanDusen NJ, Zhou P, Sweat ME, Wang Y, Prondzynski M, Chen J, Zhang Y, Wang P, Kelly DP, Pu WT. In Vivo Dissection of Chamber-Selective Enhancers Reveals Estrogen-Related Receptor as a Regulator of Ventricular Cardiomyocyte Identity. Circulation 2023; 147:881-896. [PMID: 36705030 PMCID: PMC10010668 DOI: 10.1161/circulationaha.122.061955] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cardiac chamber-selective transcriptional programs underpin the structural and functional differences between atrial and ventricular cardiomyocytes (aCMs and vCMs). The mechanisms responsible for these chamber-selective transcriptional programs remain largely undefined. METHODS We nominated candidate chamber-selective enhancers (CSEs) by determining the genome-wide occupancy of 7 key cardiac transcription factors (GATA4, MEF2A, MEF2C, NKX2-5, SRF, TBX5, TEAD1) and transcriptional coactivator P300 in atria and ventricles. Candidate enhancers were tested using an adeno-associated virus-mediated massively parallel reporter assay. Chromatin features of CSEs were evaluated by performing assay of transposase accessible chromatin sequencing and acetylation of histone H3 at lysine 27-HiChIP on aCMs and vCMs. CSE sequence requirements were determined by systematic tiling mutagenesis of 29 CSEs at 5 bp resolution. Estrogen-related receptor (ERR) function in cardiomyocytes was evaluated by Cre-loxP-mediated inactivation of ERRα and ERRγ in cardiomyocytes. RESULTS We identified 134 066 and 97 506 regions reproducibly occupied by at least 1 transcription factor or P300, in atria or ventricles, respectively. Enhancer activities of 2639 regions bound by transcription factors or P300 were tested in aCMs and vCMs by adeno-associated virus-mediated massively parallel reporter assay. This identified 1092 active enhancers in aCMs or vCMs. Several overlapped loci associated with cardiovascular disease through genome-wide association studies, and 229 exhibited chamber-selective activity in aCMs or vCMs. Many CSEs exhibited differential chromatin accessibility between aCMs and vCMs, and CSEs were enriched for aCM- or vCM-selective acetylation of histone H3 at lysine 27-anchored loops. Tiling mutagenesis of 29 CSEs identified the binding motif of ERRα/γ as important for ventricular enhancer activity. The requirement of ERRα/γ to activate ventricular CSEs and promote vCM identity was confirmed by loss of the vCM gene profile in ERRα/γ knockout vCMs. CONCLUSIONS We identified 229 CSEs that could be useful research tools or direct therapeutic gene expression. We showed that chamber-selective multi-transcription factor, P300 occupancy, open chromatin, and chromatin looping are predictive features of CSEs. We found that ERRα/γ are essential for maintenance of ventricular identity. Finally, our gene expression, epigenetic, 3-dimensional genome, and enhancer activity atlas provide key resources for future studies of chamber-selective gene regulation.
Collapse
Affiliation(s)
- Yangpo Cao
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Xiaoran Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Brynn N Akerberg
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Haiyun Yuan
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, China (H.Y.)
| | - Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (T.S., D.P.K.)
| | - Feng Xiao
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Nathan J VanDusen
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis (N.J.V.)
| | - Pingzhu Zhou
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Mason E Sweat
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Yi Wang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Maksymilian Prondzynski
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Jian Chen
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Yan Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Peizhe Wang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (T.S., D.P.K.)
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.).,Harvard Stem Cell Institute, Cambridge, MA (W.T.P.)
| |
Collapse
|
10
|
Martyniak A, Jeż M, Dulak J, Stępniewski J. Adaptation of cardiomyogenesis to the generation and maturation of cardiomyocytes from human pluripotent stem cells. IUBMB Life 2023; 75:8-29. [PMID: 36263833 DOI: 10.1002/iub.2685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/05/2022] [Indexed: 12/29/2022]
Abstract
The advent of methods for efficient generation and cardiac differentiation of pluripotent stem cells opened new avenues for disease modelling, drug testing, and cell therapies of the heart. However, cardiomyocytes (CM) obtained from such cells demonstrate an immature, foetal-like phenotype that involves spontaneous contractions, irregular morphology, expression of embryonic isoforms of sarcomere components, and low level of ion channels. These and other features may affect cellular response to putative therapeutic compounds and the efficient integration into the host myocardium after in vivo delivery. Therefore, novel strategies to increase the maturity of pluripotent stem cell-derived CM are of utmost importance. Several approaches have already been developed relying on molecular changes that occur during foetal and postnatal maturation of the heart, its electromechanical activity, and the cellular composition. As a better understanding of these determinants may facilitate the generation of efficient protocols for in vitro acquisition of an adult-like phenotype by immature CM, this review summarizes the most important molecular factors that govern CM during embryonic development, postnatal changes that trigger heart maturation, as well as protocols that are currently used to generate mature pluripotent stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alicja Martyniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mateusz Jeż
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
11
|
Harnessing conserved signaling and metabolic pathways to enhance the maturation of functional engineered tissues. NPJ Regen Med 2022; 7:44. [PMID: 36057642 PMCID: PMC9440900 DOI: 10.1038/s41536-022-00246-3] [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: 02/22/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
The development of induced-pluripotent stem cell (iPSC)-derived cell types offers promise for basic science, drug testing, disease modeling, personalized medicine, and translatable cell therapies across many tissue types. However, in practice many iPSC-derived cells have presented as immature in physiological function, and despite efforts to recapitulate adult maturity, most have yet to meet the necessary benchmarks for the intended tissues. Here, we summarize the available state of knowledge surrounding the physiological mechanisms underlying cell maturation in several key tissues. Common signaling consolidators, as well as potential synergies between critical signaling pathways are explored. Finally, current practices in physiologically relevant tissue engineering and experimental design are critically examined, with the goal of integrating greater decision paradigms and frameworks towards achieving efficient maturation strategies, which in turn may produce higher-valued iPSC-derived tissues.
Collapse
|
12
|
Joseph LC, Shi J, Nguyen QN, Pensiero V, Goulbourne C, Bauer RC, Zhang H, Morrow JP. Combined metabolomic and transcriptomic profiling approaches reveal the cardiac response to high-fat diet. iScience 2022; 25:104184. [PMID: 35494220 PMCID: PMC9038541 DOI: 10.1016/j.isci.2022.104184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/04/2022] [Accepted: 03/29/2022] [Indexed: 12/24/2022] Open
Abstract
The response of vital organs to different types of nutrition or diet is a fundamental question in physiology. We examined the cardiac response to 4 weeks of high-fat diet in mice, measuring cardiac metabolites and mRNA. Metabolomics showed dramatic differences after a high-fat diet, including increases in several acyl-carnitine species. The RNA-seq data showed changes consistent with adaptations to use more fatty acid as substrate and an increase in the antioxidant protein catalase. Changes in mRNA were correlated with changes in protein level for several highly responsive genes. We also found significant sex differences in both metabolomics and RNA-seq datasets, both at baseline and after high fat diet. This work reveals the response of a vital organ to dietary intervention at both metabolomic and transcriptomic levels, which is a fundamental question in physiology. This work also reveals significant sex differences in cardiac metabolites and gene expression.
Collapse
Affiliation(s)
- Leroy C. Joseph
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Jianting Shi
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
- Cardiometabolic Genomics Program, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | - Quynh N. Nguyen
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Victoria Pensiero
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Chris Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, USA
| | - Robert C. Bauer
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Hanrui Zhang
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
- Cardiometabolic Genomics Program, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | - John P. Morrow
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| |
Collapse
|
13
|
Scholtes C, Giguère V. Transcriptional control of energy metabolism by nuclear receptors. Nat Rev Mol Cell Biol 2022; 23:750-770. [DOI: 10.1038/s41580-022-00486-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2022] [Indexed: 12/11/2022]
|
14
|
The nuclear receptor ERR cooperates with the cardiogenic factor GATA4 to orchestrate cardiomyocyte maturation. Nat Commun 2022; 13:1991. [PMID: 35418170 PMCID: PMC9008061 DOI: 10.1038/s41467-022-29733-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 03/30/2022] [Indexed: 12/19/2022] Open
Abstract
Estrogen-related receptors (ERR) α and γ were shown recently to serve as regulators of cardiac maturation, yet the underlying mechanisms have not been delineated. Herein, we find that ERR signaling is necessary for induction of genes involved in mitochondrial and cardiac-specific contractile processes during human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) differentiation. Genomic interrogation studies demonstrate that ERRγ occupies many cardiomyocyte enhancers/super-enhancers, often co-localizing with the cardiogenic factor GATA4. ERRγ interacts with GATA4 to cooperatively activate transcription of targets involved in cardiomyocyte-specific processes such as contractile function, whereas ERRγ-mediated control of metabolic genes occurs independent of GATA4. Both mechanisms require the transcriptional coregulator PGC-1α. A disease-causing GATA4 mutation is shown to diminish PGC-1α/ERR/GATA4 cooperativity and expression of ERR target genes are downregulated in human heart failure samples suggesting that dysregulation of this circuitry may contribute to congenital and acquired forms of heart failure. Mature cardiac muscle requires high mitochondrial ATP production and specialized contractile proteins. Here the authors demonstrate that cardiomyocyte-specific contractile maturation involves cooperation between the nuclear receptor ERRγ and cardiogenic transcription factor GATA4, but ERRγ controls metabolic genes independently.
Collapse
|
15
|
Pavez-Giani MG, Cyganek L. Recent Advances in Modeling Mitochondrial Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Front Cell Dev Biol 2022; 9:800529. [PMID: 35083221 PMCID: PMC8784695 DOI: 10.3389/fcell.2021.800529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022] Open
Abstract
Around one third of patients with mitochondrial disorders develop a kind of cardiomyopathy. In these cases, severity is quite variable ranging from asymptomatic status to severe manifestations including heart failure, arrhythmias, and sudden cardiac death. ATP is primarily generated in the mitochondrial respiratory chain via oxidative phosphorylation by utilizing fatty acids and carbohydrates. Genes in both the nuclear and the mitochondrial DNA encode components of this metabolic route and, although mutations in these genes are extremely rare, the risk to develop cardiac symptoms is significantly higher in this patient cohort. Additionally, infants with cardiovascular compromise in mitochondrial deficiency display a worse late survival compared to patients without cardiac symptoms. At this point, the mechanisms behind cardiac disease progression related to mitochondrial gene mutations are poorly understood and current therapies are unable to substantially restore the cardiac performance and to reduce the disease burden. Therefore, new strategies are needed to uncover the pathophysiological mechanisms and to identify new therapeutic options for mitochondrial cardiomyopathies. Here, human induced pluripotent stem cell (iPSC) technology has emerged to provide a suitable patient-specific model system by recapitulating major characteristics of the disease in vitro, as well as to offer a powerful platform for pre-clinical drug development and for the testing of novel therapeutic options. In the present review, we summarize recent advances in iPSC-based disease modeling of mitochondrial cardiomyopathies and explore the patho-mechanistic insights as well as new therapeutic approaches that were uncovered with this experimental platform. Further, we discuss the challenges and limitations of this technology and provide an overview of the latest techniques to promote metabolic and functional maturation of iPSC-derived cardiomyocytes that might be necessary for modeling of mitochondrial disorders.
Collapse
Affiliation(s)
- Mario G Pavez-Giani
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| |
Collapse
|
16
|
Yamamoto T, Sano M. Deranged Myocardial Fatty Acid Metabolism in Heart Failure. Int J Mol Sci 2022; 23:996. [PMID: 35055179 PMCID: PMC8779056 DOI: 10.3390/ijms23020996] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
The heart requires fatty acids to maintain its activity. Various mechanisms regulate myocardial fatty acid metabolism, such as energy production using fatty acids as fuel, for which it is known that coordinated control of fatty acid uptake, β-oxidation, and mitochondrial oxidative phosphorylation steps are important for efficient adenosine triphosphate (ATP) production without unwanted side effects. The fatty acids taken up by cardiomyocytes are not only used as substrates for energy production but also for the synthesis of triglycerides and the replacement reaction of fatty acid chains in cell membrane phospholipids. Alterations in fatty acid metabolism affect the structure and function of the heart. Recently, breakthrough studies have focused on the key transcription factors that regulate fatty acid metabolism in cardiomyocytes and the signaling systems that modify their functions. In this article, we reviewed the latest research on the role of fatty acid metabolism in the pathogenesis of heart failure and provide an outlook on future challenges.
Collapse
Affiliation(s)
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| |
Collapse
|
17
|
Beak JY, Kang HS, Huang W, Deshmukh R, Hong SJ, Kadakia N, Aghajanian A, Gerrish K, Jetten A, Jensen B. The nuclear receptor RORα preserves cardiomyocyte mitochondrial function by regulating caveolin-3-mediated mitophagy. J Biol Chem 2021; 297:101358. [PMID: 34756888 PMCID: PMC8626585 DOI: 10.1016/j.jbc.2021.101358] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/23/2023] Open
Abstract
Preserving optimal mitochondrial function is critical in the heart, which is the most ATP-avid organ in the body. Recently, we showed that global deficiency of the nuclear receptor RORα in the "staggerer" mouse exacerbates angiotensin II-induced cardiac hypertrophy and compromises cardiomyocyte mitochondrial function. However, the mechanisms underlying these observations have not been defined previously. Here, we used pharmacological and genetic gain- and loss-of-function tools to demonstrate that RORα regulates cardiomyocyte mitophagy to preserve mitochondrial abundance and function. We found that cardiomyocyte mitochondria in staggerer mice with lack of functional RORα were less numerous and exhibited fewer mitophagy events than those in WT controls. The hearts of our novel cardiomyocyte-specific RORα KO mouse line demonstrated impaired contractile function, enhanced oxidative stress, increased apoptosis, and reduced autophagic flux relative to Cre(-) littermates. We found that cardiomyocyte mitochondria in "staggerer" mice with lack of functional RORα were upregulated by hypoxia, a classical inducer of mitophagy. The loss of RORα blunted mitophagy and broadly compromised mitochondrial function in normoxic and hypoxic conditions in vivo and in vitro. We also show that RORα is a direct transcriptional regulator of the mitophagy mediator caveolin-3 in cardiomyocytes and that enhanced expression of RORα increases caveolin-3 abundance and enhances mitophagy. Finally, knockdown of RORα impairs cardiomyocyte mitophagy, compromises mitochondrial function, and induces apoptosis, but these defects could be rescued by caveolin-3 overexpression. Collectively, these findings reveal a novel role for RORα in regulating mitophagy through caveolin-3 and expand our currently limited understanding of the mechanisms underlying RORα-mediated cardioprotection.
Collapse
Affiliation(s)
- Ju Youn Beak
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Hong Soon Kang
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, USA
| | - Wei Huang
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Rishi Deshmukh
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Seok Jae Hong
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Nishi Kadakia
- Campbell University School of Osteopathic Medicine, Lillington, North Carolina, USA
| | - Amir Aghajanian
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Kevin Gerrish
- Molecular Genomics Core Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Anton Jetten
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, USA
| | - Brian Jensen
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA; Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.
| |
Collapse
|
18
|
Abstract
The design of the energy metabolism system in striated muscle remains a major area of investigation. Here, we review our current understanding and emerging hypotheses regarding the metabolic support of muscle contraction. Maintenance of ATP free energy, so called energy homeostasis, via mitochondrial oxidative phosphorylation is critical to sustained contractile activity, and this major design criterion is the focus of this review. Cell volume invested in mitochondria reduces the space available for generating contractile force, and this spatial balance between mitochondria acontractile elements to meet the varying sustained power demands across muscle types is another important design criterion. This is accomplished with remarkably similar mass-specific mitochondrial protein composition across muscle types, implying that it is the organization of mitochondria within the muscle cell that is critical to supporting sustained muscle function. Beyond the production of ATP, ubiquitous distribution of ATPases throughout the muscle requires rapid distribution of potential energy across these large cells. Distribution of potential energy has long been thought to occur primarily through facilitated metabolite diffusion, but recent analysis has questioned the importance of this process under normal physiological conditions. Recent structural and functional studies have supported the hypothesis that the mitochondrial reticulum provides a rapid energy distribution system via the conduction of the mitochondrial membrane potential to maintain metabolic homeostasis during contractile activity. We extensively review this aspect of the energy metabolism design contrasting it with metabolite diffusion models and how mitochondrial structure can play a role in the delivery of energy in the striated muscle.
Collapse
Affiliation(s)
- Brian Glancy
- Muscle Energetics Laboratory, National Heart, Lung, and Blood Insititute and National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, Maryland
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Insititute, Bethesda, Maryland
| | - Robert S Balaban
- Muscle Energetics Laboratory, National Heart, Lung, and Blood Insititute and National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, Maryland
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Insititute, Bethesda, Maryland
| |
Collapse
|
19
|
Cao Y, Liu K, Xiong Y, Zhao C, Liu L. Increased expression of fragmented tRNA promoted neuronal necrosis. Cell Death Dis 2021; 12:823. [PMID: 34462418 PMCID: PMC8405691 DOI: 10.1038/s41419-021-04108-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
Neuronal necrosis induced by excessive glutamate release is well known to contribute morbidity and mortality in ischemic stroke. Over the past decades, strategies on targeting glutamate receptor did not achieve desirable clinical outcomes. Finding the downstream mechanism of the glutamate receptor activation may provide new targets to suppress the cell death. Previously, our study demonstrated that the increase of H3K4 trimethylation (H3K4me3) played a key detrimental role on neuronal necrosis; however, the mechanism of this histone modification is unclear. Through a genome-wide small RNA sequencing, we identified several tRNA-derived fragments (tRFs) and piwi-interacting RNA (piRNAs) species were enriched in glutamate-induced neuronal necrosis in rat primary neuron cultures, and this enrichment was dependent on the H3K4me3 increase. Strikingly, when we transfected several synthesized tRFs and piRNA species into neurons, the tRFs but not the piRNAs induced neuron swelling and death. The cell death morphology recapitulated neuronal necrosis induced by glutamate. For the cytotoxic effect of tRFs, our data suggested that protein synthesis was inhibited likely through induction of ribosomal stalling. By proteomic analysis of tRFs effect, the most affected pathway was enriched in the mitochondrial metabolism. Consistently, mitochondrial fragmentation was increased in neuronal necrosis, and suppression of mitochondrial fission by genetic manipulation or drug rescued neuronal necrosis. Using our previously established Drosophila model of neuronal necrosis, we found that inhibition of small RNA transcription, blocking RNA transport from nucleus to cytosol, or knocking down Ago1/2 to suppress the RNA interference effect, all rescued the fly death, suggesting transcription and processing of small RNAs contribute to neuronal necrosis. Together, these results indicate that the abnormal transcription of tRFs may play a key role downstream of the H3K4me3 increase. This provides a potential new strategy to suppress neuronal necrosis.
Collapse
Affiliation(s)
- Yanyan Cao
- grid.24696.3f0000 0004 0369 153XDepartment of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, China ,grid.453074.10000 0000 9797 0900Department of Neurology, First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Kai Liu
- grid.49470.3e0000 0001 2331 6153College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Xiong
- grid.24696.3f0000 0004 0369 153XDepartment of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, China
| | - Chunyue Zhao
- grid.64939.310000 0000 9999 1211Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
| | - Lei Liu
- grid.24696.3f0000 0004 0369 153XDepartment of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, China
| |
Collapse
|
20
|
Tahri-Joutey M, Andreoletti P, Surapureddi S, Nasser B, Cherkaoui-Malki M, Latruffe N. Mechanisms Mediating the Regulation of Peroxisomal Fatty Acid Beta-Oxidation by PPARα. Int J Mol Sci 2021; 22:ijms22168969. [PMID: 34445672 PMCID: PMC8396561 DOI: 10.3390/ijms22168969] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/14/2021] [Accepted: 08/15/2021] [Indexed: 12/12/2022] Open
Abstract
In mammalian cells, two cellular organelles, mitochondria and peroxisomes, share the ability to degrade fatty acid chains. Although each organelle harbors its own fatty acid β-oxidation pathway, a distinct mitochondrial system feeds the oxidative phosphorylation pathway for ATP synthesis. At the same time, the peroxisomal β-oxidation pathway participates in cellular thermogenesis. A scientific milestone in 1965 helped discover the hepatomegaly effect in rat liver by clofibrate, subsequently identified as a peroxisome proliferator in rodents and an activator of the peroxisomal fatty acid β-oxidation pathway. These peroxisome proliferators were later identified as activating ligands of Peroxisome Proliferator-Activated Receptor α (PPARα), cloned in 1990. The ligand-activated heterodimer PPARα/RXRα recognizes a DNA sequence, called PPRE (Peroxisome Proliferator Response Element), corresponding to two half-consensus hexanucleotide motifs, AGGTCA, separated by one nucleotide. Accordingly, the assembled complex containing PPRE/PPARα/RXRα/ligands/Coregulators controls the expression of the genes involved in liver peroxisomal fatty acid β-oxidation. This review mobilizes a considerable number of findings that discuss miscellaneous axes, covering the detailed expression pattern of PPARα in species and tissues, the lessons from several PPARα KO mouse models and the modulation of PPARα function by dietary micronutrients.
Collapse
Affiliation(s)
- Mounia Tahri-Joutey
- Bio-PeroxIL Laboratory, University of Bourgogne Franche-Comté, 21000 Dijon, France; (M.T.-J.); (P.A.); (M.C.-M.)
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Sciences & Techniques, University Hassan I, BP 577, 26000 Settat, Morocco;
| | - Pierre Andreoletti
- Bio-PeroxIL Laboratory, University of Bourgogne Franche-Comté, 21000 Dijon, France; (M.T.-J.); (P.A.); (M.C.-M.)
| | - Sailesh Surapureddi
- Office of Pollution Prevention and Toxics, United States Environmental Protection Agency, Washington, DC 20460, USA;
| | - Boubker Nasser
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Sciences & Techniques, University Hassan I, BP 577, 26000 Settat, Morocco;
| | - Mustapha Cherkaoui-Malki
- Bio-PeroxIL Laboratory, University of Bourgogne Franche-Comté, 21000 Dijon, France; (M.T.-J.); (P.A.); (M.C.-M.)
| | - Norbert Latruffe
- Bio-PeroxIL Laboratory, University of Bourgogne Franche-Comté, 21000 Dijon, France; (M.T.-J.); (P.A.); (M.C.-M.)
- Correspondence:
| |
Collapse
|
21
|
Zhao Y, Ling S, Li J, Zhong G, Du R, Li Y, Wang Y, Liu C, Jin X, Liu W, Liu T, Li Y, Zhao D, Sun W, Liu Z, Liu Z, Pan J, Yuan X, Gao X, Xing W, Chang YZ, Li Y. 3' untranslated region of Ckip-1 inhibits cardiac hypertrophy independently of its cognate protein. Eur Heart J 2021; 42:3786-3799. [PMID: 34347073 DOI: 10.1093/eurheartj/ehab503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/13/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS 3' untranslated region (3' UTR) of mRNA is more conserved than other non-coding sequences in vertebrate genomes, and its sequence space has substantially expanded during the evolution of higher organisms, which substantiates their significance in biological regulation. However, the independent role of 3' UTR in cardiovascular disease was largely unknown. METHODS AND RESULTS Using bioinformatics, RNA fluorescent in situ hybridization and quantitative real-time polymerase chain reaction, we found that 3' UTR and coding sequence regions of Ckip-1 mRNA exhibited diverse expression and localization in cardiomyocytes. We generated cardiac-specific Ckip-1 3' UTR overexpression mice under wild type and casein kinase 2 interacting protein-1 (CKIP-1) knockout background. Cardiac remodelling was assessed by histological, echocardiography, and molecular analyses at 4 weeks after transverse aortic constriction (TAC) surgery. The results showed that cardiac Ckip-1 3' UTR significantly inhibited TAC-induced cardiac hypertrophy independent of CKIP-1 protein. To determine the mechanism of Ckip-1 3' UTR in cardiac hypertrophy, we performed transcriptome and metabolomics analyses, RNA immunoprecipitation, biotin-based RNA pull-down, and reporter gene assays. We found that Ckip-1 3' UTR promoted fatty acid metabolism through AMPK-PPARα-CPT1b axis, leading to its protection against pathological cardiac hypertrophy. Moreover, Ckip-1 3' UTR RNA therapy using adeno-associated virus obviously alleviates cardiac hypertrophy and improves heart function. CONCLUSIONS These findings disclose that Ckip-1 3' UTR inhibits cardiac hypertrophy independently of its cognate protein. Ckip-1 3' UTR is an effective RNA-based therapy tool for treating cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Yinlong Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China.,Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, No.20 Road East 2nd Ring South, Yuhua District, Shijiazhuang 050200, China
| | - Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Jianwei Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Guohui Zhong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Ruikai Du
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Youyou Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Yanqing Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Caizhi Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Xiaoyan Jin
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Wei Liu
- Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Tong Liu
- Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Weijia Sun
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Zizhong Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Zifan Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China.,Department of Cardiovascular Medicine, Chinese PLA General Hospital & Chinese PLA Medical School, No.28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Junjie Pan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China.,Department of Cardiology, Medical College of Soochow University, No.1 Shizi Road, Gusu District, Suzhou 215006, China
| | - Xinxin Yuan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Xingcheng Gao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Wenjuan Xing
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, No.20 Road East 2nd Ring South, Yuhua District, Shijiazhuang 050200, China
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing 100094, China
| |
Collapse
|
22
|
Paredes A, Santos-Clemente R, Ricote M. Untangling the Cooperative Role of Nuclear Receptors in Cardiovascular Physiology and Disease. Int J Mol Sci 2021; 22:ijms22157775. [PMID: 34360540 PMCID: PMC8346021 DOI: 10.3390/ijms22157775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The heart is the first organ to acquire its physiological function during development, enabling it to supply the organism with oxygen and nutrients. Given this early commitment, cardiomyocytes were traditionally considered transcriptionally stable cells fully committed to contractile function. However, growing evidence suggests that the maintenance of cardiac function in health and disease depends on transcriptional and epigenetic regulation. Several studies have revealed that the complex transcriptional alterations underlying cardiovascular disease (CVD) manifestations such as myocardial infarction and hypertrophy is mediated by cardiac retinoid X receptors (RXR) and their partners. RXRs are members of the nuclear receptor (NR) superfamily of ligand-activated transcription factors and drive essential biological processes such as ion handling, mitochondrial biogenesis, and glucose and lipid metabolism. RXRs are thus attractive molecular targets for the development of effective pharmacological strategies for CVD treatment and prevention. In this review, we summarize current knowledge of RXR partnership biology in cardiac homeostasis and disease, providing an up-to-date view of the molecular mechanisms and cellular pathways that sustain cardiomyocyte physiology.
Collapse
|
23
|
Kwik M, Hainzl S, Oppelt J, Tichy B, Koller U, Bernardinelli E, Steiner M, Zara G, Nofziger C, Weis S, Paulmichl M, Dossena S, Patsch W, Soyal SM. Selective Activation of CNS and Reference PPARGC1A Promoters Is Associated with Distinct Gene Programs Relevant for Neurodegenerative Diseases. Int J Mol Sci 2021; 22:3296. [PMID: 33804860 PMCID: PMC8036390 DOI: 10.3390/ijms22073296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022] Open
Abstract
The transcriptional regulator peroxisome proliferator activated receptor gamma coactivator 1A (PGC-1α), encoded by PPARGC1A, has been linked to neurodegenerative diseases. Recently discovered CNS-specific PPARGC1A transcripts are initiated far upstream of the reference promoter, spliced to exon 2 of the reference gene, and are more abundant than reference gene transcripts in post-mortem human brain samples. The proteins translated from the CNS and reference transcripts differ only at their N-terminal regions. To dissect functional differences between CNS-specific isoforms and reference proteins, we used clustered regularly interspaced short palindromic repeats transcriptional activation (CRISPRa) for selective endogenous activation of the CNS or the reference promoters in SH-SY5Y cells. Expression and/or exon usage of the targets was ascertained by RNA sequencing. Compared to controls, more differentially expressed genes were observed after activation of the CNS than the reference gene promoter, while the magnitude of alternative exon usage was comparable between activation of the two promoters. Promoter-selective associations were observed with canonical signaling pathways, mitochondrial and nervous system functions and neurological diseases. The distinct N-terminal as well as the shared downstream regions of PGC-1α isoforms affect the exon usage of numerous genes. Furthermore, associations of risk genes of amyotrophic lateral sclerosis and Parkinson's disease were noted with differentially expressed genes resulting from the activation of the CNS and reference gene promoter, respectively. Thus, CNS-specific isoforms markedly amplify the biological functions of PPARGC1A and CNS-specific isoforms and reference proteins have common, complementary and selective functions relevant for neurodegenerative diseases.
Collapse
Affiliation(s)
- Markus Kwik
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Strubergasse 18, 5020 Salzburg, Austria; (M.K.); (E.B.); (G.Z.); (S.D.)
| | - Stefan Hainzl
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.H.); (U.K.)
| | - Jan Oppelt
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (J.O.); (B.T.)
- Department of Pathology and Laboratory Medicine, Perlman School of Medicine, University of Pennsylvania, 614 Stellar-Chance Labs, 422 Curie Blvd, Philadelphia, PA 19104-6100, USA
| | - Boris Tichy
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (J.O.); (B.T.)
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.H.); (U.K.)
| | - Emanuele Bernardinelli
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Strubergasse 18, 5020 Salzburg, Austria; (M.K.); (E.B.); (G.Z.); (S.D.)
| | - Markus Steiner
- Third Medical Department, Cancer Research Institute, Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria;
| | - Greta Zara
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Strubergasse 18, 5020 Salzburg, Austria; (M.K.); (E.B.); (G.Z.); (S.D.)
| | | | - Serge Weis
- Division of Neuropathology, Neuromed Campus, Kepler University Hospital, 4020 Linz, Austria;
| | - Markus Paulmichl
- Department of Personalized Medicine, Humanomed, 9020 Klagenfurt, Austria;
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Strubergasse 18, 5020 Salzburg, Austria; (M.K.); (E.B.); (G.Z.); (S.D.)
| | - Wolfgang Patsch
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Strubergasse 18, 5020 Salzburg, Austria; (M.K.); (E.B.); (G.Z.); (S.D.)
| | - Selma M. Soyal
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Strubergasse 18, 5020 Salzburg, Austria; (M.K.); (E.B.); (G.Z.); (S.D.)
| |
Collapse
|
24
|
Li E, Li X, Huang J, Xu C, Liang Q, Ren K, Bai A, Lu C, Qian R, Sun N. BMAL1 regulates mitochondrial fission and mitophagy through mitochondrial protein BNIP3 and is critical in the development of dilated cardiomyopathy. Protein Cell 2020; 11:661-679. [PMID: 32277346 PMCID: PMC7452999 DOI: 10.1007/s13238-020-00713-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 03/18/2020] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of circadian rhythms associates with cardiovascular disorders. It is known that deletion of the core circadian gene Bmal1 in mice causes dilated cardiomyopathy. However, the biological rhythm regulation system in mouse is very different from that of humans. Whether BMAL1 plays a role in regulating human heart function remains unclear. Here we generated a BMAL1 knockout human embryonic stem cell (hESC) model and further derived human BMAL1 deficient cardiomyocytes. We show that BMAL1 deficient hESC-derived cardiomyocytes exhibited typical phenotypes of dilated cardiomyopathy including attenuated contractility, calcium dysregulation, and disorganized myofilaments. In addition, mitochondrial fission and mitophagy were suppressed in BMAL1 deficient hESC-cardiomyocytes, which resulted in significantly attenuated mitochondrial oxidative phosphorylation and compromised cardiomyocyte function. We also found that BMAL1 binds to the E-box element in the promoter region of BNIP3 gene and specifically controls BNIP3 protein expression. BMAL1 knockout directly reduced BNIP3 protein level, causing compromised mitophagy and mitochondria dysfunction and thereby leading to compromised cardiomyocyte function. Our data indicated that the core circadian gene BMAL1 is critical for normal mitochondria activities and cardiac function. Circadian rhythm disruption may directly link to compromised heart function and dilated cardiomyopathy in humans.
Collapse
Affiliation(s)
- Ermin Li
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiuya Li
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jie Huang
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chen Xu
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qianqian Liang
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Kehan Ren
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Aobing Bai
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, 200032, China.
- Research Center on Aging and Medicine, Fudan University, Shanghai, 200032, China.
| | - Ruizhe Qian
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, 200032, China.
- Research Center on Aging and Medicine, Fudan University, Shanghai, 200032, China.
| | - Ning Sun
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Shanghai Key Lab of Birth Defect, Children's Hospital of Fudan University, Shanghai, 201102, China.
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, 200032, China.
- Research Center on Aging and Medicine, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
25
|
Affiliation(s)
- Tomoya Sakamoto
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Daniel P. Kelly
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
26
|
Sahraoui A, Dewachter C, Vegh G, Mc Entee K, Naeije R, Bouguerra SA, Dewachter L. High fat diet altered cardiac metabolic gene profile in Psammomys obesus gerbils. Lipids Health Dis 2020; 19:123. [PMID: 32493392 PMCID: PMC7271448 DOI: 10.1186/s12944-020-01301-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 05/22/2020] [Indexed: 01/18/2023] Open
Abstract
Background In metabolic disorders, myocardial fatty infiltration is critically associated with lipotoxic cardiomyopathy. Methods Twenty Psammomys obesus gerbils were randomly assigned to normal plant or high fat diet. Sixteen weeks later, myocardium was sampled for pathobiological evaluation. Results A sixteen-week high fat diet resulted in myocardial structure disorganization, with collagen deposits, lipid accumulation, cardiomyocyte apoptosis and inflammatory cell infiltration. Myocardial expressions of glucose transporter GLUT1 and pyruvate dehydrogenase (PDH) inhibitor, PDH kinase (PDK)4 increased, while insulin-regulated GLUT4 expression remained unchanged. Myocardial expressions of molecules regulating fatty acid transport, CD36 and fatty acid binding protein (FABP)3, were increased, while expression of rate-controlling fatty acid β-oxidation, carnitine palmitoyl transferase (CPT)1B decreased. Myocardial expression of AMP-activated protein kinase (AMPK), decreased, while expression of peroxisome proliferator activated receptors (PPAR)-α and -γ did not change. Conclusion In high fat diet fed Psammomys obesus, an original experimental model of nutritionally induced metabolic syndrome mixing genetic predisposition and environment interactions, a short period of high fat feeding was sufficient to induce myocardial structural alterations, associated with altered myocardial metabolic gene expression in favor of lipid accumulation.
Collapse
Affiliation(s)
- Abdelhamid Sahraoui
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 808, Lennik Road, 1070, Brussels, Belgium.,Team of Cellular and Molecular Physiopathology, Faculty of Biological Sciences, Houari Boumediene University of Sciences and Technology, El Alia, Algiers, Algeria.,Faculté des Sciences de la Nature et de la Vie & des Sciences de la Terre, University Djilali Bounaama of Khemis Miliana, 44225, Khemis Miliana, Algeria
| | - Céline Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 808, Lennik Road, 1070, Brussels, Belgium.,Department of Cardiology, Cliniques Universitaires de Bruxelles, Hôpital Académique Erasme, Bruxelles, Belgium
| | - Grégory Vegh
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 808, Lennik Road, 1070, Brussels, Belgium
| | - Kathleen Mc Entee
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 808, Lennik Road, 1070, Brussels, Belgium
| | - Robert Naeije
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 808, Lennik Road, 1070, Brussels, Belgium
| | - Souhila Aouichat Bouguerra
- Team of Cellular and Molecular Physiopathology, Faculty of Biological Sciences, Houari Boumediene University of Sciences and Technology, El Alia, Algiers, Algeria
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 808, Lennik Road, 1070, Brussels, Belgium.
| |
Collapse
|
27
|
Gong X, Yu Z, Huang Z, Xie L, Zhou N, Wang J, Liang Y, Qin S, Nie Z, Wei L, Li Z, Wang S, Su Y, Ge J. Protective effects of cardiac resynchronization therapy in a canine model with experimental heart failure by improving mitochondrial function: a mitochondrial proteomics study. J Interv Card Electrophysiol 2020; 61:123-135. [DOI: 10.1007/s10840-020-00768-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/04/2020] [Indexed: 12/18/2022]
|
28
|
Xu X, Chen C, Lu WJ, Su YL, Shi JY, Liu YC, Wang L, Xiao CX, Wu X, Lu Q. Pyrroloquinoline quinone can prevent chronic heart failure by regulating mitochondrial function. Cardiovasc Diagn Ther 2020; 10:453-469. [PMID: 32695625 DOI: 10.21037/cdt-20-129] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Myocardial mitochondrial dysfunction is the leading cause of chronic heart failure (CHF). Increased reactive oxygen species (ROS) levels, disruption of mitochondrial biogenesis and mitochondrial Ca2+([Ca2+]m) homeostasis and reduction of the mitochondrial membrane potential (ΔΨm) cause myocardial mitochondrial dysfunction. Therefore, treating CHF by targeting mitochondrial function is a focus of current research. For the first time, this study investigated the effects of the strong antioxidant pyrroloquinoline quinone (PQQ) on mitochondrial function in a cardiac pressure overload model, and the mechanism by which PQQ regulates [Ca2+]m homeostasis was explored in depth. Methods After transaortic constriction (TAC), normal saline and PQQ (0.4, 2 and 10 mg/kg) were administered intragastrically to Sprague Dawley (SD) rats for 12 weeks. In vitro, neonatal rat left ventricle myocytes (NRVMs) were pretreated with 200 nm angiotensin II (Ang II) with or without PQQ (1, 10 and 100 μM). Rat heart remodelling was verified by assessment of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) levels (qRT-PCR), cell surface area (wheat germ agglutinin (WGA) staining in vivo and α-actin in vitro) and echocardiography. Myocardial mitochondrial morphology was assessed by transmission electron microscopy. Western blotting was used to assess mitochondrial biogenesis [peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and transcription factor A, mitochondrial (TFAM)]. The ΔΨm was determined by tetraethyl benzimidazolyl carbocyanine iodide (JC-1) staining and flow cytometry, and ROS levels were measured by dichloro-dihydro-fluorescein diacetate (DCFH-DA) and MitoSOX Red staining. [Ca2+]m was measured by isolating rat mitochondria, and mitochondrial Ca2+ channel proteins [the mitochondrial Na+/Ca2+ exchanger (NCLX) and mitochondrial Ca2+ uniporter (MCU)] were detected by Western blot. Results In vivo and in vitro, PQQ pretreatment improved pressure overload-induced cardiac remodelling and cell hypertrophy, thus preventing the occurrence of CHF. PQQ also prevented mitochondrial morphology damage and reduced the PGC-1α and TFAM downregulation caused by TAC or Ang II. In addition, in NRVMs treated with Ang II + PQQ, PQQ regulated ROS levels and increased the ΔΨm. PQQ also regulated [Ca2+]m homeostasis and prohibited [Ca2+]m overloading by increasing NCLX expression. Conclusions These results show that PQQ can prevent [Ca2+]m overload by increasing NCLX expression and thereby reducing ROS production and protecting the ΔΨm. At the same time, PQQ can increase PGC-1α and TFAM expression to regulate mitochondrial biogenesis. These factors can prevent mitochondrial dysfunction, thereby reducing cardiac damage caused by pressure overload and preventing the occurrence of CHF.
Collapse
Affiliation(s)
- Xuan Xu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Chu Chen
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wen-Jiang Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yi-Ling Su
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jia-Yu Shi
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu-Chen Liu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Li Wang
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Chen-Xi Xiao
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiang Wu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qi Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
29
|
Soyal SM, Bonova P, Kwik M, Zara G, Auer S, Scharler C, Strunk D, Nofziger C, Paulmichl M, Patsch W. The Expression of CNS-Specific PPARGC1A Transcripts Is Regulated by Hypoxia and a Variable GT Repeat Polymorphism. Mol Neurobiol 2020; 57:752-764. [PMID: 31471878 PMCID: PMC7031416 DOI: 10.1007/s12035-019-01731-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022]
Abstract
PPARGC1A encodes a transcriptional co-activator also termed peroxisome proliferator-activated receptor (PPAR) gamma coactivator 1-alpha (PGC-1α) which orchestrates multiple transcriptional programs. We have recently identified CNS-specific transcripts that are initiated far upstream of the reference gene (RG) promoter. The regulation of these isoforms may be relevant, as experimental and genetic studies implicated the PPARGC1A locus in neurodegenerative diseases. We therefore studied cis- and trans-regulatory elements activating the CNS promoter in comparison to the RG promoter in human neuronal cell lines. A naturally occurring variable guanidine thymidine (GT) repeat polymorphism within a microsatellite region in the proximal CNS promoter increases promoter activity in neuronal cell lines. Both the RG and the CNS promoters are activated by ESRRA, and the PGC-1α isoforms co-activate ESRRA on their own promoters suggesting an autoregulatory feedback loop. The proximal CNS, but not the RG, promoter is induced by FOXA2 and co-activated by PGC-1α resulting in robust activation. Furthermore, the CNS, but not the RG, promoter is targeted by the canonical hypoxia response involving HIF1A. Importantly, the transactivation by HIF1A is modulated by the size of the GT polymorphism. Increased expression of CNS-specific transcripts in response to hypoxia was observed in an established rat model, while RG transcripts encoding the full-length reference protein were not increased. These results suggest a role of the CNS region of the PPARGC1A locus in ischemia and warrant further studies in humans as the activity of the CNS promoter as well as its induction by hypoxia is subject to inter-individual variability due to the GT polymorphism.
Collapse
Affiliation(s)
- Selma M Soyal
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020, Salzburg, Austria.
| | - Petra Bonova
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Markus Kwik
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Greta Zara
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Simon Auer
- Institute for Medical and Chemical Laboratory Diagnostics, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Cornelia Scharler
- Institute of Experimental and Clinical Cell Therapy, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Dirk Strunk
- Institute of Experimental and Clinical Cell Therapy, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, 5020, Salzburg, Austria
| | | | - Markus Paulmichl
- PharmGenetix GmbH, Niederalm, 5081, Salzburg, Austria
- Department of Personalized Medicine, Humanomed, 9020, Klagenfurt, Austria
| | - Wolfgang Patsch
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020, Salzburg, Austria.
| |
Collapse
|
30
|
Hansmann G, Calvier L, Risbano MG, Chan SY. Activation of the Metabolic Master Regulator PPARγ: A Potential PIOneering Therapy for Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2020; 62:143-156. [PMID: 31577451 PMCID: PMC6993553 DOI: 10.1165/rcmb.2019-0226ps] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/02/2019] [Indexed: 12/24/2022] Open
Abstract
Translational research is essential to the development of reverse-remodeling strategies for the treatment of pulmonary vascular disease, pulmonary hypertension, and heart failure via mechanistic in vivo studies using animal models resembling human pulmonary arterial hypertension (PAH), cardiovascular remodeling, and progressive right heart failure. Since 2007, peroxisome proliferator-activated receptor γ (PPARγ) agonists have emerged as promising novel, antiproliferative, antiinflammatory, insulin-sensitizing, efficient medications for the treatment of PAH. However, early diabetes study results, their subsequent misinterpretations, errors in published review articles, and rumors regarding potential adverse effects in the literature have dampened enthusiasm for considering pharmacological PPARγ activation for the treatment of cardiovascular diseases, including PAH. Most recently, the thiazolidinedione class PPARγ agonist pioglitazone underwent a clinical revival, especially based on the IRIS (Insulin Resistance Intervention After Stroke) study, a randomized controlled trial in 3,876 patients without diabetes status post-transient ischemic attack/ischemic stroke who were clinically followed for 4.8 years. We discuss preclinical basic translational findings and randomized controlled trials related to the beneficial and adverse effects of PPARγ agonists of the thiazolidinedione class, with a particular focus on the last 5 years. The objective is a data-driven approach to set the preclinical and clinical study record straight. The convincing recent clinical trial data on the lack of significant toxicity in high-risk populations justify the timely conduct of clinical studies to achieve "repurposing" or "repositioning" of pioglitazone for the treatment of clinical PAH.
Collapse
Affiliation(s)
- Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany; and
| | - Laurent Calvier
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany; and
| | - Michael G. Risbano
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, and
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, and
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
31
|
Glancy B. Visualizing Mitochondrial Form and Function within the Cell. Trends Mol Med 2020; 26:58-70. [PMID: 31706841 PMCID: PMC6938546 DOI: 10.1016/j.molmed.2019.09.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022]
Abstract
The specific cellular role of mitochondria is influenced by the surrounding environment because effective mitochondrial function requires the delivery of inputs (e.g., oxygen) and export of products (e.g., signaling molecules) to and from other cellular components, respectively. Recent technological developments in mitochondrial imaging have led to a more precise and comprehensive understanding of the spatial relationships governing the function of this complex organelle, opening a new era of mitochondrial research. Here, I highlight current imaging approaches for visualizing mitochondrial form and function within complex cellular environments. Increasing clarity of mitochondrial behavior within cells will continue to lend mechanistic insights into the role of mitochondria under normal and pathological conditions and point to spatially regulated processes that can be targeted to improve cellular function.
Collapse
Affiliation(s)
- Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
32
|
Emmett MJ, Lazar MA. Integrative regulation of physiology by histone deacetylase 3. Nat Rev Mol Cell Biol 2019; 20:102-115. [PMID: 30390028 DOI: 10.1038/s41580-018-0076-0] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cell-type-specific gene expression is physiologically modulated by the binding of transcription factors to genomic enhancer sequences, to which chromatin modifiers such as histone deacetylases (HDACs) are recruited. Drugs that inhibit HDACs are in clinical use but lack specificity. HDAC3 is a stoichiometric component of nuclear receptor co-repressor complexes whose enzymatic activity depends on this interaction. HDAC3 is required for many aspects of mammalian development and physiology, for example, for controlling metabolism and circadian rhythms. In this Review, we discuss the mechanisms by which HDAC3 regulates cell type-specific enhancers, the structure of HDAC3 and its function as part of nuclear receptor co-repressors, its enzymatic activity and its post-translational modifications. We then discuss the plethora of tissue-specific physiological functions of HDAC3.
Collapse
Affiliation(s)
- Matthew J Emmett
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA. .,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Trevino MB, Zhang X, Standley RA, Wang M, Han X, Reis FCG, Periasamy M, Yu G, Kelly DP, Goodpaster BH, Vega RB, Coen PM. Loss of mitochondrial energetics is associated with poor recovery of muscle function but not mass following disuse atrophy. Am J Physiol Endocrinol Metab 2019; 317:E899-E910. [PMID: 31479303 PMCID: PMC6879870 DOI: 10.1152/ajpendo.00161.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 12/20/2022]
Abstract
Skeletal muscle atrophy is a clinically important outcome of disuse because of injury, immobilization, or bed rest. Disuse atrophy is accompanied by mitochondrial dysfunction, which likely contributes to activation of the muscle atrophy program. However, the linkage of muscle mass and mitochondrial energetics during disuse atrophy and its recovery is incompletely understood. Transcriptomic analysis of muscle biopsies from healthy older adults subject to complete bed rest revealed marked inhibition of mitochondrial energy metabolic pathways. To determine the temporal sequence of muscle atrophy and changes in intramyocellular lipid and mitochondrial energetics, we conducted a time course of hind limb unloading-induced atrophy in adult mice. Mitochondrial respiration and calcium retention capacity were diminished, whereas H2O2 emission was increased within 3 days of unloading before significant muscle atrophy. These changes were associated with a decrease in total cardiolipin and profound changes in remodeled cardiolipin species. Hind limb unloading performed in muscle-specific peroxisome proliferator-activated receptor-γ coactivator-1α/β knockout mice, a model of mitochondrial dysfunction, did not affect muscle atrophy but impacted muscle function. These data suggest early mitochondrial remodeling affects muscle function but not mass during disuse atrophy. Early alterations in mitochondrial energetics and lipid remodeling may represent novel targets to prevent muscle functional impairment caused by disuse and to enhance recovery from periods of muscle atrophy.
Collapse
Affiliation(s)
- Michelle B Trevino
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Xiaolei Zhang
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, Florida
| | - Robert A Standley
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, Florida
| | - Miao Wang
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Xianlin Han
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Felipe C G Reis
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Muthu Periasamy
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Gongxin Yu
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, Florida
| | - Daniel P Kelly
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
- Cardiovascular Research Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bret H Goodpaster
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, Florida
| | - Rick B Vega
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, Florida
| | - Paul M Coen
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, Florida
| |
Collapse
|
34
|
Zhu X, Shen W, Yao K, Wang H, Liu B, Li T, Song L, Diao D, Mao G, Huang P, Li C, Zhang H, Zou Y, Qiu Y, Zhao Y, Wang W, Yang Y, Hu Z, Auwerx J, Loscalzo J, Zhou Y, Ju Z. Fine-Tuning of PGC1α Expression Regulates Cardiac Function and Longevity. Circ Res 2019; 125:707-719. [PMID: 31412728 DOI: 10.1161/circresaha.119.315529] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE PGC1α (peroxisome proliferator-activated receptor gamma coactivator 1α) represents an attractive target interfering bioenergetics and mitochondrial homeostasis, yet multiple attempts have failed to upregulate PGC1α expression as a therapy, for instance, causing cardiomyopathy. OBJECTIVE To determine whether a fine-tuning of PGC1α expression is essential for cardiac homeostasis in a context-dependent manner. METHODS AND RESULTS Moderate cardiac-specific PGC1α overexpression through a ROSA26 locus knock-in strategy was utilized in WT (wild type) mice and in G3Terc-/- (third generation of telomerase deficient; hereafter as G3) mouse model, respectively. Ultrastructure, mitochondrial stress, echocardiographic, and a variety of biological approaches were applied to assess mitochondrial physiology and cardiac function. While WT mice showed a relatively consistent PGC1α expression from 3 to 12 months old, age-matched G3 mice exhibited declined PGC1α expression and compromised mitochondrial function. Cardiac-specific overexpression of PGC1α (PGC1αOE) promoted mitochondrial and cardiac function in 3-month-old WT mice but accelerated cardiac aging and significantly shortened life span in 12-month-old WT mice because of increased mitochondrial damage and reactive oxygen species insult. In contrast, cardiac-specific PGC1α knock in in G3 (G3 PGC1αOE) mice restored mitochondrial homeostasis and attenuated senescence-associated secretory phenotypes, thereby preserving cardiac performance with age and extending health span. Mechanistically, age-dependent defect in mitophagy is associated with accumulation of damaged mitochondria that leads to cardiac impairment and premature death in 12-month-old WT PGC1αOE mice. In the context of telomere dysfunction, PGC1α induction replenished energy supply through restoring the compromised mitochondrial biogenesis and thus is beneficial to old G3 heart. CONCLUSIONS Fine-tuning the expression of PGC1α is crucial for the cardiac homeostasis because the balance between mitochondrial biogenesis and clearance is vital for regulating mitochondrial function and homeostasis. These results reinforce the importance of carefully evaluating the PGC1α-boosting strategies in a context-dependent manner to facilitate clinical translation of novel cardioprotective therapies.
Collapse
Affiliation(s)
- Xudong Zhu
- From the Institute of Aging Research, Hangzhou Normal University School of Medicine, China (X.Z., H.W., T.L.)
| | - Weiyan Shen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, China (W.S., H.W., B.L., D.D., Z.J.)
| | - Ke Yao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China (K.Y., Z.H.)
| | - Hu Wang
- From the Institute of Aging Research, Hangzhou Normal University School of Medicine, China (X.Z., H.W., T.L.).,Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, China (W.S., H.W., B.L., D.D., Z.J.)
| | - Bo Liu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, China (W.S., H.W., B.L., D.D., Z.J.)
| | - Tangliang Li
- From the Institute of Aging Research, Hangzhou Normal University School of Medicine, China (X.Z., H.W., T.L.)
| | - Lijuan Song
- Department of Cardiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (L.S.)
| | - Daojun Diao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, China (W.S., H.W., B.L., D.D., Z.J.)
| | - Genxiang Mao
- Department of Geriatrics, Zhejiang Provincial Key Lab of Geriatrics, Geriatrics Research Institute of Zhejiang Province, Zhejiang Hospital, Hangzhou, China (G.M.)
| | - Ping Huang
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Institute of Forensic Sciences, Ministry of Justice, China (P.H., C.L.)
| | - Chengtao Li
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Institute of Forensic Sciences, Ministry of Justice, China (P.H., C.L.)
| | - Hongbo Zhang
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Federale de Lausanne, Switzerland (H.Z., J.A.)
| | - Yejun Zou
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology (Y. Zou, Y. Zhao, Y.Y.)
| | - Yugang Qiu
- School of Rehabilitation Medicine, Weifang Medical University, China (Y.Q.)
| | - Yuzheng Zhao
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology (Y. Zou, Y. Zhao, Y.Y.)
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, China (W.W.)
| | - Yi Yang
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology (Y. Zou, Y. Zhao, Y.Y.)
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China (K.Y., Z.H.)
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Federale de Lausanne, Switzerland (H.Z., J.A.)
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.)
| | - Yong Zhou
- Beijing Sanbo Brain Hospital, Capital Medical University, China (Y. Zhou)
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, China (W.S., H.W., B.L., D.D., Z.J.)
| |
Collapse
|
35
|
Allosteric, transcriptional and post-translational control of mitochondrial energy metabolism. Biochem J 2019; 476:1695-1712. [PMID: 31217327 DOI: 10.1042/bcj20180617] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/24/2022]
Abstract
The heart is the organ with highest energy turnover rate (per unit weight) in our body. The heart relies on its flexible and powerful catabolic capacity to continuously generate large amounts of ATP utilizing many energy substrates including fatty acids, carbohydrates (glucose and lactate), ketones and amino acids. The normal health mainly utilizes fatty acids (40-60%) and glucose (20-40%) for ATP production while ketones and amino acids have a minor contribution (10-15% and 1-2%, respectively). Mitochondrial oxidative phosphorylation is the major contributor to cardiac energy production (95%) while cytosolic glycolysis has a marginal contribution (5%). The heart can dramatically and swiftly switch between energy-producing pathways and/or alter the share from each of the energy substrates based on cardiac workload, availability of each energy substrate and neuronal and hormonal activity. The heart is equipped with a highly sophisticated and powerful mitochondrial machinery which synchronizes cardiac energy production from different substrates and orchestrates the rate of ATP production to accommodate its contractility demands. This review discusses mitochondrial cardiac energy metabolism and how it is regulated. This includes a discussion on the allosteric control of cardiac energy metabolism by short-chain coenzyme A esters, including malonyl CoA and its effect on cardiac metabolic preference. We also discuss the transcriptional level of energy regulation and its role in the maturation of cardiac metabolism after birth and cardiac adaptability for different metabolic conditions and energy demands. The role post-translational modifications, namely phosphorylation, acetylation, malonylation, succinylation and glutarylation, play in regulating mitochondrial energy metabolism is also discussed.
Collapse
|
36
|
Nakamura M, Liu T, Husain S, Zhai P, Warren JS, Hsu CP, Matsuda T, Phiel CJ, Cox JE, Tian B, Li H, Sadoshima J. Glycogen Synthase Kinase-3α Promotes Fatty Acid Uptake and Lipotoxic Cardiomyopathy. Cell Metab 2019; 29:1119-1134.e12. [PMID: 30745182 PMCID: PMC6677269 DOI: 10.1016/j.cmet.2019.01.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/27/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023]
Abstract
Obesity induces lipotoxic cardiomyopathy, a condition in which lipid accumulation in cardiomyocytes causes cardiac dysfunction. Here, we show that glycogen synthase kinase-3α (GSK-3α) mediates lipid accumulation in the heart. Fatty acids (FAs) upregulate GSK-3α, which phosphorylates PPARα at Ser280 in the ligand-binding domain (LBD). This modification ligand independently enhances transcription of a subset of PPARα targets, selectively stimulating FA uptake and storage, but not oxidation, thereby promoting lipid accumulation. Constitutively active GSK-3α, but not GSK-3β, was sufficient to drive PPARα signaling, while cardiac-specific knockdown of GSK-3α, but not GSK-3β, or replacement of PPARα Ser280 with Ala conferred resistance to lipotoxicity in the heart. Fibrates, PPARα ligands, inhibited phosphorylation of PPARα at Ser280 by inhibiting the interaction of GSK-3α with the LBD of PPARα, thereby reversing lipotoxic cardiomyopathy. These results suggest that GSK-3α promotes lipid anabolism through PPARα-Ser280 phosphorylation, which underlies the development of lipotoxic cardiomyopathy in the context of obesity.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Biochemistry & Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Seema Husain
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Junco S Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei City, Taiwan
| | - Takahisa Matsuda
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Christopher J Phiel
- Department of Integrative Biology, University of Colorado Denver, Denver, CO, USA
| | - James E Cox
- Metabolomics Core Research Facility and Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Bin Tian
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Biochemistry & Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
37
|
Yurista SR, Silljé HH, Oberdorf‐Maass SU, Schouten E, Pavez Giani MG, Hillebrands J, van Goor H, van Veldhuisen DJ, de Boer RA, Westenbrink BD. Sodium–glucose co‐transporter 2 inhibition with empagliflozin improves cardiac function in non‐diabetic rats with left ventricular dysfunction after myocardial infarction. Eur J Heart Fail 2019; 21:862-873. [DOI: 10.1002/ejhf.1473] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/06/2019] [Accepted: 03/17/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Salva R. Yurista
- Department of Cardiology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - Herman H.W. Silljé
- Department of Cardiology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - Silke U. Oberdorf‐Maass
- Department of Cardiology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - Elisabeth‐Maria Schouten
- Department of Cardiology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - Mario G. Pavez Giani
- Department of Cardiology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - Jan‐Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - Dirk J. van Veldhuisen
- Department of Cardiology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| | - B. Daan Westenbrink
- Department of Cardiology, University Medical Center GroningenUniversity of Groningen Groningen The Netherlands
| |
Collapse
|
38
|
Protection of Myocardial Ischemia-Reperfusion by Therapeutic Hypercapnia: a Mechanism Involving Improvements in Mitochondrial Biogenesis and Function. J Cardiovasc Transl Res 2019; 12:467-477. [PMID: 30980235 DOI: 10.1007/s12265-019-09886-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
Previous studies proposed that acidic reperfusion may be a protective strategy for myocardial ischemia-reperfusion therapy with potential of clinical transformation. In this study, we investigated whether therapeutic hypercapnia could mimic acidosis postconditioning in isolated hearts with a 30-min left coronary artery ligation-reperfusion model in rats. Therapeutic hypercapnia (inhalation 20% CO2 for 10 min) is cardioprotective with a strict therapeutic time window and acidity: it reduced the infarct ratio and serum myocardial enzyme and increased the myocardial ATP content. Furthermore, mitochondrial morphology damage, the loss of mitochondrial membrane potential, and the formation of mitochondrial permeability transition pore were effectively inhibited, indicating the improvements in mitochondrial function. The expression of the mitochondrial biogenesis regulators was upregulated simultaneously. These findings indicated therapeutic hypercapnia in animals can mimic ex vivo acidosis postconditioning to alleviate myocardial ischemia-reperfusion injury. The effect is related to improvement in mitochondrial function and regulation of the mitochondrial biogenesis pathway.
Collapse
|
39
|
Nakamura M, Sadoshima J. Cardiomyopathy in obesity, insulin resistance and diabetes. J Physiol 2019; 598:2977-2993. [PMID: 30869158 DOI: 10.1113/jp276747] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity, insulin resistance and diabetes is increasing rapidly. Most patients with these disorders have hypertriglyceridaemia and increased plasma levels of fatty acids, which are taken up and stored in lipid droplets in the heart. Intramyocardial lipids that exceed the capacity for storage and oxidation can be lipotoxic and induce non-ischaemic and non-hypertensive cardiomyopathy, termed diabetic or lipotoxic cardiomyopathy. The clinical features of diabetic cardiomyopathy are cardiac hypertrophy and diastolic dysfunction, which lead to heart failure, especially heart failure with preserved ejection fraction. Although the pathogenesis of the cardiomyopathy is multifactorial, diabetic dyslipidaemia and intramyocardial lipid accumulation are the key pathological features, triggering cellular signalling and modifications of proteins and lipids via generation of toxic metabolic intermediates. Most clinical studies have shown no beneficial effect of anti-diabetic agents and statins on outcomes in heart failure patients without atherosclerotic diseases, indicating the importance of identifying underlying mechanisms and early interventions for diabetic cardiomyopathy. Here, we summarize the molecular mechanisms of diabetic cardiomyopathy, with a special emphasis on cardiac lipotoxicity, and discuss the role of peroxisome proliferator-activated receptor α and dysregulated fatty acid metabolism as potential therapeutic targets.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| |
Collapse
|
40
|
Gill AW. Postnatal cardiovascular adaptation. Arch Dis Child Fetal Neonatal Ed 2019; 104:F220-F224. [PMID: 30049726 DOI: 10.1136/archdischild-2017-314453] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022]
Abstract
The heart undergoes rapid transformations in function during the transition to extrauterine life. Our understanding of the adaptive physiology underlying this process is able to inform the clinical management of infants who are struggling to complete this complex transition. Much of our knowledge of the cardiac transition is derived from the preterm infant in whom the preparative adaptations are incomplete and clinical sequelae all too common. This review will re-examine the cardiac transition highlighting the physiology that drives it and suggest appropriate clinical intervention to support the process.
Collapse
Affiliation(s)
- Andrew William Gill
- Centre for Neonatal Research and Education, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
41
|
Pepin ME, Koentges C, Pfeil K, Gollmer J, Kersting S, Wiese S, Hoffmann MM, Odening KE, von zur Mühlen C, Diehl P, Stachon P, Wolf D, Wende AR, Bode C, Zirlik A, Bugger H. Dysregulation of the Mitochondrial Proteome Occurs in Mice Lacking Adiponectin Receptor 1. Front Endocrinol (Lausanne) 2019; 10:872. [PMID: 31920982 PMCID: PMC6923683 DOI: 10.3389/fendo.2019.00872] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/28/2019] [Indexed: 12/23/2022] Open
Abstract
Decreased serum adiponectin levels in type 2 diabetes has been linked to the onset of mitochondrial dysfunction in diabetic complications by impairing AMPK-SIRT1-PGC-1α signaling via impaired adiponectin receptor 1 (AdipoR1) signaling. Here, we aimed to characterize the previously undefined role of disrupted AdipoR1 signaling on the mitochondrial protein composition of cardiac, renal, and hepatic tissues as three organs principally associated with diabetic complications. Comparative proteomics were performed in mitochondria isolated from the heart, kidneys and liver of Adipor1 -/- mice. A total of 790, 1,573, and 1,833 proteins were identified in cardiac, renal and hepatic mitochondria, respectively. While 121, 98, and 78 proteins were differentially regulated in cardiac, renal, and hepatic tissue of Adipor1-/- mice, respectively; only 15 proteins were regulated in the same direction across all investigated tissues. Enrichment analysis of differentially expressed proteins revealed disproportionate representation of proteins involved in oxidative phosphorylation conserved across tissue types. Curated pathway analysis identified HNF4, NRF1, LONP, RICTOR, SURF1, insulin receptor, and PGC-1α as candidate upstream regulators. In high fat-fed non-transgenic mice with obesity and insulin resistance, AdipoR1 gene expression was markedly reduced in heart (-70%), kidney (-80%), and liver (-90%) (all P < 0.05) as compared to low fat-fed mice. NRF1 was the only upstream regulator downregulated both in Adipor1-/- mice and in high fat-fed mice, suggesting common mechanisms of regulation. Thus, AdipoR1 signaling regulates mitochondrial protein composition across all investigated tissues in a functionally conserved, yet molecularly distinct, manner. The biological significance and potential implications of impaired AdipoR1 signaling are discussed.
Collapse
Affiliation(s)
- Mark E. Pepin
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christoph Koentges
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
| | - Katharina Pfeil
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Johannes Gollmer
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Sophia Kersting
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Ulm University, Ulm, Germany
| | - Michael M. Hoffmann
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Katja E. Odening
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Constantin von zur Mühlen
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Diehl
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Adam R. Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christoph Bode
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Zirlik
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
- Division of Cardiology, Medical University of Graz, Graz, Austria
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heiko Bugger
- Division of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany
- Division of Cardiology, Medical University of Graz, Graz, Austria
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- *Correspondence: Heiko Bugger
| |
Collapse
|
42
|
Beak JY, Kang HS, Huang W, Myers PH, Bowles DE, Jetten AM, Jensen BC. The nuclear receptor RORα protects against angiotensin II-induced cardiac hypertrophy and heart failure. Am J Physiol Heart Circ Physiol 2019; 316:H186-H200. [PMID: 30387679 PMCID: PMC6383360 DOI: 10.1152/ajpheart.00531.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/04/2018] [Accepted: 10/19/2018] [Indexed: 01/01/2023]
Abstract
The nuclear receptor retinoic acid-related orphan receptor-α (RORα) regulates numerous critical biological processes, including central nervous system development, lymphocyte differentiation, and lipid metabolism. RORα has been recently identified in the heart, but very little is known about its role in cardiac physiology. We sought to determine whether RORα regulates myocardial hypertrophy and cardiomyocyte survival in the context of angiotensin II (ANG II) stimulation. For in vivo characterization of the function of RORα in the context of pathological cardiac hypertrophy and heart failure, we used the "staggerer" (RORαsg/sg) mouse, which harbors a germline mutation encoding a truncated and globally nonfunctional RORα. RORαsg/sg and wild-type littermate mice were infused with ANG II or vehicle for 14 days. For in vitro experiments, we overexpressed or silenced RORα in neonatal rat ventricular myocytes (NRVMs) and human cardiac fibroblasts exposed to ANG II. RORαsg/sg mice developed exaggerated myocardial hypertrophy and contractile dysfunction after ANG II treatment. In vitro gain- and loss-of-function experiments were consistent with the discovery that RORα inhibits ANG II-induced pathological hypertrophy and cardiomyocyte death in vivo. RORα directly repressed IL-6 transcription. Loss of RORα function led to enhanced IL-6 expression, proinflammatory STAT3 activation (phopho-STAT3 Tyr705), and decreased mitochondrial number and function, oxidative stress, hypertrophy, and death of cardiomyocytes upon ANG II exposure. RORα was less abundant in failing compared with nonfailing human heart tissue. In conclusion, RORα protects against ANG II-mediated pathological hypertrophy and heart failure by suppressing the IL-6-STAT3 pathway and enhancing mitochondrial function. NEW & NOTEWORTHY Mice lacking retinoic acid-related orphan receptor-α (RORα) develop exaggerated cardiac hypertrophy after angiotensin II infusion. Loss of RORα leads to enhanced IL-6 expression and NF-κB nuclear translocation. RORα maintains mitochondrial function and reduces oxidative stress after angiotensin II. The abundance of RORα is reduced in failing mouse and human hearts.
Collapse
MESH Headings
- Angiotensin II/toxicity
- Animals
- Cardiomegaly/etiology
- Cardiomegaly/genetics
- Cardiomegaly/metabolism
- Cells, Cultured
- Female
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Heart Failure/etiology
- Heart Failure/genetics
- Heart Failure/metabolism
- Humans
- Interleukin-6/metabolism
- Loss of Function Mutation
- Mice
- Mice, Inbred C57BL
- Middle Aged
- Mitochondria, Heart/metabolism
- Myocardial Contraction
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- Nuclear Receptor Subfamily 1, Group F, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism
- Rats
- Rats, Sprague-Dawley
- STAT3 Transcription Factor/metabolism
Collapse
Affiliation(s)
- Ju Youn Beak
- McAllister Heart Institute University of North Carolina School of Medicine , Chapel Hill, North Carolina
| | - Hong Soon Kang
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park, North Carolina
| | - Wei Huang
- McAllister Heart Institute University of North Carolina School of Medicine , Chapel Hill, North Carolina
| | - Page H Myers
- Veterinary Medicine Section, Comparative Medicine Branch, National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park, North Carolina
| | - Dawn E Bowles
- Department of Surgery, Duke University Medical Center , Durham, North Carolina
| | - Anton M Jetten
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park, North Carolina
| | - Brian C Jensen
- McAllister Heart Institute University of North Carolina School of Medicine , Chapel Hill, North Carolina
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
43
|
Saffitz JE. Editorial commentary: Nuclear receptors and a network biology approach to understanding and treating heart disease. Trends Cardiovasc Med 2018; 29:438-439. [PMID: 30559047 DOI: 10.1016/j.tcm.2018.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 11/26/2022]
Affiliation(s)
- Jeffrey E Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston 02215, MA, United States.
| |
Collapse
|
44
|
Piquereau J, Ventura-Clapier R. Maturation of Cardiac Energy Metabolism During Perinatal Development. Front Physiol 2018; 9:959. [PMID: 30072919 PMCID: PMC6060230 DOI: 10.3389/fphys.2018.00959] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/29/2018] [Indexed: 12/26/2022] Open
Abstract
As one of the highest energy consumer organ in mammals, the heart has to be provided with a high amount of energy as soon as its first beats in utero. During the development of this organ, energy is produced within the cardiac muscle cell depending on substrate availability, oxygen pressure and cardiac workload that drastically change at birth. Thus, energy metabolism relying essentially on carbohydrates in fetal heart is very different from the adult one and birth is the trigger of a profound maturation which ensures the transition to a highly oxidative metabolism depending on lipid utilization. To face the substantial increase in cardiac workload resulting from the growth of the organism during the postnatal period, the heart not only develops its capacity for energy production but also undergoes a hypertrophic growth to adapt its contractile capacity to its new function. This leads to a profound cytoarchitectural remodeling of the cardiomyocyte which becomes a highly compartmentalized structure. As a consequence, within the mature cardiac muscle, energy transfer between energy producing and consuming compartments requires organized energy transfer systems that are established in the early postnatal life. This review aims at describing the major rearrangements of energy metabolism during the perinatal development.
Collapse
Affiliation(s)
- Jérôme Piquereau
- Signalling and Cardiovascular Pathophysiology - UMR-S 1180, Université Paris-Sud, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Châtenay-Malabry, France
| | - Renée Ventura-Clapier
- Signalling and Cardiovascular Pathophysiology - UMR-S 1180, Université Paris-Sud, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
45
|
Whitehead N, Gill JF, Brink M, Handschin C. Moderate Modulation of Cardiac PGC-1α Expression Partially Affects Age-Associated Transcriptional Remodeling of the Heart. Front Physiol 2018; 9:242. [PMID: 29618980 PMCID: PMC5871735 DOI: 10.3389/fphys.2018.00242] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/06/2018] [Indexed: 01/09/2023] Open
Abstract
Aging is associated with a decline in cardiac function due to a decreased myocardial reserve. This adverse cardiac remodeling comprises of a variety of changes, including a reduction in mitochondrial function and a decline in the expression of the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a central regulator of mitochondrial biogenesis and metabolic adaptation in the myocardium. To study the etiological involvement of PGC-1α in cardiac aging, we used mouse models mimicking the modest down- and upregulation of this coactivator in the old and the exercised heart, respectively. Young mice with reduced cardiac expression of PGC-1α recapitulated part of the age-related impairment in mitochondrial gene expression, but otherwise did not aggravate the aging process. Inversely however, moderate overexpression of PGC-1α counteracts numerous key age-related remodeling changes, e.g., by improving blood pressure, age-associated apoptosis, and collagen accumulation, as well as in the expression of many, but not all cardiac genes involved in mitochondrial biogenesis, dynamics, metabolism, calcium handling and contractility. Thus, while the reduction of PGC-1α in the heart is insufficient to cause an aging phenotype, moderate overexpression reduces pathological remodeling of older hearts and could thereby contribute to the beneficial effects of exercise on cardiac function in aging.
Collapse
Affiliation(s)
| | | | - Marijke Brink
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | | |
Collapse
|
46
|
Diguet N, Trammell SAJ, Tannous C, Deloux R, Piquereau J, Mougenot N, Gouge A, Gressette M, Manoury B, Blanc J, Breton M, Decaux JF, Lavery GG, Baczkó I, Zoll J, Garnier A, Li Z, Brenner C, Mericskay M. Nicotinamide Riboside Preserves Cardiac Function in a Mouse Model of Dilated Cardiomyopathy. Circulation 2017; 137:2256-2273. [PMID: 29217642 DOI: 10.1161/circulationaha.116.026099] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 11/06/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Myocardial metabolic impairment is a major feature in chronic heart failure. As the major coenzyme in fuel oxidation and oxidative phosphorylation and a substrate for enzymes signaling energy stress and oxidative stress response, nicotinamide adenine dinucleotide (NAD+) is emerging as a metabolic target in a number of diseases including heart failure. Little is known on the mechanisms regulating homeostasis of NAD+ in the failing heart. METHODS To explore possible alterations of NAD+ homeostasis in the failing heart, we quantified the expression of NAD+ biosynthetic enzymes in the human failing heart and in the heart of a mouse model of dilated cardiomyopathy (DCM) triggered by Serum Response Factor transcription factor depletion in the heart (SRFHKO) or of cardiac hypertrophy triggered by transverse aorta constriction. We studied the impact of NAD+ precursor supplementation on cardiac function in both mouse models. RESULTS We observed a 30% loss in levels of NAD+ in the murine failing heart of both DCM and transverse aorta constriction mice that was accompanied by a decrease in expression of the nicotinamide phosphoribosyltransferase enzyme that recycles the nicotinamide precursor, whereas the nicotinamide riboside kinase 2 (NMRK2) that phosphorylates the nicotinamide riboside precursor is increased, to a higher level in the DCM (40-fold) than in transverse aorta constriction (4-fold). This shift was also observed in human failing heart biopsies in comparison with nonfailing controls. We show that the Nmrk2 gene is an AMP-activated protein kinase and peroxisome proliferator-activated receptor α responsive gene that is activated by energy stress and NAD+ depletion in isolated rat cardiomyocytes. Nicotinamide riboside efficiently rescues NAD+ synthesis in response to FK866-mediated inhibition of nicotinamide phosphoribosyltransferase and stimulates glycolysis in cardiomyocytes. Accordingly, we show that nicotinamide riboside supplementation in food attenuates the development of heart failure in mice, more robustly in DCM, and partially after transverse aorta constriction, by stabilizing myocardial NAD+ levels in the failing heart. Nicotinamide riboside treatment also robustly increases the myocardial levels of 3 metabolites, nicotinic acid adenine dinucleotide, methylnicotinamide, and N1-methyl-4-pyridone-5-carboxamide, that can be used as validation biomarkers for the treatment. CONCLUSIONS The data show that nicotinamide riboside, the most energy-efficient among NAD precursors, could be useful for treatment of heart failure, notably in the context of DCM, a disease with few therapeutic options.
Collapse
Affiliation(s)
- Nicolas Diguet
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Samuel A J Trammell
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City (S.A.J.T., C.B.)
| | - Cynthia Tannous
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Robin Deloux
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | | | - Nathalie Mougenot
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Plateforme PECMV, UMS28, Paris, France (N.M.)
| | - Anne Gouge
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Mélanie Gressette
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Boris Manoury
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Jocelyne Blanc
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Marie Breton
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Jean-François Decaux
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, United Kingdom (G.G.L.)
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Hungary (I.B.)
| | - Joffrey Zoll
- Physiology Department, Faculty of Medicine and EA3072, Université de Strasbourg, France (J.Z.)
| | - Anne Garnier
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Zhenlin Li
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City (S.A.J.T., C.B.)
| | - Mathias Mericskay
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.).
| |
Collapse
|
47
|
Warren JS, Oka SI, Zablocki D, Sadoshima J. Metabolic reprogramming via PPARα signaling in cardiac hypertrophy and failure: From metabolomics to epigenetics. Am J Physiol Heart Circ Physiol 2017. [PMID: 28646024 DOI: 10.1152/ajpheart.00103.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Studies using omics-based approaches have advanced our knowledge of metabolic remodeling in cardiac hypertrophy and failure. Metabolomic analysis of the failing heart has revealed global changes in mitochondrial substrate metabolism. Peroxisome proliferator-activated receptor-α (PPARα) plays a critical role in synergistic regulation of cardiac metabolism through transcriptional control. Metabolic reprogramming via PPARα signaling in heart failure ultimately propagates into myocardial energetics. However, emerging evidence suggests that the expression level of PPARα per se does not always explain the energetic state in the heart. The transcriptional activities of PPARα are dynamic, yet highly coordinated. An additional level of complexity in the PPARα regulatory mechanism arises from its ability to interact with various partners, which ultimately determines the metabolic phenotype of the diseased heart. This review summarizes our current knowledge of the PPARα regulatory mechanisms in cardiac metabolism and the possible role of PPARα in epigenetic modifications in the diseased heart. In addition, we discuss how metabolomics can contribute to a better understanding of the role of PPARα in the progression of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Junco Shibayama Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah; .,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah; and
| | - Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|