1
|
Li L, Niemann B, Knapp F, Werner S, Mühlfeld C, Schneider JP, Jurida LM, Molenda N, Schmitz ML, Yin X, Mayr M, Schulz R, Kracht M, Rohrbach S. Comparison of the stage-dependent mitochondrial changes in response to pressure overload between the diseased right and left ventricle in the rat. Basic Res Cardiol 2024; 119:587-611. [PMID: 38758338 DOI: 10.1007/s00395-024-01051-3] [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: 08/28/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024]
Abstract
The right ventricle (RV) differs developmentally, anatomically and functionally from the left ventricle (LV). Therefore, characteristics of LV adaptation to chronic pressure overload cannot easily be extrapolated to the RV. Mitochondrial abnormalities are considered a crucial contributor in heart failure (HF), but have never been compared directly between RV and LV tissues and cardiomyocytes. To identify ventricle-specific mitochondrial molecular and functional signatures, we established rat models with two slowly developing disease stages (compensated and decompensated) in response to pulmonary artery banding (PAB) or ascending aortic banding (AOB). Genome-wide transcriptomic and proteomic analyses were used to identify differentially expressed mitochondrial genes and proteins and were accompanied by a detailed characterization of mitochondrial function and morphology. Two clearly distinguishable disease stages, which culminated in a comparable systolic impairment of the respective ventricle, were observed. Mitochondrial respiration was similarly impaired at the decompensated stage, while respiratory chain activity or mitochondrial biogenesis were more severely deteriorated in the failing LV. Bioinformatics analyses of the RNA-seq. and proteomic data sets identified specifically deregulated mitochondrial components and pathways. Although the top regulated mitochondrial genes and proteins differed between the RV and LV, the overall changes in tissue and cardiomyocyte gene expression were highly similar. In conclusion, mitochondrial dysfuntion contributes to disease progression in right and left heart failure. Ventricle-specific differences in mitochondrial gene and protein expression are mostly related to the extent of observed changes, suggesting that despite developmental, anatomical and functional differences mitochondrial adaptations to chronic pressure overload are comparable in both ventricles.
Collapse
MESH Headings
- Animals
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Male
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Heart Failure/pathology
- Heart Failure/genetics
- Disease Models, Animal
- Proteomics
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/genetics
- Ventricular Dysfunction, Right/pathology
- Ventricular Function, Right
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Heart Ventricles/pathology
- Rats
- Ventricular Function, Left
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/genetics
- Transcriptome
- Rats, Sprague-Dawley
- Mitochondrial Proteins/metabolism
- Mitochondrial Proteins/genetics
Collapse
Affiliation(s)
- Ling Li
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Bernd Niemann
- Department of Cardiac and Vascular Surgery, Justus Liebig University Giessen, Rudolf-Buchheim-Street. 8, 35392, Giessen, Germany
| | - Fabienne Knapp
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Sebastian Werner
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Christian Mühlfeld
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Street. 1, 30625, Hannover, Germany
| | - Jan Philipp Schneider
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Street. 1, 30625, Hannover, Germany
| | - Liane M Jurida
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Nicole Molenda
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus Liebig University Giessen, Friedrichstr. 24, 35392, Giessen, Germany
| | - Xiaoke Yin
- School of Cardiovascular and Metabolic Medicine and Science, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Manuel Mayr
- School of Cardiovascular and Metabolic Medicine and Science, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany.
| |
Collapse
|
2
|
Rai NK, Venugopal H, Rajesh R, Ancha P, Venkatesh S. Mitochondrial complex-1 as a therapeutic target for cardiac diseases. Mol Cell Biochem 2024:10.1007/s11010-024-05074-1. [PMID: 39033212 DOI: 10.1007/s11010-024-05074-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Mitochondrial dysfunction is critical for the development and progression of cardiovascular diseases (CVDs). Complex-1 (CI) is an essential component of the mitochondrial electron transport chain that participates in oxidative phosphorylation and energy production. CI is the largest multisubunit complex (~ 1 Mda) and comprises 45 protein subunits encoded by seven mt-DNA genes and 38 nuclear genes. These subunits function as the enzyme nicotinamide adenine dinucleotide hydrogen (NADH): ubiquinone oxidoreductase. CI dysregulation has been implicated in various CVDs, including heart failure, ischemic heart disease, pressure overload, hypertrophy, and cardiomyopathy. Several studies demonstrated that impaired CI function contributes to increased oxidative stress, altered calcium homeostasis, and mitochondrial DNA damage in cardiac cells, leading to cardiomyocyte dysfunction and apoptosis. CI dysfunction has been associated with endothelial dysfunction, inflammation, and vascular remodeling, critical processes in developing atherosclerosis and hypertension. Although CI is crucial in physiological and pathological conditions, no potential therapeutics targeting CI are available to treat CVDs. We believe that a lack of understanding of CI's precise mechanisms and contributions to CVDs limits the development of therapeutic strategies. In this review, we comprehensively analyze the role of CI in cardiovascular health and disease to shed light on its potential therapeutic target role in CVDs.
Collapse
Affiliation(s)
- Neeraj Kumar Rai
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, School of Medicine, West Virginia University, Morgantown, 26505, WV, USA
- Nora Eccles Harrison Cardiovascular Research and Training Institute, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Harikrishnan Venugopal
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ritika Rajesh
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, School of Medicine, West Virginia University, Morgantown, 26505, WV, USA
| | - Pranavi Ancha
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, School of Medicine, West Virginia University, Morgantown, 26505, WV, USA
| | - Sundararajan Venkatesh
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, School of Medicine, West Virginia University, Morgantown, 26505, WV, USA.
| |
Collapse
|
3
|
Zeng C, Wu J, Li J. Pyruvate Kinase M2: A Potential Regulator of Cardiac Injury Through Glycolytic and Non-glycolytic Pathways. J Cardiovasc Pharmacol 2024; 84:1-9. [PMID: 38560918 PMCID: PMC11230662 DOI: 10.1097/fjc.0000000000001568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
ABSTRACT Adult animals are unable to regenerate heart cells due to postnatal cardiomyocyte cycle arrest, leading to higher mortality rates in cardiomyopathy. However, reprogramming of energy metabolism in cardiomyocytes provides a new perspective on the contribution of glycolysis to repair, regeneration, and fibrosis after cardiac injury. Pyruvate kinase (PK) is a key enzyme in the glycolysis process. This review focuses on the glycolysis function of PKM2, although PKM1 and PKM2 both play significant roles in the process after cardiac injury. PKM2 exists in both low-activity dimer and high-activity tetramer forms. PKM2 dimers promote aerobic glycolysis but have low catalytic activity, leading to the accumulation of glycolytic intermediates. These intermediates enter the pentose phosphate pathway to promote cardiomyocyte proliferation and heart regeneration. Additionally, they activate adenosine triphosphate (ATP)-sensitive K + (K ATP ) channels, protecting the heart against ischemic damage. PKM2 tetramers function similar to PKM1 in glycolysis, promoting pyruvate oxidation and subsequently ATP generation to protect the heart from ischemic damage. They also activate KDM5 through the accumulation of αKG, thereby promoting cardiomyocyte proliferation and cardiac regeneration. Apart from glycolysis, PKM2 interacts with transcription factors like Jmjd4, RAC1, β-catenin, and hypoxia-inducible factor (HIF)-1α, playing various roles in homeostasis maintenance, remodeling, survival regulation, and neovascularization promotion. However, PKM2 has also been implicated in promoting cardiac fibrosis through mechanisms like sirtuin (SIRT) 3 deletion, TG2 expression enhancement, and activation of transforming growth factor-β1 (TGF-β1)/Smad2/3 and Jak2/Stat3 signals. Overall, PKM2 shows promising potential as a therapeutic target for promoting cardiomyocyte proliferation and cardiac regeneration and addressing cardiac fibrosis after injury.
Collapse
Affiliation(s)
- Chenxin Zeng
- The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Jiangfeng Wu
- The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, China; and
| | - Junming Li
- The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
4
|
Wang XB, Cui NH, Fang ZQ, Gao MJ, Cai D. Platelet bioenergetic profiling uncovers a metabolic pattern of high dependency on mitochondrial fatty acid oxidation in type 2 diabetic patients who developed in-stent restenosis. Redox Biol 2024; 72:103146. [PMID: 38579589 PMCID: PMC11000186 DOI: 10.1016/j.redox.2024.103146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024] Open
Abstract
Although platelet bioenergetic dysfunction is evident early in the pathogenesis of diabetic macrovascular complications, the bioenergetic characteristics in type 2 diabetic patients who developed coronary in-stent restenosis (ISR) and their effects on platelet function remain unclear. Here, we performed platelet bioenergetic profiling to characterize the bioenergetic alterations in 28 type 2 diabetic patients with ISR compared with 28 type 2 diabetic patients without ISR (non-ISR) and 28 healthy individuals. Generally, platelets from type 2 diabetic patients with ISR exhibited a specific bioenergetic alteration characterized by high dependency on fatty acid (FA) oxidation, which subsequently induced complex III deficiency, causing decreased mitochondrial respiration, increased mitochondrial oxidant production, and low efficiency of mitochondrial ATP generation. This pattern of bioenergetic dysfunction showed close relationships with both α-granule and dense granule secretion as measured by surface P-selectin expression, ATP release, and profiles of granule cargo proteins in platelet releasates. Importantly, ex vivo reproduction of high dependency on FA oxidation by exposing non-ISR platelets to its agonist mimicked the bioenergetic dysfunction observed in ISR platelets and enhanced platelet secretion, whereas pharmaceutical inhibition of FA oxidation normalized the respiratory and redox states of ISR platelets and diminished platelet secretion. Further, causal mediation analyses identified a strong association between high dependency on FA oxidation and increased angiographical severity of ISR, which was significantly mediated by the status of platelet secretion. Our findings, for the first time, uncover a pattern of bioenergetic dysfunction in ISR and enhance current understanding of the mechanistic link of high dependency on FA oxidation to platelet abnormalities in the context of diabetes.
Collapse
Affiliation(s)
- Xue-Bin Wang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Ning-Hua Cui
- Zhengzhou Key Laboratory of Children's Infection and Immunity, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zi-Qi Fang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mi-Jie Gao
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dan Cai
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| |
Collapse
|
5
|
Zhang L, Xie F, Zhang F, Lu B. The potential roles of exosomes in pathological cardiomyocyte hypertrophy mechanisms and therapy: A review. Medicine (Baltimore) 2024; 103:e37994. [PMID: 38669371 PMCID: PMC11049793 DOI: 10.1097/md.0000000000037994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Pathological cardiac hypertrophy, characterized by the enlargement of cardiac muscle cells, leads to serious cardiac conditions and stands as a major global health issue. Exosomes, comprising small lipid bilayer vesicles, are produced by various cell types and found in numerous bodily fluids. They play a pivotal role in intercellular communication by transferring bioactive cargos to recipient cells or activating signaling pathways in target cells. Exosomes from cardiomyocytes, endothelial cells, fibroblasts, and stem cells are key in regulating processes like cardiac hypertrophy, cardiomyocyte survival, apoptosis, fibrosis, and angiogenesis within the context of cardiovascular diseases. This review delves into exosomes' roles in pathological cardiac hypertrophy, first elucidating their impact on cell communication and signaling pathways. It then advances to discuss how exosomes affect key hypertrophic processes, including metabolism, fibrosis, oxidative stress, and angiogenesis. The review culminates by evaluating the potential of exosomes as biomarkers and their significance in targeted therapeutic strategies, thus emphasizing their critical role in the pathophysiology and management of cardiac hypertrophy.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Xie
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fengmei Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Beiyao Lu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Liu LX, Zheng XH, Hai JH, Zhang CM, Ti Y, Chen TS, Bu PL. SIRT3 regulates cardiolipin biosynthesis in pressure overload-induced cardiac remodeling by PPARγ-mediated mechanism. PLoS One 2024; 19:e0301990. [PMID: 38625851 PMCID: PMC11020683 DOI: 10.1371/journal.pone.0301990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/25/2024] [Indexed: 04/18/2024] Open
Abstract
Cardiac remodeling is the primary pathological feature of chronic heart failure (HF). Exploring the characteristics of cardiac remodeling in the very early stages of HF and identifying targets for intervention are essential for discovering novel mechanisms and therapeutic strategies. Silent mating type information regulation 2 homolog 3 (SIRT3), as a major mitochondrial nicotinamide adenine dinucleotide (NAD)-dependent deacetylase, is required for mitochondrial metabolism. However, whether SIRT3 plays a role in cardiac remodeling by regulating the biosynthesis of mitochondrial cardiolipin (CL) is unknown. In this study, we induced pressure overload in wild-type (WT) and SIRT3 knockout (SIRT3-/-) mice via transverse aortic constriction (TAC). Compared with WT mouse hearts, the hearts of SIRT3-/- mice exhibited more-pronounced cardiac remodeling and fibrosis, greater reactive oxygen species (ROS) production, decreased mitochondrial-membrane potential (ΔΨm), and abnormal mitochondrial morphology after TAC. Furthermore, SIRT3 deletion aggravated TAC-induced decrease in total CL content, which might be associated with the downregulation of the CL synthesis related enzymes cardiolipin synthase 1 (CRLS1) and phospholipid-lysophospholipid transacylase (TAFAZZIN). In our in vitro experiments, SIRT3 overexpression prevented angiotensin II (AngII)- induced aberrant mitochondrial function, CL biosynthesis disorder, and peroxisome proliferator-activated receptor gamma (PPARγ) downregulation in cardiomyocytes; meanwhile, SIRT3 knockdown exacerbated these effects. Moreover, the addition of GW9662, a PPARγ antagonist, partially counteracted the beneficial effects of SIRT3 overexpression. In conclusion, SIRT3 regulated PPARγ-mediated CL biosynthesis, maintained the structure and function of mitochondria, and thereby protected the myocardium against cardiac remodeling.
Collapse
Affiliation(s)
- Ling-Xin Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xue-Hui Zheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing-Han Hai
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chun-Mei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Ti
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tong-Shuai Chen
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Pei-Li Bu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Heyne E, Zeeb S, Junker C, Petzinna A, Schrepper A, Doenst T, Koch LG, Britton SL, Schwarzer M. Exercise Training Differentially Affects Skeletal Muscle Mitochondria in Rats with Inherited High or Low Exercise Capacity. Cells 2024; 13:393. [PMID: 38474357 DOI: 10.3390/cells13050393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Exercise capacity has been related to morbidity and mortality. It consists of an inherited and an acquired part and is dependent on mitochondrial function. We assessed skeletal muscle mitochondrial function in rats with divergent inherited exercise capacity and analyzed the effect of exercise training. Female high (HCR)- and low (LCR)-capacity runners were trained with individually adapted high-intensity intervals or kept sedentary. Interfibrillar (IFM) and subsarcolemmal (SSM) mitochondria from gastrocnemius muscle were isolated and functionally assessed (age: 15 weeks). Sedentary HCR presented with higher exercise capacity than LCR paralleled by higher citrate synthase activity and IFM respiratory capacity in skeletal muscle of HCR. Exercise training increased exercise capacity in both HCR and LCR, but this was more pronounced in LCR. In addition, exercise increased skeletal muscle mitochondrial mass more in LCR. Instead, maximal respiratory capacity was increased following exercise in HCRs' IFM only. The results suggest that differences in skeletal muscle mitochondrial subpopulations are mainly inherited. Exercise training resulted in different mitochondrial adaptations and in higher trainability of LCR. HCR primarily increased skeletal muscle mitochondrial quality while LCR increased mitochondrial quantity in response to exercise training, suggesting that inherited aerobic exercise capacity differentially affects the mitochondrial response to exercise training.
Collapse
Affiliation(s)
- Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany
| | - Susanne Zeeb
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany
| | - Celina Junker
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany
| | - Andreas Petzinna
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany
| | - Lauren G Koch
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University Toledo, Toledo, OH 43606, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany
| |
Collapse
|
8
|
Bhullar SK, Dhalla NS. Adaptive and maladaptive roles of different angiotensin receptors in the development of cardiac hypertrophy and heart failure. Can J Physiol Pharmacol 2024; 102:86-104. [PMID: 37748204 DOI: 10.1139/cjpp-2023-0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Angiotensin II (Ang II) is formed by the action of angiotensin-converting enzyme (ACE) in the renin-angiotensin system. This hormone is known to induce cardiac hypertrophy and heart failure and its actions are mediated by the interaction of both pro- and antihypertrophic Ang II receptors (AT1R and AT2R). Ang II is also metabolized by ACE 2 to Ang-(1-7), which elicits the activation of Mas receptors (MasR) for inducing antihypertrophic actions. Since heart failure under different pathophysiological situations is preceded by adaptive and maladaptive cardiac hypertrophy, we have reviewed the existing literature to gain some information regarding the roles of AT1R, AT2R, and MasR in both acute and chronic conditions of cardiac hypertrophy. It appears that the activation of AT1R may be involved in the development of adaptive and maladaptive cardiac hypertrophy as well as subsequent heart failure because both ACE inhibitors and AT1R antagonists exert beneficial effects. On the other hand, the activation of both AT2R and MasR may prevent the occurrence of maladaptive cardiac hypertrophy and delay the progression of heart failure, and thus therapy with different activators of these antihypertrophic receptors under chronic pathological stages may prove beneficial. Accordingly, it is suggested that a great deal of effort should be made to develop appropriate activators of both AT2R and MasR for the treatment of heart failure subjects.
Collapse
Affiliation(s)
- Sukhwinder K Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
9
|
Gao S, Liu XP, Li TT, Chen L, Feng YP, Wang YK, Yin YJ, Little PJ, Wu XQ, Xu SW, Jiang XD. Animal models of heart failure with preserved ejection fraction (HFpEF): from metabolic pathobiology to drug discovery. Acta Pharmacol Sin 2024; 45:23-35. [PMID: 37644131 PMCID: PMC10770177 DOI: 10.1038/s41401-023-01152-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is currently a preeminent challenge for cardiovascular medicine. It has a poor prognosis, increasing mortality, and is escalating in prevalence worldwide. Despite accounting for over 50% of all HF patients, the mechanistic underpinnings driving HFpEF are poorly understood, thus impeding the discovery and development of mechanism-based therapies. HFpEF is a disease syndrome driven by diverse comorbidities, including hypertension, diabetes and obesity, pulmonary hypertension, aging, and atrial fibrillation. There is a lack of high-fidelity animal models that faithfully recapitulate the HFpEF phenotype, owing primarily to the disease heterogeneity, which has hampered our understanding of the complex pathophysiology of HFpEF. This review provides an updated overview of the currently available animal models of HFpEF and discusses their characteristics from the perspective of energy metabolism. Interventional strategies for efficiently utilizing energy substrates in preclinical HFpEF models are also discussed.
Collapse
Affiliation(s)
- Si Gao
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Xue-Ping Liu
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Ting-Ting Li
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Li Chen
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yi-Ping Feng
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yu-Kun Wang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yan-Jun Yin
- School of Pharmacy, Bengbu Medical College, Bengbu, 233000, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, 4102, Australia
| | - Xiao-Qian Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Suo-Wen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Xu-Dong Jiang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China.
| |
Collapse
|
10
|
Alves PKN, Schauer A, Augstein A, Männel A, Barthel P, Joachim D, Friedrich J, Prieto ME, Moriscot AS, Linke A, Adams V. Leucine Supplementation Improves Diastolic Function in HFpEF by HDAC4 Inhibition. Cells 2023; 12:2561. [PMID: 37947639 PMCID: PMC10648219 DOI: 10.3390/cells12212561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome associated with a high morbidity and mortality rate. Leucine supplementation has been demonstrated to attenuate cardiac dysfunction in animal models of cachexia and heart failure with reduced ejection fraction (HFrEF). So far, no data exist on leucine supplementation on cardiac function in HFpEF. Thus, the current study aimed to investigate the effect of leucine supplementation on myocardial function and key signaling pathways in an established HFpEF rat model. Female ZSF1 rats were randomized into three groups: Control (untreated lean rats), HFpEF (untreated obese rats), and HFpEF_Leu (obese rats receiving standard chow enriched with 3% leucine). Leucine supplementation started at 20 weeks of age after an established HFpEF was confirmed in obese rats. In all animals, cardiac function was assessed by echocardiography at baseline and throughout the experiment. At the age of 32 weeks, hemodynamics were measured invasively, and myocardial tissue was collected for assessment of mitochondrial function and for histological and molecular analyses. Leucine had already improved diastolic function after 4 weeks of treatment. This was accompanied by improved hemodynamics and reduced stiffness, as well as by reduced left ventricular fibrosis and hypertrophy. Cardiac mitochondrial respiratory function was improved by leucine without alteration of the cardiac mitochondrial content. Lastly, leucine supplementation suppressed the expression and nuclear localization of HDAC4 and was associated with Protein kinase A activation. Our data show that leucine supplementation improves diastolic function and decreases remodeling processes in a rat model of HFpEF. Beneficial effects were associated with HDAC4/TGF-β1/Collagenase downregulation and indicate a potential use in the treatment of HFpEF.
Collapse
Affiliation(s)
- Paula Ketilly Nascimento Alves
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil;
| | - Antje Schauer
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Antje Augstein
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Anita Männel
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Peggy Barthel
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Dirk Joachim
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Janet Friedrich
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Maria-Elisa Prieto
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Anselmo Sigari Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil;
| | - Axel Linke
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| | - Volker Adams
- Laboratory of Experimental and Molecular Cardiology, TU Dresden, Heart Center Dresden, 01307 Dresden, Germany; (P.K.N.A.); (A.S.); (A.A.); (A.M.); (P.B.); (D.J.); (J.F.); (A.L.)
| |
Collapse
|
11
|
Qian J, Zhang J, Cao J, Wang X, Zhang W, Chen X. The Regulatory Effect of Receptor-Interacting Protein Kinase 3 on CaMKIIδ in TAC-Induced Myocardial Hypertrophy. Int J Mol Sci 2023; 24:14529. [PMID: 37833985 PMCID: PMC10572717 DOI: 10.3390/ijms241914529] [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: 08/16/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
Necroptosis is a newly discovered mechanism of cell death, and its key regulatory role is attributed to the interaction of receptor-interacting protein kinases (RIPKs) RIPK1 and RIPK3. Ca2+/calmodulin-dependent protein kinase (CaMKII) is a newly discovered RIPK3 substrate, and its alternative splicing plays a fundamental role in cardiovascular diseases. In the present study, we aimed to explore the role and mechanism of necroptosis and alternative splicing of CaMKIIδ in myocardial hypertrophy. Transverse aortic constriction (TAC) was performed on wild-type and knockout mice to establish the model of myocardial hypertrophy. After 3 weeks, echocardiography, cardiac index, cross-sectional area of myocardial cells, hypertrophic gene expression, myocardial damage, and fibers were assessed. Moreover, we detected the levels of inflammatory factors (IL-6 and TNF-α) and examined the expressions of necroptosis-related proteins RIPK3, RIPK1, and phosphorylated MLKL. Meanwhile, we tested the expression levels of splicing factors ASF/SF2 and SC-35 in an attempt to explore CaMKII δ. The relationship between variable splicing disorder and the expression levels of splicing factors ASF/SF2 and SC-35. Further, we also investigated CaMKII activation, oxidative stress, and mitochondrial ultrastructure. In addition, wild-type mice were administered with a recombinant adeno-associated virus (AAV) carrying RIPK3, followed by TAC surgery to construct a model of myocardial hypertrophy, and the above-mentioned indicators were tested after 3 weeks. The results showed that RIPK3 deficiency could alleviate cardiac dysfunction, myocardial injury, aggravation of necrosis, and CaMKII activation induced by TAC surgery in mice with myocardial hypertrophy. Tail vein injection of AAV could reverse cardiac dysfunction, myocardial damage, aggravation of necrosis, and CaMKII activation in mice with myocardial hypertrophy. These results proved that RIPK3 could be used as a molecular intervention target for the prevention and treatment of myocardial hypertrophy.
Collapse
Affiliation(s)
- Jianan Qian
- School of Pharmacy, Nantong University, Nantong 226001, China; (J.Q.); (J.Z.); (J.C.); (X.W.)
| | - Jingjing Zhang
- School of Pharmacy, Nantong University, Nantong 226001, China; (J.Q.); (J.Z.); (J.C.); (X.W.)
- School of Medicine, Nantong University, Nantong 226001, China
| | - Ji Cao
- School of Pharmacy, Nantong University, Nantong 226001, China; (J.Q.); (J.Z.); (J.C.); (X.W.)
| | - Xue Wang
- School of Pharmacy, Nantong University, Nantong 226001, China; (J.Q.); (J.Z.); (J.C.); (X.W.)
| | - Wei Zhang
- School of Pharmacy, Nantong University, Nantong 226001, China; (J.Q.); (J.Z.); (J.C.); (X.W.)
- School of Medicine, Nantong University, Nantong 226001, China
| | - Xiangfan Chen
- School of Pharmacy, Nantong University, Nantong 226001, China; (J.Q.); (J.Z.); (J.C.); (X.W.)
- School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
12
|
Schenkl C, Heyne E, Doenst T, Schulze PC, Nguyen TD. Targeting Mitochondrial Metabolism to Save the Failing Heart. Life (Basel) 2023; 13:life13041027. [PMID: 37109556 PMCID: PMC10143865 DOI: 10.3390/life13041027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite considerable progress in treating cardiac disorders, the prevalence of heart failure (HF) keeps growing, making it a global medical and economic burden. HF is characterized by profound metabolic remodeling, which mostly occurs in the mitochondria. Although it is well established that the failing heart is energy-deficient, the role of mitochondria in the pathophysiology of HF extends beyond the energetic aspects. Changes in substrate oxidation, tricarboxylic acid cycle and the respiratory chain have emerged as key players in regulating myocardial energy homeostasis, Ca2+ handling, oxidative stress and inflammation. This work aims to highlight metabolic alterations in the mitochondria and their far-reaching effects on the pathophysiology of HF. Based on this knowledge, we will also discuss potential metabolic approaches to improve cardiac function.
Collapse
Affiliation(s)
- Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Paul Christian Schulze
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Tien Dung Nguyen
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
13
|
Abstract
Chronic kidney disease is associated with an increased risk for the development and progression of cardiovascular disorders including hypertension, dyslipidemia, and coronary artery disease. Chronic kidney disease may also affect the myocardium through complex systemic changes, resulting in structural remodeling such as hypertrophy and fibrosis, as well as impairments in both diastolic and systolic function. These cardiac changes in the setting of chronic kidney disease define a specific cardiomyopathic phenotype known as uremic cardiomyopathy. Cardiac function is tightly linked to its metabolism, and research over the past 3 decades has revealed significant metabolic remodeling in the myocardium during the development of heart failure. Because the concept of uremic cardiomyopathy has only been recognized in recent years, there are limited data on metabolism in the uremic heart. Nonetheless, recent findings suggest overlapping mechanisms with heart failure. This work reviews key features of metabolic remodeling in the failing heart in the general population and extends this to patients with chronic kidney disease. The knowledge of similarities and differences in cardiac metabolism between heart failure and uremic cardiomyopathy may help identify new targets for mechanistic and therapeutic research on uremic cardiomyopathy.
Collapse
Affiliation(s)
- T Dung Nguyen
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | | |
Collapse
|
14
|
Ali SA, Bommaraju S, Patwa J, Khare P, Rachamalla M, Niyogi S, Datusalia AK. Melatonin Attenuates Extracellular Matrix Accumulation and Cardiac Injury Manifested by Copper. Biol Trace Elem Res 2022:10.1007/s12011-022-03509-8. [PMID: 36449149 DOI: 10.1007/s12011-022-03509-8] [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: 10/09/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022]
Abstract
Copper-induced cardiac injury is not widely reported in spite of its ability to cause oxidative damage and tissue injury. Structural and morphological changes in the cardiac tissue are triggered via oxidative stress and inflammatory responses following copper exposure. The varied and unavoidable exposure of copper through contaminated food and water warrants a safe and effective agent against its harmful effects. Since the heart is highly sensitive to changes in the redox balance, the present study was undertaken to examine the protective effects of melatonin against copper-induced cardiac injury. Sprague Dawley (SD) rats were exposed to 100 ppm of elemental copper via drinking water for 4 months. The cardiac tissue was evaluated for various biochemical, histological, and protein expression studies. Animals exposed to copper exhibited induced oxidative stress and cardiac injury compared to normal control. To this end, we found that melatonin treatment ameliorated copper-induced alterations in tissue oxidative variables like ROS, nitrate, MDA, and GSH. In addition, histological examinations unravelled decreased cardiac muscle dilation, atrophy, and cardiomyopathy in melatonin-treated rats. Furthermore, melatonin-treated rats were associated with reduced tissue copper levels, collagen deposition, α-SMA, and increased HO-1 expression as compared to rats exposed exclusively to copper. Moreover, the levels of NF-κB and cardiac markers such as CK-MB, cTnI, and cTnT were found to be decreased in the melatonin-treated animals. Altogether, melatonin-triggered increase in antioxidant capacity resulting in reduced aggregation of ECM components demonstrates the therapeutic potential of melatonin in the treatment of cardiac injury and tissue fibrosis.
Collapse
Affiliation(s)
- Syed Afroz Ali
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, 226002, India
| | - Sumadhura Bommaraju
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, 226002, India
| | - Jayant Patwa
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, 226002, India
| | - Puja Khare
- Crop Production and Protection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, Canada
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, Canada
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, 226002, India.
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, 226002, India.
| |
Collapse
|
15
|
Winzer EB, Augstein A, Schauer A, Mueller S, Fischer-Schaepmann T, Goto K, Hommel J, van Craenenbroeck EM, Wisløff U, Pieske B, Halle M, Linke A, Adams V. Impact of Different Training Modalities on Molecular Alterations in Skeletal Muscle of Patients With Heart Failure With Preserved Ejection Fraction: A Substudy of the OptimEx Trial. Circ Heart Fail 2022; 15:e009124. [DOI: 10.1161/circheartfailure.121.009124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background:
Exercise intolerance is a cardinal feature of heart failure with preserved ejection fraction and so far exercise training (ET) is the most effective treatment. Since the improvement in exercise capacity is only weakly associated with changes in diastolic function other mechanisms, like changes in the skeletal muscle, contribute to improvement in peak oxygen consumption. The aim of the present study was to analyze molecular changes in skeletal muscle of patients with heart failure with preserved ejection fraction performing different ET modalities.
Methods:
Skeletal muscle biopsies were taken at study begin and after 3 and 12 months from patients with heart failure with preserved ejection fraction randomized either into a control group (guideline based advice for ET), a high-intensity interval training group (HIIT) or a moderate continuous training group. The first 3 months of ET were supervised in-hospital followed by 9 months home-based ET. Protein and mRNA expression of atrophy-related proteins, enzyme activities of enzymes linked to energy metabolism and satellite cells (SCs) were quantified.
Results:
Exercise capacity improved 3 months after moderate continuous exercise training and HIIT. This beneficial effect was lost after 12 months. HIIT mainly improved markers of energy metabolism and the amount and function of SC, with minor changes in markers for muscle atrophy. Only slight changes were observed after moderate continuous exercise training. The molecular changes were no longer detectable after 12 months.
Conclusions:
Despite similar improvements in exercise capacity by HIIT and moderate continuous exercise training after 3 months, only HIIT altered proteins related to energy metabolism and amount/function of SC. These effects were lost after switching from in-hospital to at-home-based ET.
Registration:
URL:
https://www.clinicaltrials.gov
; Unique identifier: NCT02078947.
Collapse
Affiliation(s)
- Ephraim B. Winzer
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Antje Augstein
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Antje Schauer
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Stephan Mueller
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany (S.M., M.H.)
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.M., M.H.)
| | - Tina Fischer-Schaepmann
- Department of Internal Medicine/Cardiology, Heart Center Leipzig – University Hospital, Helios Stiftungsprofessur, Germany (T.F.-S.)
| | - Keita Goto
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Jennifer Hommel
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Emeline M. van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Belgium (E.M.v.C.)
- Department of Cardiology, Antwerp University Hospital, Belgium (E.M.v.C.)
| | - Ulrik Wisløff
- Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (U.W.)
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Germany (B.P.)
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany (S.M., M.H.)
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.M., M.H.)
| | - Axel Linke
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Germany (V.A.)
| |
Collapse
|
16
|
Mitophagy: A Potential Target for Pressure Overload-Induced Cardiac Remodelling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2849985. [PMID: 36204518 PMCID: PMC9532135 DOI: 10.1155/2022/2849985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 07/16/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022]
Abstract
The pathological mechanisms underlying cardiac remodelling and cardiac dysfunction caused by pressure overload are poorly understood. Mitochondrial damage and functional dysfunction, including mitochondrial bioenergetic disorder, oxidative stress, and mtDNA damage, contribute to heart injury caused by pressure overload. Mitophagy, an important regulator of mitochondrial homeostasis and function, is triggered by mitochondrial damage and participates in the pathological process of cardiovascular diseases. Recent studies indicate that mitophagy plays a critical role in the pressure overload model, but evidence on the causal relationship between mitophagy abnormality and pressure overload-induced heart injury is inconclusive. This review summarises the mechanism, role, and regulation of mitophagy in the pressure overload model. It also pays special attention to active compounds that may regulate mitophagy in pressure overload, which provide clues for possible clinical applications.
Collapse
|
17
|
Ca2+/Calmodulin-Dependent Protein Kinase II Regulation by Inhibitor of RIPK3 Protects against Cardiac Hypertrophy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7941374. [PMID: 36046685 PMCID: PMC9423983 DOI: 10.1155/2022/7941374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022]
Abstract
The activity of Ca2+/calmodulin-dependent protein kinase II δ (CaMKII δ) is central to the mechanisms of cardiovascular diseases. Receptor-interacting protein kinase 3- (RIPK3-) mediated necroptosis has been reported to contribute to cardiac dysfunction. However, the potential protective role of inhibition of RIPK3, a regulator of CaMKII, on cardiac hypertrophy remains unclear. The present study is aimed at investigating how the RIPK3 inhibitor GSK'872 regulates CaMKII activity and exploring its effect on hypertrophic cardiomyopathy (HCM). Wild-type (WT) and RIPK3 gene knockout (RIPK3−/−) mice were implanted subcutaneously with Alzet miniosmotic pumps (200 μL) and perfused with angiotensin II (AMP-AngII) to induce cardiac hypertrophy. After WT mice were induced by AngII for 72 hours, they were injected with GSK'872 with an intraperitoneal (IP) dose of 6 mg/kg once a day for two weeks. After this, they were physiologically examined for Echocardiography, myocardial injury, CaMKII activity, necroptosis, RIPK3 expression, mixed lineage kinase domain-like protein (MLKL) phosphorylation, and mitochondrial ultrastructure. The results indicated that deletion of the RIPK3 gene or administration of GSK'872 could reduce CaMKII activity, alleviate oxidative stress, reduce necroptosis, and reverse myocardial injury and cardiac dysfunction caused by AngII-induced cardiac hypertrophy in mice. The present study demonstrated that CaMKII activation and necroptosis augment cardiac hypertrophy in a RIPK3-dependent manner, which may provide therapeutic strategies for HCM. RIPK3 inhibitor GSK'872 has a protective effect on cardiac hypertrophy and could be an efficacious targeted medicine for HCM in clinical treatment.
Collapse
|
18
|
Ramachandra CJA, Kp MMJ, Chua J, Hernandez-Resendiz S, Liehn EA, Knöll R, Gan LM, Michaëlsson E, Jonsson MKB, Ryden-Markinhuhta K, Bhat RV, Fritsche-Danielson R, Lin YH, Sadayappan S, Tang HC, Wong P, Shim W, Hausenloy DJ. Inhibiting cardiac myeloperoxidase alleviates the relaxation defect in hypertrophic cardiomyocytes. Cardiovasc Res 2022; 118:517-530. [PMID: 33705529 PMCID: PMC8803077 DOI: 10.1093/cvr/cvab077] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/12/2020] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is characterized by cardiomyocyte hypertrophy and disarray, and myocardial stiffness due to interstitial fibrosis, which result in impaired left ventricular filling and diastolic dysfunction. The latter manifests as exercise intolerance, angina, and dyspnoea. There is currently no specific treatment for improving diastolic function in HCM. Here, we investigated whether myeloperoxidase (MPO) is expressed in cardiomyocytes and provides a novel therapeutic target for alleviating diastolic dysfunction in HCM. METHODS AND RESULTS Human cardiomyocytes derived from control-induced pluripotent stem cells (iPSC-CMs) were shown to express MPO, with MPO levels being increased in iPSC-CMs generated from two HCM patients harbouring sarcomeric mutations in the MYBPC3 and MYH7 genes. The presence of cardiomyocyte MPO was associated with higher chlorination and peroxidation activity, increased levels of 3-chlorotyrosine-modified cardiac myosin binding protein-C (MYBPC3), attenuated phosphorylation of MYBPC3 at Ser-282, perturbed calcium signalling, and impaired cardiomyocyte relaxation. Interestingly, treatment with the MPO inhibitor, AZD5904, reduced 3-chlorotyrosine-modified MYBPC3 levels, restored MYBPC3 phosphorylation, and alleviated the calcium signalling and relaxation defects. Finally, we found that MPO protein was expressed in healthy adult murine and human cardiomyocytes, and MPO levels were increased in diseased hearts with left ventricular hypertrophy. CONCLUSION This study demonstrates that MPO inhibition alleviates the relaxation defect in hypertrophic iPSC-CMs through MYBPC3 phosphorylation. These findings highlight cardiomyocyte MPO as a novel therapeutic target for improving myocardial relaxation associated with HCM, a treatment strategy which can be readily investigated in the clinical setting, given that MPO inhibitors are already available for clinical testing.
Collapse
MESH Headings
- Animals
- Cardiac Myosins/genetics
- Cardiac Myosins/metabolism
- Cardiomyopathy, Hypertrophic/drug therapy
- Cardiomyopathy, Hypertrophic/enzymology
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/physiopathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line
- Disease Models, Animal
- Enzyme Inhibitors/pharmacology
- Humans
- Hypertrophy, Left Ventricular/drug therapy
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/physiopathology
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/enzymology
- Induced Pluripotent Stem Cells/pathology
- Male
- Mice, Inbred C57BL
- Mutation, Missense
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Peroxidase/antagonists & inhibitors
- Peroxidase/metabolism
- Phosphorylation
- Reactive Oxygen Species/metabolism
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
- Ventricular Function, Left/drug effects
- Mice
Collapse
Affiliation(s)
- Chrishan J A Ramachandra
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
| | - Myu Mai Ja Kp
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
| | - Jasper Chua
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Faculty of Science, National University of Singapore, Singapore,
Singapore
| | - Sauri Hernandez-Resendiz
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
| | - Elisa A Liehn
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
| | - Ralph Knöll
- Bioscience, Cardiovascular, Renal & Metabolism, BioPharmaceuticals
R&D, AstraZeneca, Gothenburg, Sweden
- Department of Medicine (MedH), Integrated Cardio Metabolic Centre
(ICMC), Heart and Vascular Theme, Karolinska Institute, Stockholm SE-171 77,
Sweden
| | - Li-Ming Gan
- Early Clinical Development, Research and Early Development Cardiovascular,
Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca,
Gothenburg, Sweden
| | - Erik Michaëlsson
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal
and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg,
Sweden
| | - Malin K B Jonsson
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal
and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg,
Sweden
| | - Katarina Ryden-Markinhuhta
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal
and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg,
Sweden
| | - Ratan V Bhat
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM),
BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Regina Fritsche-Danielson
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM),
BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ying-Hsi Lin
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine,
Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati,
OH, USA
| | - Hak Chiaw Tang
- Department of Cardiology, National Heart Centre Singapore,
Singapore, Singapore
| | - Philip Wong
- Department of Cardiology, National Heart Centre Singapore,
Singapore, Singapore
| | - Winston Shim
- Health and Social Sciences Cluster, Singapore Institute of
Technology, Singapore, Singapore
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of
Singapore, Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London,
London, UK
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia
University, Taichung, Taiwan
| |
Collapse
|
19
|
Addition of Popular Exogenous Antioxidant Agent, PBN, to Culture Media May Be an Important Step to Optimization of Myogenic Stem/Progenitor Cell Preparation Protocol. Antioxidants (Basel) 2021; 10:antiox10060959. [PMID: 34203726 PMCID: PMC8232265 DOI: 10.3390/antiox10060959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 11/17/2022] Open
Abstract
The aim of the study was to modify human skeletal muscle-derived stem/progenitor cells (SkMDS/PCs) and demonstrate the optimal cell preparation protocol for application in post-infarction hearts. We used conditioned SkMDS/PC culture medium with α-phenyl-N-tert-butyl nitrone (PBN). SkMDS/PCs were cultured under hypoxic conditions and the results were compared to the standard ones. We observed a significant increase of CD-56 positive phenotypic marker the ability to form functional myotubes, increase in the proportion of young cells in cell primary suspensions, and a decrease in the percentage of apoptotic cells among PBN-conditioned cells in normoxia an hypoxia. We also observed significantly higher levels of SOD3 expression; maintained expression of SOD1, SOD2, and CAT; a higher level of BCL2 gene expression; and a rather significant decrease in Hsp70 gene expression in PBN-conditioned SkMDS/PCs compared to the WT population under hypoxic conditions. In addition, significant increase of myogenic genes expression was observed after PBN addition to culture medium, compared to WT population under hypoxia. Interestingly, PBN addition significantly increased the lengths of telomeres under hypoxia. Based on the data obtained, we can postulate that PBN conditioning of human SkMDS/PCs could be a promising step in improving myogenic cell preparation protocol for pro-regenerative treatment of post-infarction hearts.
Collapse
|
20
|
Adams V, Wunderlich S, Mangner N, Hommel J, Esefeld K, Gielen S, Halle M, Ellingsen Ø, Van Craenenbroeck EM, Wisløff U, Pieske B, Linke A, Winzer EB. Ubiquitin-proteasome-system and enzymes of energy metabolism in skeletal muscle of patients with HFpEF and HFrEF. ESC Heart Fail 2021; 8:2556-2568. [PMID: 33955206 PMCID: PMC8318515 DOI: 10.1002/ehf2.13405] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/17/2021] [Accepted: 04/22/2021] [Indexed: 01/14/2023] Open
Abstract
Background Skeletal muscle (SM) alterations contribute to exercise intolerance in heart failure patients with preserved (HFpEF) or reduced (HFrEF) left ventricular ejection fraction (LVEF). Protein degradation via the ubiquitin‐proteasome‐system (UPS), nuclear apoptosis, and reduced mitochondrial energy supply is associated with SM weakness in HFrEF. These mechanisms are incompletely studied in HFpEF, and a direct comparison between these groups is missing. Methods and results Patients with HFpEF (LVEF ≥ 50%, septal E/e′ > 15 or >8 and NT‐proBNP > 220 pg/mL, n = 20), HFrEF (LVEF ≤ 35%, n = 20) and sedentary control subjects (Con, n = 12) were studied. Inflammatory markers were measured in serum, and markers of the UPS, nuclear apoptosis, and energy metabolism were determined in percutaneous SM biopsies. Both HFpEF and HFrEF showed increased proteolysis (MuRF‐1 protein expression, ubiquitination, and proteasome activity) with proteasome activity significantly related to interleukin‐6. Proteolysis was more pronounced in patients with lower exercise capacity as indicated by peak oxygen uptake in per cent predicted below the median. Markers of apoptosis did not differ between groups. Mitochondrial energy supply was reduced in HFpEF and HFrEF (complex‐I activity: −31% and −53%; malate dehydrogenase activity: −20% and −29%; both P < 0.05 vs. Con). In contrast, short‐term energy supply via creatine kinase was increased in HFpEF but decreased in HFrEF (47% and −45%; P < 0.05 vs. Con). Conclusions Similarly to HFrEF, skeletal muscle in HFpEF is characterized by increased proteolysis linked to systemic inflammation and reduced exercise capacity. Energy metabolism is disturbed in both groups; however, its regulation seems to be severity‐dependent.
Collapse
Affiliation(s)
- Volker Adams
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany.,Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| | - Sebastian Wunderlich
- Department of Internal Medicine/Cardiology, Heart Center Leipzig - University Hospital, Leipzig, Germany
| | - Norman Mangner
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| | - Jennifer Hommel
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| | - Katrin Esefeld
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Prevention and Sports Medicine, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Stephan Gielen
- Department of Cardiology, Angiology and Intensive Care, Klinikum Lippe, Detmold, Germany
| | - Martin Halle
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Prevention and Sports Medicine, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Øyvind Ellingsen
- Department of Cardiology, St. Olavs University Hospital, Trondheim, Norway.,The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Emeline M Van Craenenbroeck
- Department of Cardiology, Antwerp University Hospital, Edegem, Belgium.,Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Ulrik Wisløff
- The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Burkert Pieske
- Department Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany.,Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| | - Ephraim B Winzer
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| |
Collapse
|
21
|
Marck PV, Pessoa MT, Xu Y, Kutz LC, Collins DM, Yan Y, King C, Wang X, Duan Q, Cai L, Xie JX, Lingrel JB, Xie Z, Tian J, Pierre SV. Cardiac Oxidative Signaling and Physiological Hypertrophy in the Na/K-ATPase α1 s/sα2 s/s Mouse Model of High Affinity for Cardiotonic Steroids. Int J Mol Sci 2021; 22:ijms22073462. [PMID: 33801629 PMCID: PMC8036649 DOI: 10.3390/ijms22073462] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 11/25/2022] Open
Abstract
The Na/K-ATPase is the specific receptor for cardiotonic steroids (CTS) such as ouabain and digoxin. At pharmacological concentrations used in the treatment of cardiac conditions, CTS inhibit the ion-pumping function of Na/K-ATPase. At much lower concentrations, in the range of those reported for endogenous CTS in the blood, they stimulate hypertrophic growth of cultured cardiac myocytes through initiation of a Na/K-ATPase-mediated and reactive oxygen species (ROS)-dependent signaling. To examine a possible effect of endogenous concentrations of CTS on cardiac structure and function in vivo, we compared mice expressing the naturally resistant Na/K-ATPase α1 and age-matched mice genetically engineered to express a mutated Na/K-ATPase α1 with high affinity for CTS. In this model, total cardiac Na/K-ATPase activity, α1, α2, and β1 protein content remained unchanged, and the cardiac Na/K-ATPase dose–response curve to ouabain shifted to the left as expected. In males aged 3–6 months, increased α1 sensitivity to CTS resulted in a significant increase in cardiac carbonylated protein content, suggesting that ROS production was elevated. A moderate but significant increase of about 15% of the heart-weight-to-tibia-length ratio accompanied by an increase in the myocyte cross-sectional area was detected. Echocardiographic analyses did not reveal any change in cardiac function, and there was no fibrosis or re-expression of the fetal gene program. RNA sequencing analysis indicated that pathways related to energy metabolism were upregulated, while those related to extracellular matrix organization were downregulated. Consistent with a functional role of the latter, an angiotensin-II challenge that triggered fibrosis in the α1r/rα2s/s mouse failed to do so in the α1s/sα2s/s. Taken together, these results are indicative of a link between circulating CTS, Na/K-ATPase α1, ROS, and physiological cardiac hypertrophy in mice under baseline laboratory conditions.
Collapse
Affiliation(s)
- Pauline V. Marck
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Marco T. Pessoa
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Yunhui Xu
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Laura C. Kutz
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Dominic M. Collins
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Yanling Yan
- Department of Biomedical Sciences, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25755, USA;
| | - Cierra King
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Qiming Duan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA;
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Jeffrey X. Xie
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Jerry B. Lingrel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Jiang Tian
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA; (P.V.M.); (M.T.P.); (Y.X.); (L.C.K.); (D.M.C.); (C.K.); (X.W.); (L.C.); (Z.X.); (J.T.)
- Correspondence: ; Tel.: +1-(304)-696-3505
| |
Collapse
|
22
|
Mangner N, Garbade J, Heyne E, van den Berg M, Winzer EB, Hommel J, Sandri M, Jozwiak-Nozdrzykowska J, Meyer AL, Lehmann S, Schmitz C, Malfatti E, Schwarzer M, Ottenheijm CAC, Bowen TS, Linke A, Adams V. Molecular Mechanisms of Diaphragm Myopathy in Humans With Severe Heart Failure. Circ Res 2021; 128:706-719. [PMID: 33535772 DOI: 10.1161/circresaha.120.318060] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Norman Mangner
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Jens Garbade
- Department of Cardiac Surgery (J.G., S.L.), Heart Center Leipzig - University Hospital, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Germany (E.H., M.S.)
| | | | - Ephraim B Winzer
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Jennifer Hommel
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Marcus Sandri
- Department of Cardiology (M.S., J.J.-N.), Heart Center Leipzig - University Hospital, Germany
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Germany (E.H., M.S.)
| | | | - Anna L Meyer
- Cardiac Surgery, Heart and Marfan Center, University of Heidelberg, Germany (A.L.M.)
| | - Sven Lehmann
- Department of Cardiac Surgery (J.G., S.L.), Heart Center Leipzig - University Hospital, Germany
| | - Clara Schmitz
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Edoardo Malfatti
- Neurology, Centre de Référence Maladies Neuromusculaires Nord-Est-Ile-de-France, CHU Raymond-Poincaré, Garches, France (E.M.). U1179 UVSQ-INSERM, Université Versailles-Saint-Quentin-en-Yvelines, France
| | | | - Coen A C Ottenheijm
- Physiology, Amsterdam UMC (location VUmc), the Netherlands (M.v.d.B., C.A.C.O.)
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom (T.S.B.)
| | - Axel Linke
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany (A.L., V.A.)
| | - Volker Adams
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany (A.L., V.A.)
| |
Collapse
|
23
|
PARIS-DJ-1 Interaction Regulates Mitochondrial Functions in Cardiomyocytes, Which Is Critically Important in Cardiac Hypertrophy. Mol Cell Biol 2020; 41:MCB.00106-20. [PMID: 33077496 DOI: 10.1128/mcb.00106-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/05/2020] [Indexed: 11/20/2022] Open
Abstract
Mitochondrial dysfunction is one of the major pathological attributes of cardiac hypertrophy and is associated with reduced expression of PGC1α in cardiomyocytes. However, the transcriptional regulation of PGC1α remains elusive. Here, we show that parkin interacting substrate (PARIS), a KRAB zinc finger protein, prevented PGC1α transcription despite the induction of cardiomyocytes with hypertrophic stimuli. Moreover, PARIS expression and its nuclear localization are enhanced in hypertrophy both in vitro and in vivo Knocking down PARIS resulted in mitochondrial biogenesis and improved respiration and other biochemical features that were compromised during hypertrophy. Furthermore, a PARIS-dependent proteome showed exclusive binding of a deSUMOylating protein called DJ-1 to PARIS in control cells, while this interaction is completely abrogated in hypertrophied cells. We further demonstrate that proteasomal degradation of DJ-1 under oxidative stress led to augmented PARIS SUMOylation and consequent repression of PGC1α promoter activity. SUMOylation-resistant mutants of PARIS failed to repress PGC1α, suggesting a critical role for PARIS SUMOylation in hypertrophy. The present study, therefore, proposes a novel regulatory pathway where DJ-1 acts as an oxidative stress sensor and contributes to the feedback loop governing PARIS-mediated mitochondrial function.
Collapse
|
24
|
Miranda-Silva D, G Rodrigues P, Alves E, Rizo D, Fonseca ACRG, Lima T, Baganha F, Conceição G, Sousa C, Gonçalves A, Miranda I, Vasques-Nóvoa F, Magalhães J, Leite-Moreira A, Falcão-Pires I. Mitochondrial Reversible Changes Determine Diastolic Function Adaptations During Myocardial (Reverse) Remodeling. Circ Heart Fail 2020; 13:e006170. [PMID: 33176457 DOI: 10.1161/circheartfailure.119.006170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Often, pressure overload-induced myocardial remodeling does not undergo complete reverse remodeling after decreasing afterload. Recently, mitochondrial abnormalities and oxidative stress have been successively implicated in the pathogenesis of several chronic pressure overload cardiac diseases. Therefore, we aim to clarify the myocardial energetic dysregulation in (reverse) remodeling, mainly focusing on the mitochondria. METHODS Thirty-five Wistar Han male rats randomly underwent sham or ascending (supravalvular) aortic banding procedure. Echocardiography revealed that banding induced concentric hypertrophy and diastolic dysfunction (early diastolic transmitral flow velocity to peak early-diastolic annular velocity ratio, E/E': sham, 13.6±2.1, banding, 18.5±4.1, P=0.014) accompanied by increased oxidative stress (dihydroethidium fluorescence: sham, 1.6×108±6.1×107, banding, 2.6×108±4.5×107, P<0.001) and augmented mitochondrial function. After 8 to 9 weeks, half of the banding animals underwent overload relief by an aortic debanding surgery (n=10). RESULTS Two weeks later, hypertrophy decreased with the decline of oxidative stress (dihydroethidium fluorescence: banding, 2.6×108±4.5×107, debanding, 1.96×108±6.8×107, P<0.001) and diastolic dysfunction improved simultaneously (E/E': banding, 18.5±4.1, debanding, 15.1±1.8, P=0.029). The reduction of energetic demands imposed by overload relief allowed the mitochondria to reduce its activity and myocardial levels of phosphocreatine, phosphocreatine/ATP, and ATP/ADP to normalize in debanding towards sham values (phosphocreatine: sham, 38.4±7.4, debanding, 35.6±8.7, P=0.71; phosphocreatine/ATP: sham, 1.22±0.23 debanding, 1.11±0.24, P=0.59; ATP/ADP: sham, 6.2±0.9, debanding, 5.6±1.6, P=0.66). Despite the decreased mitochondrial area, complex III and V expression increased in debanding compared with sham or banding. Autophagy and mitophagy-related markers increased in banding and remained higher in debanding rats. CONCLUSIONS During compensatory and maladaptive hypertrophy, mitochondria become more active. However, as the disease progresses, the myocardial energetic demands increase and the myocardium becomes energy deficient. During reverse remodeling, the concomitant attenuation of cardiac hypertrophy and oxidative stress allowed myocardial energetics, left ventricle hypertrophy, and diastolic dysfunction to recover. Autophagy and mitophagy are probably involved in the myocardial adaptation to overload and to unload. We conclude that these mitochondrial reversible changes underlie diastolic function adaptations during myocardial (reverse) remodeling.
Collapse
Affiliation(s)
- Daniela Miranda-Silva
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Patrícia G Rodrigues
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Estela Alves
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - David Rizo
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - Ana Catarina R G Fonseca
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Portugal (A.C.R.G.F.)
| | - Tânia Lima
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Fabiana Baganha
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Gloria Conceição
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Cláudia Sousa
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Alexandre Gonçalves
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Isabel Miranda
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Francisco Vasques-Nóvoa
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - José Magalhães
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| |
Collapse
|
25
|
Adams V, Gußen V, Zozulya S, Cruz A, Moriscot A, Linke A, Labeit S. Small-Molecule Chemical Knockdown of MuRF1 in Melanoma Bearing Mice Attenuates Tumor Cachexia Associated Myopathy. Cells 2020; 9:E2272. [PMID: 33050629 PMCID: PMC7600862 DOI: 10.3390/cells9102272] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
: Patients with malignant tumors frequently suffer during disease progression from a syndrome referred to as cancer cachexia (CaCax): CaCax includes skeletal muscle atrophy and weakness, loss of bodyweight, and fat tissues. Currently, there are no FDA (Food and Drug Administration) approved treatments available for CaCax. Here, we studied skeletal muscle atrophy and dysfunction in a murine CaCax model by injecting B16F10 melanoma cells into mouse thighs and followed mice during melanoma outgrowth. Skeletal muscles developed progressive weakness as detected by wire hang tests (WHTs) during days 13-23. Individual muscles analyzed at day 24 had atrophy, mitochondrial dysfunction, augmented metabolic reactive oxygen species (ROS) stress, and a catabolically activated ubiquitin proteasome system (UPS), including upregulated MuRF1. Accordingly, we tested as an experimental intervention of recently identified small molecules, Myomed-205 and -946, that inhibit MuRF1 activity and MuRF1/MuRF2 expression. Results indicate that MuRF1 inhibitor fed attenuated induction of MuRF1 in tumor stressed muscles. In addition, the compounds augmented muscle performance in WHTs and attenuated muscle weight loss. Myomed-205 and -946 also rescued citrate synthase and complex-1 activities in tumor-stressed muscles, possibly suggesting that mitochondrial-metabolic and muscle wasting effects in this CaCax model are mechanistically connected. Inhibition of MuRF1 during tumor cachexia may represent a suitable strategy to attenuate skeletal muscle atrophy and dysfunction.
Collapse
Affiliation(s)
- Volker Adams
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, 1307 Dresden, Germany; (V.G.); (A.L.)
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, 01067 Dresden, Germany
| | - Victoria Gußen
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, 1307 Dresden, Germany; (V.G.); (A.L.)
| | - Sergey Zozulya
- Department of Drug Research, Enamine-Bienta Ltd., 02000 Kiev, Ukraine;
| | - André Cruz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (A.C.); (A.M.)
| | - Anselmo Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (A.C.); (A.M.)
| | - Axel Linke
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, 1307 Dresden, Germany; (V.G.); (A.L.)
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, 01067 Dresden, Germany
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany;
| |
Collapse
|
26
|
Zhou N, Chen X, Xi J, Ma B, Leimena C, Stoll S, Qin G, Wang C, Qiu H. Genomic characterization reveals novel mechanisms underlying the valosin-containing protein-mediated cardiac protection against heart failure. Redox Biol 2020; 36:101662. [PMID: 32795937 PMCID: PMC7426568 DOI: 10.1016/j.redox.2020.101662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/20/2020] [Accepted: 07/26/2020] [Indexed: 12/22/2022] Open
Abstract
Chronic hypertension is a key risk factor for heart failure. However, the underlying molecular mechanisms are not fully understood. Our previous studies found that the valosin-containing protein (VCP), an ATPase-associated protein, was significantly decreased in the hypertensive heart tissues. In this study, we tested the hypothesis that restoration of VCP protected the heart against pressure overload-induced heart failure. With a cardiac-specific transgenic (TG) mouse model, we showed that a moderate increase of VCP was able to attenuate chronic pressure overload-induced maladaptive cardiac hypertrophy and dysfunction. RNA sequencing and a comprehensive bioinformatic analysis further demonstrated that overexpression of VCP in the heart normalized the pressure overload-stimulated hypertrophic signals and repressed the stress-induced inflammatory response. In addition, VCP overexpression promoted cell survival by enhancing the mitochondria resistance to the oxidative stress via activating the Rictor-mediated-gene networks. VCP was also found to be involved in the regulation of the alternative splicing and differential isoform expression for some genes that are related to ATP production and protein synthesis by interacting with long no-coding RNAs and histone deacetylases, indicating a novel epigenetic regulation of VCP in integrating coding and noncoding genomic network in the stressed heart. In summary, our study demonstrated that the rescuing of a deficient VCP in the heart could prevent pressure overload-induced heart failure by rectifying cardiac hypertrophic and inflammatory signaling and enhancing the cardiac resistance to oxidative stress, which brought in novel insights into the understanding of the mechanism of VCP in protecting patients from hypertensive heart failure. Deficiency of VCP contributes to the pathogenesis of hypertensive heart failure. Rescue of VCP prevents stress-induced cardiac remodeling and cell death. VCP attenuates stress-induced inflammatory and hypertrophic signaling. VCP promotes cardiac resistance to oxidative stress. VCP mediates a novel epigenetic integrating regulation in the stressed heart.
Collapse
Affiliation(s)
- Ning Zhou
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA; Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xin Chen
- Center for Genomics & Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jing Xi
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Ben Ma
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA; Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA, 30303, USA
| | - Christiana Leimena
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Shaunrick Stoll
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Charles Wang
- Center for Genomics & Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA.
| | - Hongyu Qiu
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA; Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
27
|
Riehle C, Bauersachs J. Small animal models of heart failure. Cardiovasc Res 2020; 115:1838-1849. [PMID: 31243437 PMCID: PMC6803815 DOI: 10.1093/cvr/cvz161] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/30/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022] Open
Abstract
Heart disease is a major cause of death worldwide with increasing prevalence, which urges the development of new therapeutic strategies. Over the last few decades, numerous small animal models have been generated to mimic various pathomechanisms contributing to heart failure (HF). Despite some limitations, these animal models have greatly advanced our understanding of the pathogenesis of the different aetiologies of HF and paved the way to understanding the underlying mechanisms and development of successful treatments. These models utilize surgical techniques, genetic modifications, and pharmacological approaches. The present review discusses the strengths and limitations of commonly used small animal HF models, which continue to provide crucial insight and facilitate the development of new treatment strategies for patients with HF.
Collapse
Affiliation(s)
- Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany
| |
Collapse
|
28
|
Yang L, Wang L, Deng Y, Sun L, Lou B, Yuan Z, Wu Y, Zhou B, Liu J, She J. Serum lipids profiling perturbances in patients with ischemic heart disease and ischemic cardiomyopathy. Lipids Health Dis 2020; 19:89. [PMID: 32386519 PMCID: PMC7210665 DOI: 10.1186/s12944-020-01269-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 04/27/2020] [Indexed: 01/07/2023] Open
Abstract
Background Ischemic heart disease (IHD) is a common cardiovascular disorder associated with inadequate blood supply to the myocardium. Chronic coronary ischemia leads to ischemic cardiomyopathy (ICM). Despite their rising prevalence and morbidity, few studies have discussed the lipids alterations in these patients. Methods In this cross-sectional study, we analyzed serum lipids profile in IHD and ICM patients using a lipidomics approach. Consecutive consenting patients admitted to the hospital for IHD and ICM were enrolled. Serum samples were obtained after overnight fasting. Non-targeted metabolomics was applied to demonstrate lipids metabolic profile in control, IHD and ICM patients. Results A total of 63 and 62 lipids were detected in negative and positive ion mode respectively. Among them, 16:0 Lyso PI, 18:1 Lyso PI in negative ion mode, and 19:0 Lyso PC, 12:0 SM d18:1/12:0, 15:0 Lyso PC, 17:0 PC, 18:1–18:0 PC in positive ion mode were significantly altered both in IHD and ICM as compared to control. 13:0 Lyso PI, 18:0 Lyso PI, 16:0 PE, 14:0 PC DMPC, 16:0 ceramide, 18:0 ceramide in negative ion mode, and 17:0 PE, 19:0 PC, 14:0 Lyso PC, 20:0 Lyso PC, 18:0 PC DSPC, 18:0–22:6 PC in positive ion mode were significantly altered only in ICM as compared to IHD and control. Conclusion Using non-targeted lipidomics profiling, we have successfully identified a group of circulating lipids that were significantly altered in IHD and ICM. The lipids metabolic signatures shed light on potential new biomarkers and therapeutics for preventing and treating ICM.
Collapse
Affiliation(s)
- Lin Yang
- Vascular surgery Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Wang
- Department of cardiovascular surgery, The general hospital of Ningxia Medical Univetsity, Yinchuan, China
| | - Yangyang Deng
- Cardiovascular Department, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710048, People's Republic of China
| | - Lizhe Sun
- Cardiovascular Department, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710048, People's Republic of China
| | - Bowen Lou
- Cardiovascular Department, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710048, People's Republic of China
| | - Zuyi Yuan
- Cardiovascular Department, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710048, People's Republic of China
| | - Yue Wu
- Cardiovascular Department, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710048, People's Republic of China
| | - Bo Zhou
- Respiratory Department, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710048, People's Republic of China.
| | - Junhui Liu
- Diagnostic Department, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710048, People's Republic of China.
| | - Jianqing She
- Cardiovascular Department, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710048, People's Republic of China.
| |
Collapse
|
29
|
Heyne E, Schrepper A, Doenst T, Schenkl C, Kreuzer K, Schwarzer M. High-fat diet affects skeletal muscle mitochondria comparable to pressure overload-induced heart failure. J Cell Mol Med 2020; 24:6741-6749. [PMID: 32363733 PMCID: PMC7299710 DOI: 10.1111/jcmm.15325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023] Open
Abstract
In heart failure, high-fat diet (HFD) may exert beneficial effects on cardiac mitochondria and contractility. Skeletal muscle mitochondrial dysfunction in heart failure is associated with myopathy. However, it is not clear if HFD affects skeletal muscle mitochondria in heart failure as well. To induce heart failure, we used pressure overload (PO) in rats fed normal chow or HFD. Interfibrillar mitochondria (IFM) and subsarcolemmal mitochondria (SSM) from gastrocnemius were isolated and functionally characterized. With PO heart failure, maximal respiratory capacity was impaired in IFM but increased in SSM of gastrocnemius. Unexpectedly, HFD affected mitochondria comparably to PO. In combination, PO and HFD showed additive effects on mitochondrial subpopulations which were reflected by isolated complex activities. While PO impaired diastolic as well as systolic cardiac function and increased glucose tolerance, HFD did not affect cardiac function but decreased glucose tolerance. We conclude that HFD and PO heart failure have comparable effects leading to more severe impairment of IFM. Glucose tolerance seems not causally related to skeletal muscle mitochondrial dysfunction. The additive effects of HFD and PO may suggest accelerated skeletal muscle mitochondrial dysfunction when heart failure is accompanied with a diet containing high fat.
Collapse
Affiliation(s)
- Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Katrin Kreuzer
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| |
Collapse
|
30
|
Miranda‐Silva D, Wüst RCI, Conceição G, Gonçalves‐Rodrigues P, Gonçalves N, Gonçalves A, Kuster DWD, Leite‐Moreira AF, Velden J, Sousa Beleza JM, Magalhães J, Stienen GJM, Falcão‐Pires I. Disturbed cardiac mitochondrial and cytosolic calcium handling in a metabolic risk-related rat model of heart failure with preserved ejection fraction. Acta Physiol (Oxf) 2020; 228:e13378. [PMID: 31520455 PMCID: PMC7064935 DOI: 10.1111/apha.13378] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022]
Abstract
AIM Calcium ions play a pivotal role in matching energy supply and demand in cardiac muscle. Mitochondrial calcium concentration is lower in animal models of heart failure with reduced ejection fraction (HFrEF), but limited information is available about mitochondrial calcium handling in heart failure with preserved ejection fraction (HFpEF). METHODS We assessed mitochondrial Ca2+ handling in intact cardiomyocytes from Zucker/fatty Spontaneously hypertensive F1 hybrid (ZSF1)-lean (control) and ZSF1-obese rats, a metabolic risk-related model of HFpEF. A mitochondrially targeted Ca2+ indicator (MitoCam) was expressed in cultured adult rat cardiomyocytes. Cytosolic and mitochondrial Ca2+ transients were measured at different stimulation frequencies. Mitochondrial respiration and swelling, and expression of key proteins were determined ex vivo. RESULTS At rest, mitochondrial Ca2+ concentration in ZSF1-obese was larger than in ZSF1-lean. The diastolic and systolic mitochondrial Ca2+ concentrations increased with stimulation frequency, but the steady-state levels were larger in ZSF1-obese. The half-widths of the contractile responses, the resting cytosolic Ca2+ concentration and the decay half-times of the cytosolic Ca2+ transients were higher in ZSF1-obese, likely because of a lower SERCA2a/phospholamban ratio. Mitochondrial respiration was lower, particularly with nicotinamide adenine dinucleotide (NADH) (complex I) substrates, and mitochondrial swelling was larger in ZSF1-obese. CONCLUSION The free mitochondrial calcium concentration is higher in HFpEF owing to alterations in mitochondrial and cytosolic Ca2+ handling. This coupling between cytosolic and mitochondrial Ca2+ levels may compensate for myocardial ATP supply in vivo under conditions of mild mitochondrial dysfunction. However, if mitochondrial Ca2+ concentration is sustainedly increased, it might trigger mitochondrial permeability transition pore opening.
Collapse
Affiliation(s)
- Daniela Miranda‐Silva
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Rob C. I. Wüst
- Department of Physiology Amsterdam UMC VUmc Amsterdam Cardiovascular Sciences Amsterdam the Netherlands
- Department of Human Movement Sciences Laboratory for Myology Faculty of Behavioural and Movement Sciences Amsterdam Movement Sciences Vrije Universiteit Amsterdam Amsterdam the Netherlands
| | - Glória Conceição
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Patrícia Gonçalves‐Rodrigues
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Nádia Gonçalves
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Alexandre Gonçalves
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Diederik W. D. Kuster
- Department of Physiology Amsterdam UMC VUmc Amsterdam Cardiovascular Sciences Amsterdam the Netherlands
| | - Adelino F. Leite‐Moreira
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Jolanda Velden
- Department of Physiology Amsterdam UMC VUmc Amsterdam Cardiovascular Sciences Amsterdam the Netherlands
- Netherlands Heart Institute Utrecht the Netherlands
| | - Jorge M. Sousa Beleza
- LaMetEx—Laboratory of Metabolism and Exercise Faculty of Sport Cardiovascular Research Center - UniC, University of Porto Porto Portugal
| | - José Magalhães
- LaMetEx—Laboratory of Metabolism and Exercise Faculty of Sport Cardiovascular Research Center - UniC, University of Porto Porto Portugal
| | - Ger J. M. Stienen
- Department of Physiology Amsterdam UMC VUmc Amsterdam Cardiovascular Sciences Amsterdam the Netherlands
| | - Inês Falcão‐Pires
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| |
Collapse
|
31
|
Adams V, Bowen TS, Werner S, Barthel P, Amberger C, Konzer A, Graumann J, Sehr P, Lewis J, Provaznik J, Benes V, Büttner P, Gasch A, Mangner N, Witt CC, Labeit D, Linke A, Labeit S. Small-molecule-mediated chemical knock-down of MuRF1/MuRF2 and attenuation of diaphragm dysfunction in chronic heart failure. J Cachexia Sarcopenia Muscle 2019; 10:1102-1115. [PMID: 31140761 PMCID: PMC6818456 DOI: 10.1002/jcsm.12448] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Chronic heart failure (CHF) leads to diaphragm myopathy that significantly impairs quality of life and worsens prognosis. In this study, we aimed to assess the efficacy of a recently discovered small-molecule inhibitor of MuRF1 in treating CHF-induced diaphragm myopathy and loss of contractile function. METHODS Myocardial infarction was induced in mice by ligation of the left anterior descending coronary artery. Sham-operated animals (sham) served as controls. One week post-left anterior descending coronary artery ligation animals were randomized into two groups-one group was fed control rodent chow, whereas the other group was fed a diet containing 0.1% of the compound ID#704946-a recently described MuRF1-interfering small molecule. Echocardiography confirmed development of CHF after 10 weeks. Functional and molecular analysis of the diaphragm was subsequently performed. RESULTS Chronic heart failure induced diaphragm fibre atrophy and contractile dysfunction by ~20%, as well as decreased activity of enzymes involved in mitochondrial energy production (P < 0.05). Treatment with compound ID#704946 in CHF mice had beneficial effects on the diaphragm: contractile function was protected, while mitochondrial enzyme activity and up-regulation of the MuRF1 and MuRF2 was attenuated after infarct. CONCLUSIONS Our murine CHF model presented with diaphragm fibre atrophy, impaired contractile function, and reduced mitochondrial enzyme activities. Compound ID#704946 rescued from this partially, possibly by targeting MuRF1/MuRF2. However, at this stage of our study, we refrain to claim specific mechanism(s) and targets of compound ID#704946, because the nature of changes after 12 weeks of feeding is likely to be complex and is not necessarily caused by direct mechanistic effects.
Collapse
Affiliation(s)
- Volker Adams
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - T Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Sarah Werner
- University Clinic of Cardiology, Heart Center Leipzig, Leipzig, Germany
| | - Peggy Barthel
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | | | - Anne Konzer
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Rhine-Main, Germany
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Rhine-Main, Germany
| | - Peter Sehr
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Joe Lewis
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jan Provaznik
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vladimir Benes
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Petra Büttner
- University Clinic of Cardiology, Heart Center Leipzig, Leipzig, Germany
| | - Alexander Gasch
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Norman Mangner
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - Christian C Witt
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Dittmar Labeit
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.,Myomedix GmbH, Neckargemünd, Germany
| | - Axel Linke
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.,Myomedix GmbH, Neckargemünd, Germany
| |
Collapse
|
32
|
Chadda KR, Fazmin IT, Ahmad S, Valli H, Edling CE, Huang CLH, Jeevaratnam K. Arrhythmogenic mechanisms of obstructive sleep apnea in heart failure patients. Sleep 2019; 41:5054592. [PMID: 30016501 DOI: 10.1093/sleep/zsy136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 07/13/2018] [Indexed: 01/01/2023] Open
Abstract
Heart failure (HF) affects 23 million people worldwide and results in 300000 annual deaths. It is associated with many comorbidities, such as obstructive sleep apnea (OSA), and risk factors for both conditions overlap. Eleven percent of HF patients have OSA and 7.7% of OSA patients have left ventricular ejection fraction <50% with arrhythmias being a significant comorbidity in HF and OSA patients. Forty percent of HF patients develop atrial fibrillation (AF) and 30%-50% of deaths from cardiac causes in HF patients are from sudden cardiac death. OSA is prevalent in 32%-49% of patients with AF and there is a dose-dependent relationship between OSA severity and resistance to anti-arrhythmic therapies. HF and OSA lead to various downstream arrhythmogenic mechanisms, including metabolic derangement, remodeling, inflammation, and autonomic imbalance. (1) Metabolic derangement and production of reactive oxidative species increase late Na+ currents, decrease outward K+ currents and downregulate connexin-43 and cell-cell coupling. (2) remodeling also features downregulated K+ currents in addition to decreased Na+/K+ ATPase currents, altered Ca2+ homeostasis, and increased density of If current. (3) Chronic inflammation leads to downregulation of both Nav1.5 channels and K+ channels, altered Ca2+ homeostasis and reduced cellular coupling from alterations of connexin expression. (4) Autonomic imbalance causes arrhythmias by evoking triggered activity through increased Ca2+ transients and reduction of excitation wavefront wavelength. Thus, consideration of these multiple pathophysiological pathways (1-4) will enable the development of novel therapeutic strategies that can be targeted against arrhythmias in the context of complex disease, such as the comorbidities of HF and OSA.
Collapse
Affiliation(s)
- Karan R Chadda
- Faculty of Health and Medical Science, University of Surrey, Guildford, United Kingdom.,Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - Ibrahim T Fazmin
- Faculty of Health and Medical Science, University of Surrey, Guildford, United Kingdom.,Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - Haseeb Valli
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - Charlotte E Edling
- Faculty of Health and Medical Science, University of Surrey, Guildford, United Kingdom
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, United Kingdom.,Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Science, University of Surrey, Guildford, United Kingdom.,Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, United Kingdom
| |
Collapse
|
33
|
Abstract
In heart failure, alterations of Na+ and Ca2+ handling, energetic deficit, and oxidative stress in cardiac myocytes are important pathophysiological hallmarks. Mitochondria are central to these processes because they are the main source for ATP, but also reactive oxygen species (ROS), and their function is critically controlled by Ca2+ During physiological variations of workload, mitochondrial Ca2+ uptake is required to match energy supply to demand but also to keep the antioxidative capacity in a reduced state to prevent excessive emission of ROS. Mitochondria take up Ca2+ via the mitochondrial Ca2+ uniporter, which exists in a multiprotein complex whose molecular components were identified only recently. In heart failure, deterioration of cytosolic Ca2+ and Na+ handling hampers mitochondrial Ca2+ uptake and the ensuing Krebs cycle-induced regeneration of the reduced forms of NADH (nicotinamide adenine dinucleotide) and NADPH (nicotinamide adenine dinucleotide phosphate), giving rise to energetic deficit and oxidative stress. ROS emission from mitochondria can trigger further ROS release from neighboring mitochondria termed ROS-induced ROS release, and cross talk between different ROS sources provides a spatially confined cellular network of redox signaling. Although low levels of ROS may serve physiological roles, higher levels interfere with excitation-contraction coupling, induce maladaptive cardiac remodeling through redox-sensitive kinases, and cell death through mitochondrial permeability transition. Targeting the dysregulated interplay between excitation-contraction coupling and mitochondrial energetics may ameliorate the progression of heart failure.
Collapse
Affiliation(s)
- Edoardo Bertero
- From the Comprehensive Heart Failure Center, University Clinic Würzburg, Germany
| | - Christoph Maack
- From the Comprehensive Heart Failure Center, University Clinic Würzburg, Germany.
| |
Collapse
|
34
|
Xu L, Su Y, Zhao Y, Sheng X, Tong R, Ying X, Gao L, Ji Q, Gao Y, Yan Y, Yuan A, Wu F, Lan F, Pu J. Melatonin differentially regulates pathological and physiological cardiac hypertrophy: Crucial role of circadian nuclear receptor RORα signaling. J Pineal Res 2019; 67:e12579. [PMID: 30958896 DOI: 10.1111/jpi.12579] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/26/2019] [Accepted: 04/02/2019] [Indexed: 12/30/2022]
Abstract
Exercise-induced physiological hypertrophy provides protection against cardiovascular disease, whereas disease-induced pathological hypertrophy leads to heart failure. Emerging evidence suggests pleiotropic roles of melatonin in cardiac disease; however, the effects of melatonin on physiological vs pathological cardiac hypertrophy remain unknown. Using swimming-induced physiological hypertrophy and pressure overload-induced pathological hypertrophy models, we found that melatonin treatment significantly improved pathological hypertrophic responses accompanied by alleviated oxidative stress in myocardium but did not affect physiological cardiac hypertrophy and oxidative stress levels. As an important mediator of melatonin, the retinoid-related orphan nuclear receptor-α (RORα) was significantly decreased in human and murine pathological hypertrophic cardiomyocytes, but not in swimming-induced physiological hypertrophic murine hearts. In vivo and in vitro loss-of-function experiments indicated that RORα deficiency significantly aggravated pathological cardiac hypertrophy, and notably weakened the anti-hypertrophic effects of melatonin. Mechanistically, RORα mediated the cardioprotection of melatonin in pathological hypertrophy mainly by transactivation of manganese-dependent superoxide dismutase (MnSOD) via binding to the RORα response element located in the promoter region of the MnSOD gene. Furthermore, MnSOD overexpression reversed the pro-hypertrophic effects of RORα deficiency, while MnSOD silencing abolished the anti-hypertrophic effects of RORα overexpression in pathological cardiac hypertrophy. Collectively, our findings provide the first evidence that melatonin exerts an anti-hypertrophic effect on pathological but not physiological cardiac hypertrophy via alleviating oxidative stress through transactivation of the antioxidant enzyme MnSOD in a RORα-dependent manner.
Collapse
Affiliation(s)
- Longwei Xu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yuanyuan Su
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yichao Zhao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xincheng Sheng
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Renyang Tong
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoying Ying
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lingchen Gao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Qingqi Ji
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Gao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Yan
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ancai Yuan
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Fujian Wu
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
35
|
Groepenhoff F, Bots SH, Kessler EL, Sickinghe AA, Eikendal ALM, Leiner T, den Ruijter HM. Sex-Specific Aspects in the Pathophysiology and Imaging of Coronary Macro- and Microvascular Disease. J Cardiovasc Transl Res 2019; 13:39-46. [PMID: 31471830 PMCID: PMC7010630 DOI: 10.1007/s12265-019-09906-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
Sex differences in coronary artery disease (CAD) are well established, with women presenting with non-obstructive CAD more often than men do. However, recent evidence has identified coronary microvascular dysfunction as the underlying cause for cardiac complaints, yet sex-specific prevalence numbers are inconclusive. This review summarises known sex-specific aspects in the pathophysiology of both macro- and microvascular dysfunction and identifies currently existing knowledge gaps. In addition, this review describes current diagnostic approaches and whether these should take underlying sex differences into account by, for example, using different techniques or cut-off values for women and men. Future research into both innovation of imaging techniques and perfusion-related sex differences is needed to fill evidence gaps and enable the implementation of the available knowledge in daily clinical practice.
Collapse
Affiliation(s)
- Floor Groepenhoff
- Laboratory for Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sophie H Bots
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elise L Kessler
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ariane A Sickinghe
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anouk L M Eikendal
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tim Leiner
- Department of Radiology, University Medical Center Utrecht, Utrecht University, P. O. Box 85500, 3508, GA, Utrecht, The Netherlands
| | - Hester M den Ruijter
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Gao JL, Zhao J, Zhu HB, Peng X, Zhu JX, Ma MH, Fu Y, Hu N, Tai Y, Xuan XC, Dong DL. Characterizations of mitochondrial uncoupling induced by chemical mitochondrial uncouplers in cardiomyocytes. Free Radic Biol Med 2018; 124:288-298. [PMID: 29935261 DOI: 10.1016/j.freeradbiomed.2018.06.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 01/05/2023]
Abstract
Induction of mild mitochondrial uncoupling is protective in a variety of disorders; however, it is unclear how to recognize the mild mitochondrial uncoupling induced by chemical mitochondrial uncouplers. The aim of the present study is to identify the pharmacological properties of mitochondrial uncoupling induced by mitochondrial uncouplers in cardiomyocytes. Neonatal rat cardiomyocytes were cultured. Protein levels were measured by using western blot technique. The whole cell respiratory function was determined by using high-resolution respirometry. The typical types of chemical mitochondrial uncouplers, carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP), niclosamide, and BAM15, induced biphasic change of STAT3 activity in cardiomyocytes, activating STAT3 at low dose and inhibiting STAT3 at high dose, though the dose range of these drugs was distinct. Low-dose uncouplers induced STAT3 activation through the mild increase of mitochondrial ROS (mitoROS) generation and the subsequent JAK/STAT3 activation in cardiomyocytes. However, high-dose uncouplers induced inhibition of STAT3, decrease of ATP production, and cardiomyocyte death. High-dose uncouplers induced STAT3 inhibition through the excessive mitoROS generation and the decreased ATP -induced AMPK activation. Low-dose mitochondrial uncouplers attenuated doxorubicin (DOX)-induced STAT3 inhibition and cardiomyocyte death, and activated STAT3 contributed to the cardioprotection of low-dose mitochondrial uncouplers. Uncoupler-induced mild mitochondrial uncoupling in cardiomyocytes is characterized by STAT3 activation and ATP increase whereas excessive mitochondrial uncoupling is characterized by STAT3 inhibition, ATP decrease and cell injury. Development of mitochondrial uncoupler with optimal dose window of inducing mild uncoupling is a promising strategy for heart protection.
Collapse
Affiliation(s)
- Jin-Lai Gao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Jing Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Hai-Bin Zhu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Xuan Peng
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Jun-Xue Zhu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Ming-Hui Ma
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Yao Fu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Nan Hu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Yu Tai
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - Xiu-Chen Xuan
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China
| | - De-Li Dong
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Baojian Road 157, Harbin 150086, Heilongjiang Province, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, PR China.
| |
Collapse
|
37
|
Smith RL, Soeters MR, Wüst RCI, Houtkooper RH. Metabolic Flexibility as an Adaptation to Energy Resources and Requirements in Health and Disease. Endocr Rev 2018; 39:489-517. [PMID: 29697773 PMCID: PMC6093334 DOI: 10.1210/er.2017-00211] [Citation(s) in RCA: 351] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 04/19/2018] [Indexed: 12/15/2022]
Abstract
The ability to efficiently adapt metabolism by substrate sensing, trafficking, storage, and utilization, dependent on availability and requirement, is known as metabolic flexibility. In this review, we discuss the breadth and depth of metabolic flexibility and its impact on health and disease. Metabolic flexibility is essential to maintain energy homeostasis in times of either caloric excess or caloric restriction, and in times of either low or high energy demand, such as during exercise. The liver, adipose tissue, and muscle govern systemic metabolic flexibility and manage nutrient sensing, uptake, transport, storage, and expenditure by communication via endocrine cues. At a molecular level, metabolic flexibility relies on the configuration of metabolic pathways, which are regulated by key metabolic enzymes and transcription factors, many of which interact closely with the mitochondria. Disrupted metabolic flexibility, or metabolic inflexibility, however, is associated with many pathological conditions including metabolic syndrome, type 2 diabetes mellitus, and cancer. Multiple factors such as dietary composition and feeding frequency, exercise training, and use of pharmacological compounds, influence metabolic flexibility and will be discussed here. Last, we outline important advances in metabolic flexibility research and discuss medical horizons and translational aspects.
Collapse
Affiliation(s)
- Reuben L Smith
- Laboratory of Genetic Metabolic Diseases, Academic Medical Center, AZ Amsterdam, Netherlands.,Amsterdam Gastroenterology and Metabolism, Academic Medical Center, AZ Amsterdam, Netherlands
| | - Maarten R Soeters
- Amsterdam Gastroenterology and Metabolism, Academic Medical Center, AZ Amsterdam, Netherlands.,Department of Endocrinology and Metabolism, Internal Medicine, Academic Medical Center, AZ Amsterdam, Netherlands
| | - Rob C I Wüst
- Laboratory of Genetic Metabolic Diseases, Academic Medical Center, AZ Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Academic Medical Center, AZ Amsterdam, Netherlands.,Amsterdam Movement Sciences, Academic Medical Center, AZ Amsterdam, Netherlands
| | - Riekelt H Houtkooper
- Laboratory of Genetic Metabolic Diseases, Academic Medical Center, AZ Amsterdam, Netherlands.,Amsterdam Gastroenterology and Metabolism, Academic Medical Center, AZ Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Academic Medical Center, AZ Amsterdam, Netherlands
| |
Collapse
|
38
|
Urmaliya V, Franchelli G. A multidimensional sight on cardiac failure: uncovered from structural to molecular level. Heart Fail Rev 2018; 22:357-370. [PMID: 28474325 DOI: 10.1007/s10741-017-9610-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Heart failure is one of the leading causes of death, with high mortality rate within 5 years after diagnosis. Treatment and prognosis options for heart failure primarily targeted on hemodynamic and neurohumoral components that drive progressive deterioration of the heart. However, given the multifactorial background that eventually leads to the "phenotype" named heart failure, better insight into the various components may lead to personalized treatment opportunities. Indeed, currently used criteria to diagnose and/or classify heart failure are possibly too focused on phenotypic improvement rather than the molecular driver of the disease and could therefore be further refined by integrating the leap of molecular and cellular knowledge. The ambiguity of the ejection fraction-based classification criteria became evident with development of advanced molecular techniques and the dawn of omics disciplines which introduced the idea that disease is caused by a myriad of cellular and molecular processes rather than a single event or pathway. The fact that different signaling pathways may underlie similar clinical manifestations calls for a more holistic study of heart failure. In this context, the systems biology approach can offer a better understanding of how different components of a system are altered during disease and how they interact with each other, potentially leading to improved diagnosis and classification of this condition. This review is aimed at addressing heart failure through a multilayer approach that covers individually some of the anatomical, morphological, functional, and tissue aspects, with focus on cellular and subcellular features as an alternative insight into new therapeutic opportunities.
Collapse
Affiliation(s)
- Vijay Urmaliya
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium.
| | | |
Collapse
|
39
|
LIU JJ, LU Y, PING NN, LI X, LIN YX, LI CF. Apocynin Ameliorates Pressure Overload-Induced Cardiac Remodeling by Inhibiting Oxidative Stress and Apoptosis. Physiol Res 2017; 66:741-752. [DOI: 10.33549/physiolres.933257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress plays an important role in pressure overload-induced cardiac remodeling. The purpose of this study was to determine whether apocynin, a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, attenuates pressure overload-induced cardiac remodeling in rats. After abdominal aorta constriction, the surviving rats were randomly divided into four groups: sham group, abdominal aorta constriction group, apocynin group, captopril group. Left ventricular pathological changes were studied using Masson’s trichrome staining. Metalloproteinase-2 (MMP-2) levels in the left ventricle were analyzed by western blot and gelatin zymography. Oxidative stress and apoptotic index were also examined in cardiomyocytes using dihydroethidium and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), respectively. Our results showed that abdominal aorta constriction significantly caused excess collagen deposition and cardiac insult. Treatment with apocynin significantly inhibited deposition of collagen and reduced the level of MMP-2. Furthermore, apocynin also decreased the NADPH oxidase activity, reactive oxygen species production and cardiomyocyte apoptotic index. Interestingly, apocynin only inhibited NADPH oxidase activity without affecting its expression or the level of angiotension II in the left ventricle. In conclusion, apocynin reduced collagen deposition, oxidative stress, and inhibited apoptosis, ultimately ameliorating cardiac remodeling by mechanisms that are independent of the renin-angiotensin system.
Collapse
Affiliation(s)
| | | | | | | | | | - C.-F. LI
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, China
| |
Collapse
|
40
|
Duicu OM, Privistirescu A, Wolf A, Petruş A, Dănilă MD, Raţiu CD, Muntean DM, Sturza A. Methylene blue improves mitochondrial respiration and decreases oxidative stress in a substrate-dependent manner in diabetic rat hearts. Can J Physiol Pharmacol 2017; 95:1376-1382. [PMID: 28738167 DOI: 10.1139/cjpp-2017-0074] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Diabetic cardiomyopathy has been systematically associated with compromised mitochondrial energetics and increased generation of reactive oxygen species (ROS) that underlie its progression to heart failure. Methylene blue is a redox drug with reported protective effects mainly on brain mitochondria. The purpose of the present study was to characterize the effects of acute administration of methylene blue on mitochondrial respiration, H2O2 production, and calcium sensitivity in rat heart mitochondria isolated from healthy and 2 months (streptozotocin-induced) diabetic rats. Mitochondrial respiratory function was assessed by high-resolution respirometry. H2O2 production and calcium retention capacity were measured spectrofluorimetrically. The addition of methylene blue (0.1 μmol·L-1) elicited an increase in oxygen consumption of mitochondria energized with complex I and II substrates in both normal and diseased mitochondria. Interestingly, methylene blue elicited a significant increase in H2O2 release in the presence of complex I substrates (glutamate and malate), but had an opposite effect in mitochondria energized with complex II substrate (succinate). No changes in the calcium retention capacity of healthy or diabetic mitochondria were found in the presence of methylene blue. In conclusion, in cardiac mitochondria isolated from diabetic and nondiabetic rat hearts, methylene blue improved respiratory function and elicited a dichotomic, substrate-dependent effect on ROS production.
Collapse
Affiliation(s)
- Oana M Duicu
- a Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., Timişoara 300041, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Andreea Privistirescu
- a Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., Timişoara 300041, Romania
| | - Adrian Wolf
- a Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., Timişoara 300041, Romania
| | - Alexandra Petruş
- a Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., Timişoara 300041, Romania
| | - Maria D Dănilă
- a Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., Timişoara 300041, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Corina D Raţiu
- a Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., Timişoara 300041, Romania
| | - Danina M Muntean
- a Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., Timişoara 300041, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Adrian Sturza
- a Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, 2, Eftimie Murgu Sq., Timişoara 300041, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| |
Collapse
|
41
|
Monoamine Oxidases, Oxidative Stress, and Altered Mitochondrial Dynamics in Cardiac Ageing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3017947. [PMID: 28546851 PMCID: PMC5435992 DOI: 10.1155/2017/3017947] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/22/2017] [Accepted: 03/22/2017] [Indexed: 01/06/2023]
Abstract
The advances in healthcare over the past several decades have resulted in populations now living longer. With this increase in longevity, a wider prevalence of cardiovascular diseases is more common and known to be a major factor in rising healthcare costs. A wealth of scientific evidence has implicated cell senescence as an important component in the etiology of these age-dependent pathologies. A number of studies indicate that an excess of reactive oxygen species (ROS) contributes to trigger and accelerate the cardiac senescence processes, and a new role of monoamine oxidases, MAO-A and MAO-B, is emerging in this context. These mitochondrial enzymes regulate the level of catecholamines and serotonin by catalyzing their oxidative deamination in the heart. MAOs' expression substantially increases with ageing (6-fold MAO-A in the heart and 4-fold MAO-B in neuronal tissue), and their involvement in cardiac diseases is supposedly related to the formation of ROS, via the hydrogen peroxide produced during the substrate degradation. Here, we will review the most recent advances in this field and describe why MAOs could be effective targets in order to prevent age-associated cardiovascular disease.
Collapse
|
42
|
Ashok A, Kanwar JR, Krishnan UM, Kanwar RK. SurR9C84A protects and recovers human cardiomyocytes from hypoxia induced apoptosis. Exp Cell Res 2016; 350:19-31. [PMID: 27816606 DOI: 10.1016/j.yexcr.2016.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 10/19/2016] [Accepted: 10/22/2016] [Indexed: 02/05/2023]
Abstract
Survivin, as an anti-apoptotic protein and a cell cycle regulator, is recently gaining importance for its regenerative potential in salvaging injured hypoxic cells of vital organs such as heart. Different strategies are being employed to upregulate survivin expression in dying hypoxic cardiomyocytes. We investigated the cardioprotective potential of a cell permeable survivin mutant protein SurR9C84A, for the management of hypoxia mediated cardiomyocyte apoptosis, in a novel and clinically relevant model employing primary human cardiomyocytes (HCM). The aim of this research work was to study the efficacy and mechanism of SurR9C84A facilitated cardioprotection and regeneration in hypoxic HCM. To mimic hypoxic microenvironment in vitro, well characterized HCM were treated with 100µm (48h) cobalt chloride to induce hypoxia. Hypoxia induced (HI) HCM were further treated with SurR9C84A (1µg/mL) in order to analyse its cardioprotective efficacy. Confocal microscopy showed rapid internalization of SurR9C84A and scanning electron microscopy revealed the reinstatement of cytoskeleton projections in HI HCM. SurR9C84A treatment increased cell viability, reduced cell death via, apoptosis (Annexin-V assay), and downregulated free cardiac troponin T and MMP-9 expression. SurR9C84A also upregulated the expression of proliferation markers (PCNA and Ki-67) and downregulated mitochondrial depolarization and ROS levels thereby, impeding cell death. Human Apoptosis Array further revealed that SurR9C84A downregulated expression of pro-apoptotic markers and augmented expression of HSPs and HTRA2/Omi. SurR9C84A treatment led to enhanced levels of survivin, VEGF, PI3K and pAkt. SurR9C84A proved non-toxic to normoxic HCM, as validated through unaltered cell proliferation and other marker levels. Its pre-treatment exhibited lesser susceptibility to hypoxia/damage. SurR9C84A holds a promising clinical potential for human cardiomyocyte survival and proliferation following hypoxic injury.
Collapse
Affiliation(s)
- Ajay Ashok
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Faculty of Health, Centre for Molecular and Medical Research (C-MMR), Deakin University, Waurn Ponds, Victoria 3216, Australia; Department of Pathology, Case Western Reserve University, 2103 Cornell Rd. WRB 5128, Cleveland, OH 44106-7288, USA
| | - Jagat Rakesh Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Faculty of Health, Centre for Molecular and Medical Research (C-MMR), Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical & Biotechnology (SCBT), SASTRA University, Thanjavur 613401, India
| | - Rupinder Kaur Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Faculty of Health, Centre for Molecular and Medical Research (C-MMR), Deakin University, Waurn Ponds, Victoria 3216, Australia.
| |
Collapse
|
43
|
Cell Death and Heart Failure in Obesity: Role of Uncoupling Proteins. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9340654. [PMID: 27642497 PMCID: PMC5011521 DOI: 10.1155/2016/9340654] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022]
Abstract
Metabolic diseases such as obesity, metabolic syndrome, and type II diabetes are often characterized by increased reactive oxygen species (ROS) generation in mitochondrial respiratory complexes, associated with fat accumulation in cardiomyocytes, skeletal muscle, and hepatocytes. Several rodents studies showed that lipid accumulation in cardiac myocytes produces lipotoxicity that causes apoptosis and leads to heart failure, a dynamic pathological process. Meanwhile, several tissues including cardiac tissue develop an adaptive mechanism against oxidative stress and lipotoxicity by overexpressing uncoupling proteins (UCPs), specific mitochondrial membrane proteins. In heart from rodent and human with obesity, UCP2 and UCP3 may protect cardiomyocytes from death and from a state progressing to heart failure by downregulating programmed cell death. UCP activation may affect cytochrome c and proapoptotic protein release from mitochondria by reducing ROS generation and apoptotic cell death. Therefore the aim of this review is to discuss recent findings regarding the role that UCPs play in cardiomyocyte survival by protecting against ROS generation and maintaining bioenergetic metabolism homeostasis to promote heart protection.
Collapse
|
44
|
Liu R, Kenney JW, Manousopoulou A, Johnston HE, Kamei M, Woelk CH, Xie J, Schwarzer M, Garbis SD, Proud CG. Quantitative Non-canonical Amino Acid Tagging (QuaNCAT) Proteomics Identifies Distinct Patterns of Protein Synthesis Rapidly Induced by Hypertrophic Agents in Cardiomyocytes, Revealing New Aspects of Metabolic Remodeling. Mol Cell Proteomics 2016; 15:3170-3189. [PMID: 27512079 PMCID: PMC5054342 DOI: 10.1074/mcp.m115.054312] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Indexed: 01/16/2023] Open
Abstract
Cardiomyocytes undergo growth and remodeling in response to specific pathological or physiological conditions. In the former, myocardial growth is a risk factor for cardiac failure and faster protein synthesis is a major factor driving cardiomyocyte growth. Our goal was to quantify the rapid effects of different pro-hypertrophic stimuli on the synthesis of specific proteins in ARVC and to determine whether such effects are caused by alterations on mRNA abundance or the translation of specific mRNAs. Cardiomyocytes have very low rates of protein synthesis, posing a challenging problem in terms of studying changes in the synthesis of specific proteins, which also applies to other nondividing primary cells. To study the rates of accumulation of specific proteins in these cells, we developed an optimized version of the Quantitative Noncanonical Amino acid Tagging LC/MS proteomic method to label and selectively enrich newly synthesized proteins in these primary cells while eliminating the suppressive effects of pre-existing and highly abundant nonisotope-tagged polypeptides. Our data revealed that a classical pathologic (phenylephrine; PE) and the recently identified insulin stimulus that also contributes to the development of pathological cardiac hypertrophy (insulin), both increased the synthesis of proteins involved in, e.g. glycolysis, the Krebs cycle and beta-oxidation, and sarcomeric components. However, insulin increased synthesis of many metabolic enzymes to a greater extent than PE. Using a novel validation method, we confirmed that synthesis of selected candidates is indeed up-regulated by PE and insulin. Synthesis of all proteins studied was up-regulated by signaling through mammalian target of rapamycin complex 1 without changes in their mRNA levels, showing the key importance of translational control in the rapid effects of hypertrophic stimuli. Expression of PKM2 was up-regulated in rat hearts following TAC. This isoform possesses specific regulatory properties, so this finding indicates it may be involved in metabolic remodeling and also serve as a novel candidate biomarker. Levels of translation factor eEF1 also increased during TAC, likely contributing to faster cell mass accumulation. Interestingly those two candidates were not up-regulated in pregnancy or exercise induced CH, indicating PKM2 and eEF1 were pathological CH specific markers. We anticipate that the methodologies described here will be valuable for other researchers studying protein synthesis in primary cells.
Collapse
Affiliation(s)
- Rui Liu
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Justin W Kenney
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Antigoni Manousopoulou
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Harvey E Johnston
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ‖Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Makoto Kamei
- §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Christopher H Woelk
- ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Jianling Xie
- §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Michael Schwarzer
- **Department of Cardiovascular Surgery, Jena University Hospital-Friedrich Schiller University of Jena, Erlanger Allee 101, 07747 Jena, Germany
| | - Spiros D Garbis
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK; ‖Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK;
| | - Christopher G Proud
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA5005, Australia
| |
Collapse
|
45
|
Wüst RCI, de Vries HJ, Wintjes LT, Rodenburg RJ, Niessen HWM, Stienen GJM. Mitochondrial complex I dysfunction and altered NAD(P)H kinetics in rat myocardium in cardiac right ventricular hypertrophy and failure. Cardiovasc Res 2016; 111:362-72. [PMID: 27402402 DOI: 10.1093/cvr/cvw176] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 06/16/2016] [Indexed: 01/31/2023] Open
Abstract
AIMS In cardiac hypertrophy (CH) and heart failure (HF), alterations occur in mitochondrial enzyme content and activities but the origin and implications of these changes for mitochondrial function need to be resolved. METHODS AND RESULTS Right ventricular CH or HF was induced by monocrotaline injection, which causes pulmonary artery hypertension, in rats. Results were compared with saline injection (CON). NAD(P)H and FAD autofluorescence were recorded in thin intact cardiac trabeculae during transitions in stimulation frequency, to assess mitochondrial complex I and complex II function, respectively. Oxygen consumption, mitochondrial morphology, protein content, and enzymatic activity were assessed. NAD(P)H autofluorescence upon an increase in stimulation frequency showed a rapid decline followed by a slow recovery. FAD autofluorescence followed a similar time course, but in opposite direction. The amplitude of the early rapid change in NAD(P)H autofluorescence was severely depressed in CH and HF compared with CON. The rapid changes in FAD autofluorescence in CH and HF were reduced to a lesser extent. Complex I-coupled respiration showed an ∼3.5-fold reduction in CH and HF; complex II-coupled respiration was depressed two-fold in HF. Western blot analyses revealed modest reductions in complex I protein content in CH and HF and in complex I activity in supercomplexes in HF. Mitochondrial volume density was similar, but mitochondrial remodelling was evident from changes in ultrastructure and fusion/fission indices in CH and HF. CONCLUSION These results suggest that the alterations in mitochondrial function observed in right ventricular CH and HF can be mainly attributed to complex I dysfunction.
Collapse
Affiliation(s)
- Rob C I Wüst
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, O
- 2 Building, De Boelelaan 1118, Amsterdam 1081 HV, The Netherlands
| | - Heder J de Vries
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, O
- 2 Building, De Boelelaan 1118, Amsterdam 1081 HV, The Netherlands
| | - Liesbeth T Wintjes
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Richard J Rodenburg
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Ger J M Stienen
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, O
- 2 Building, De Boelelaan 1118, Amsterdam 1081 HV, The Netherlands Faculty of Science, Department of Physics and Astronomy, VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Park SY, Trinity JD, Gifford JR, Diakos NA, McCreath L, Drakos S, Richardson RS. Mitochondrial function in heart failure: The impact of ischemic and non-ischemic etiology. Int J Cardiol 2016; 220:711-7. [PMID: 27394972 DOI: 10.1016/j.ijcard.2016.06.147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/15/2016] [Accepted: 06/24/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Although cardiac mitochondrial dysfunction is associated with heart failure (HF), this is a complex syndrome with two predominant etiologies, ischemic HF (iHF) and non-ischemic HF (niHF), and the exact impact of mitochondrial dysfunction in these two distinct forms of HF is unknown. METHODS AND RESULTS To determine the impact of HF etiology on mitochondrial function, respiration was measured in permeabilized cardiac muscle fibers from patients with iHF (n=17), niHF (n=18), and healthy donor hearts (HdH). Oxidative phosphorylation capacity (OXPHOS), assessed as state 3 respiration, fell progressively from HdH to niHF, to iHF (Complex I+II: 54±1; 34±4; 27±3pmol·s(-1)·mg(-1)) as did citrate synthase activity (CSA: 206±18; 129±6; 82±6nmol·mg(-1)·min(-1)). Although still significantly lower than HdH, normalization of OXPHOS by CSA negated the difference in mass specific OXPHOS between iHF and niHF. Interestingly, Complex I state 2 respiration increased progressively from HdH, to niHF, to iHF, whether or not normalized for CSA (0.6±0.2; 1.1±0.3; 2.3±0.3; pmol·mg(-1)·CSA), such that the respiratory control ratio (RCR), fell in the same manner across groups. Finally, both the total free radical levels (60±6; 46±4AU) and level of mitochondrial derived superoxide (1.0±0.2; 0.7±0.1AU) were greater in iHF compared to niHF, respectively. CONCLUSIONS Thus, the HF-related attenuation in OXPHOS actually appears to be independent of etiology when the lower mitochondrial content of iHF is taken into account. However, these findings provide evidence of deleterious intrinsic mitochondrial changes in iHF, compared to niHF, including greater proton leak, attenuated OXPHOS efficiency, and augmented free radical levels.
Collapse
Affiliation(s)
- Song-Young Park
- Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, USA; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Joel D Trinity
- Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Jayson R Gifford
- Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Nikolaos A Diakos
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Lauren McCreath
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Stavros Drakos
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Russell S Richardson
- Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, USA; Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
47
|
Zhang L, Ma C, Zhang C, Ma M, Zhang F, Zhang L, Chen Y, Cao F, Li S, Zhu D. Reactive oxygen species effect PASMCs apoptosis via regulation of dynamin-related protein 1 in hypoxic pulmonary hypertension. Histochem Cell Biol 2016; 146:71-84. [DOI: 10.1007/s00418-016-1424-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2016] [Indexed: 01/27/2023]
|
48
|
Maurya CK, Arha D, Rai AK, Kumar SK, Pandey J, Avisetti DR, Kalivendi SV, Klip A, Tamrakar AK. NOD2 activation induces oxidative stress contributing to mitochondrial dysfunction and insulin resistance in skeletal muscle cells. Free Radic Biol Med 2015; 89:158-69. [PMID: 26404168 DOI: 10.1016/j.freeradbiomed.2015.07.154] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/20/2015] [Accepted: 07/24/2015] [Indexed: 01/11/2023]
Abstract
Nucleotide-binding oligomerization domain protein-2 (NOD2) activation in skeletal muscle cells has been associated with insulin resistance, but the underlying mechanisms are not yet clear. Here we demonstrate the implication of oxidative stress in the development of mitochondrial dysfunction and insulin resistance in response to NOD2 activation in skeletal muscle cells. Treatment with the selective NOD2 ligand muramyl dipeptide (MDP) increased mitochondrial reactive oxygen species (ROS) generation in L6 myotubes. MDP-induced ROS production was associated with increased levels of protein carbonyls and reduction in citrate synthase activity, cellular ATP level, and mitochondrial membrane potential, as well as altered expression of genes involved in mitochondrial function and metabolism. Antioxidant treatment attenuated MDP-induced ROS production and restored mitochondrial functions. In addition, the presence of antioxidant prevented NOD2-mediated activation of MAPK kinases and the inflammatory response. This was associated with reduced serine phosphorylation of insulin receptor substrate-1 (IRS-1) and improved insulin-stimulated tyrosine phosphorylation of IRS-1 and downstream activation of Akt phosphorylation. These data indicate that oxidative stress plays a role in NOD2 activation-induced inflammatory response and that MDP-induced oxidative stress correlates with impairment of mitochondrial functions and induction of insulin resistance in skeletal muscle cells.
Collapse
Affiliation(s)
- Chandan K Maurya
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Deepti Arha
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Amit K Rai
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shashi Kant Kumar
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Jyotsana Pandey
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Deepa R Avisetti
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Shasi V Kalivendi
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Akhilesh K Tamrakar
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
49
|
Liu MH, Lin XL, Yuan C, He J, Tan TP, Wu SJ, Yu S, Chen L, Liu J, Tian W, Chen YD, Fu HY, Li J, Zhang Y. Hydrogen sulfide attenuates doxorubicin-induced cardiotoxicity by inhibiting the expression of peroxiredoxin III in H9c2 cells. Mol Med Rep 2015; 13:367-72. [PMID: 26573464 DOI: 10.3892/mmr.2015.4544] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 10/20/2015] [Indexed: 11/06/2022] Open
Abstract
Doxorubicin (DOX) is a widely used chemotherapeutic agent, which can give rise to severe cardiotoxicity, limiting its clinical use. Preliminary evidence suggests that hydrogen sulfide (H2S) may exert protective effects on DOX‑induced cardiotoxicity. Therefore, the aim of the present study was to investigate whether peroxiredoxin III is involved in the cardioprotection of H2S against DOX‑induced cardiotoxicity. The results demonstrated that DOX not only markedly induced injuries, including cytotoxicity and apoptosis, it also increased the expression levels of peroxiredoxin III. Notably, pretreatment with sodium hydrosulfide significantly attenuated the DOX‑induced decrease in cell viability and increase in apoptosis, and also reversed the increased expression levels of peroxiredoxin III in H9c2 cardiomyocytes. In addition, pretreatment of the H9c2 cells with N‑acetyl‑L‑cysteine, a scavenger of reactive oxygen species, prior to exposure to DOX markedly decreased the expression levels of peroxiredoxin III. In conclusion, the results of the present study suggested that exogenous H2S attenuates DOX‑induced cardiotoxicity by inhibiting the expression of peroxiredoxin III in H9c2 cells. In the present study, the apoptosis of H9c2 cardiomyocytes was assessed using an methyl thiazolyl tetrazolium assay and Hoechst staining. The levels of Prx III and cystathionine-γ-lyase were examined by western blotting.
Collapse
Affiliation(s)
- Mi-Hua Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiao-Long Lin
- Department of Pathology, Affiliated Huizhou Hospital of Guangzhou Medical University, The Third People's Hospital of Huizhou, Huizhou, Guangdong 516002, P.R. China
| | - Cong Yuan
- Department of Cardiology, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Jun He
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Tian-Ping Tan
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shao-Jian Wu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shan Yu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Li Chen
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jun Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei Tian
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yu-Dan Chen
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong-Yun Fu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jian Li
- Department of Ultrasonic Diagnosis, Bo'ai Hospital of Zhongshan, Zhongshan, Guangdong 528403, P.R. China
| | - Yuan Zhang
- Department of Pathology, Mawangdui Hospital, Changsha, Hunan 410016, P.R. China
| |
Collapse
|
50
|
Liu MH, Zhang Y, He J, Tan TP, Wu SJ, Fu HY, Chen YD, Liu J, LE QF, Hu HJ, Yuan C, Lin XL. Upregulation of peroxiredoxin III in doxorubicin-induced cytotoxicity and the FoxO3a-dependent expression in H9c2 cardiac cells. Exp Ther Med 2015; 10:1515-1520. [PMID: 26622517 DOI: 10.3892/etm.2015.2693] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 08/03/2015] [Indexed: 12/30/2022] Open
Abstract
Doxorubicin (DOX) is an efficient drug used in cancer therapy; however, it produces reactive oxygen species (ROS) that induce severe cytotoxicity, limiting its clinical application. The aim of the present study was to investigate the role of peroxiredoxin III (Prx III) in DOX-induced H9c2 cell injuries. Following DOX treatment, the expression of phosphorylated-FoxO3a (p-FoxO3a) was decreased and Prx III expression was increased in H9c2 cells. In order to detect whether oxidative stress was involved in the induction of Prx III expression by FoxO3a, exogenous H2O2 was used to induce oxidative stress in the H9c2 cells. Apoptosis of H9c2 cardiomyocytes was assessed using methyl thiazolyl tetrazolium assay and Hoechst staining. The levels of Prx III and p-FoxO3a were evaluated using western blot analysis. As expected, H2O2 was found to mimic the effect of DOX, decreasing the expression of p-FoxO3a and increasing the expression of Prx III. In addition, the study evaluated whether the transcription factor FoxO3a was essential for the expression of Prx III. Pretreatment of H9c2 cells with N-acetyl-L-cysteine (NAC), a scavenger of ROS, prior to exposure to DOX dramatically increased the phosphorylation of FoxO3a and led to a marked reduction in Prx III expression in the H9c2 cells. In conclusion, the results of the current study suggest that FoxO3a mediates the expression of Prx III in DOX-induced injuries.
Collapse
Affiliation(s)
- Mi-Hua Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yuan Zhang
- Department of Pathology, Hunan Mawangdui Hospital, Changsha, Hunan 410016, P.R. China
| | - Jun He
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Tian-Ping Tan
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shao-Jian Wu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong-Yun Fu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yu-Dan Chen
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jun Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Qun-Fang LE
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Heng-Jing Hu
- Department of Cardiology/Cardiac Catheterisation Lab, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Cong Yuan
- Department of Cardiology, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Xiao-Long Lin
- Department of Pathology, The Third People's Hospital of Huizhou, Guangzhou Medical University, Huizhou, Guangdong 516002, P.R. China
| |
Collapse
|