51
|
Aluganti Narasimhulu C, Singla DK. Doxorubicin-induced apoptosis enhances monocyte infiltration and adverse cardiac remodeling in diabetic animals. Can J Physiol Pharmacol 2022; 100:441-452. [PMID: 34932406 PMCID: PMC10720696 DOI: 10.1139/cjpp-2021-0596] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetic cancer patients were treated with doxorubicin (DOX), a potent chemotherapeutic drug that induces cardiac toxicity; however, molecular mechanisms of cardiac toxicity in this specific disease progression in patients and animal models are completely unknown. Therefore, we designed a study to understand the effects of DOX-induced cardiac toxicity in diabetic animals and the involved pathophysiological mechanisms. C57BL/6 J mice were divided into four DOX- and diabetic (streptozotocin; STZ) - treated groups; control, STZ, DOX, and DOX+STZ. At day 14, animals were sacrificed, echocardiography was used to examine heart function, and heart and blood samples were collected to investigate apoptotic mechanisms (caspase 3, BAX, B-Cell leukemia/lymphoma 2 (Bcl2)), inflammation, and cardiac remodeling. Our data shows a significant (p < 0.05) increase in glucose levels, apoptotic markers, and monocyte infiltration at the site of apoptosis and triggered inflammatory immune response (tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6)), in DOX+STZ animals compared with control and experimental groups. We also observed significant (p < 0.05) increase in myofibrillar area, fibrosis, and significantly decreased (p < 0.05) cardiac function in DOX-treated diabetic animals compared with controls. In conclusion, our data suggest that DOX induces significantly increased apoptosis, fibrosis, and structural alterations in diabetic hearts compared with non-diabetic animals.
Collapse
Affiliation(s)
- Chandrakala Aluganti Narasimhulu
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
52
|
Signaling cascades in the failing heart and emerging therapeutic strategies. Signal Transduct Target Ther 2022; 7:134. [PMID: 35461308 PMCID: PMC9035186 DOI: 10.1038/s41392-022-00972-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure is the end stage of cardiac diseases. With a high prevalence and a high mortality rate worldwide, chronic heart failure is one of the heaviest health-related burdens. In addition to the standard neurohormonal blockade therapy, several medications have been developed for chronic heart failure treatment, but the population-wide improvement in chronic heart failure prognosis over time has been modest, and novel therapies are still needed. Mechanistic discovery and technical innovation are powerful driving forces for therapeutic development. On the one hand, the past decades have witnessed great progress in understanding the mechanism of chronic heart failure. It is now known that chronic heart failure is not only a matter involving cardiomyocytes. Instead, chronic heart failure involves numerous signaling pathways in noncardiomyocytes, including fibroblasts, immune cells, vascular cells, and lymphatic endothelial cells, and crosstalk among these cells. The complex regulatory network includes protein-protein, protein-RNA, and RNA-RNA interactions. These achievements in mechanistic studies provide novel insights for future therapeutic targets. On the other hand, with the development of modern biological techniques, targeting a protein pharmacologically is no longer the sole option for treating chronic heart failure. Gene therapy can directly manipulate the expression level of genes; gene editing techniques provide hope for curing hereditary cardiomyopathy; cell therapy aims to replace dysfunctional cardiomyocytes; and xenotransplantation may solve the problem of donor heart shortages. In this paper, we reviewed these two aspects in the field of failing heart signaling cascades and emerging therapeutic strategies based on modern biological techniques.
Collapse
|
53
|
Nikolov A, Popovski N. Extracellular Matrix in Heart Disease: Focus on Circulating Collagen Type I and III Derived Peptides as Biomarkers of Myocardial Fibrosis and Their Potential in the Prognosis of Heart Failure: A Concise Review. Metabolites 2022; 12:297. [PMID: 35448484 PMCID: PMC9025448 DOI: 10.3390/metabo12040297] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence indicates that two major proteins are responsible for the structural coherence of bounding cardiomyocytes. These biomolecules are known as myocardial fibrillar collagen type I (COL1) and type III (COL3). In addition, fibronectin, laminin, fibrillin, elastin, glycoproteins, and proteoglycans take part in the formation of cardiac extracellular matrix (ECM). In physiological conditions, collagen synthesis and degradation in human cardiac ECM are well-regulated processes, but they can be impaired in certain cardiovascular diseases, such as heart failure (HF). Myocardial remodeling is part of the central mechanism of HF and involves cardiomyocyte injury and cardiac fibrosis due to increased fibrillar collagen accumulation. COL1 and COL3 are predominantly involved in this process. Specific products identified as collagen-derived peptides are released in the circulation as a result of abnormal COL1 and COL3 turnover and myocardial remodeling in HF and can be detected in patients' sera. The role of these products in the pathogenesis of cardiac fibrosis and the possible clinical implications are the focus of numerous investigations. This paper reviews recent studies on COL1- and COL3-derived peptides in patients with HF. Their potential application as indicators of myocardial fibrosis and prognostic markers of HF is also highlighted.
Collapse
Affiliation(s)
- Asparuh Nikolov
- Cardiovascular Research Working Group, Division of Medicine, Institute for Scientific Research, Medical University-Pleven, 5800 Pleven, Bulgaria
| | - Nikola Popovski
- Clinic of Obstetrics and Gynaecology, Department of Obstetrics and Gynaecology, University Hospital Pleven, Medical University-Pleven, 5800 Pleven, Bulgaria
| |
Collapse
|
54
|
Bourque K, Hawey C, Jiang A, Mazarura GR, Hébert TE. Biosensor-based profiling to track cellular signalling in patient-derived models of dilated cardiomyopathy. Cell Signal 2022; 91:110239. [PMID: 34990783 DOI: 10.1016/j.cellsig.2021.110239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathies (DCM) represent a diverse group of cardiovascular diseases impacting the structure and function of the myocardium. To better treat these diseases, we need to understand the impact of such cardiomyopathies on critical signalling pathways that drive disease progression downstream of receptors we often target therapeutically. Our understanding of cellular signalling events has progressed substantially in the last few years, in large part due to the design, validation and use of biosensor-based approaches to studying such events in cells, tissues and in some cases, living animals. Another transformative development has been the use of human induced pluripotent stem cells (hiPSCs) to generate disease-relevant models from individual patients. We highlight the importance of going beyond monocellular cultures to incorporate the influence of paracrine signalling mediators. Finally, we discuss the recent coalition of these approaches in the context of DCM. We discuss recent work in generating patient-derived models of cardiomyopathies and the utility of using signalling biosensors to track disease progression and test potential therapeutic strategies that can be later used to inform treatment options in patients.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Alyson Jiang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Grace R Mazarura
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
55
|
Rodriguez ER, Santos-Martins C, Tan CD. Pathology of cardiac transplantation. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
56
|
Bhullar S, Shah A, Dhalla N. Mechanisms for the development of heart failure and improvement of cardiac function by angiotensin-converting enzyme inhibitors. SCRIPTA MEDICA 2022. [DOI: 10.5937/scriptamed53-36256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors, which prevent the conversion of angiotensin I to angiotensin II, are well-known for the treatments of cardiovascular diseases, such as heart failure, hypertension and acute coronary syndrome. Several of these inhibitors including captopril, enalapril, ramipril, zofenopril and imidapril attenuate vasoconstriction, cardiac hypertrophy and adverse cardiac remodeling, improve clinical outcomes in patients with cardiac dysfunction and decrease mortality. Extensive experimental and clinical research over the past 35 years has revealed that the beneficial effects of ACE inhibitors in heart failure are associated with full or partial prevention of adverse cardiac remodeling. Since cardiac function is mainly determined by coordinated activities of different subcellular organelles, including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils, for regulating the intracellular concentration of Ca2+ and myocardial metabolism, there is ample evidence to suggest that adverse cardiac remodelling and cardiac dysfunction in the failing heart are the consequence of subcellular defects. In fact, the improvement of cardiac function by different ACE inhibitors has been demonstrated to be related to the attenuation of abnormalities in subcellular organelles for Ca2+-handling, metabolic alterations, signal transduction defects and gene expression changes in failing cardiomyocytes. Various ACE inhibitors have also been shown to delay the progression of heart failure by reducing the formation of angiotensin II, the development of oxidative stress, the level of inflammatory cytokines and the occurrence of subcellular defects. These observations support the view that ACE inhibitors improve cardiac function in the failing heart by multiple mechanisms including the reduction of oxidative stress, myocardial inflammation and Ca2+-handling abnormalities in cardiomyocytes.
Collapse
|
57
|
Pathophysiology of heart failure and an overview of therapies. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
58
|
Hsu Y, Huang K, Cheng K. Resuscitating the Field of Cardiac Regeneration: Seeking Answers from Basic Biology. Adv Biol (Weinh) 2021; 6:e2101133. [PMID: 34939372 DOI: 10.1002/adbi.202101133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Heart failure (HF) is one of the leading causes for hospital admissions worldwide. HF patients are classified based on the chronic changes in left ventricular ejection fraction (LVEF) as preserved (LVEF ≥ 50%), reduced (LVEF ≤ 40%), or mid-ranged (40% < LVEF < 50%) HFs. Treatments nowadays can prevent HFrEF progress, whereas only a few of the treatments have been proven to be effective in improving the survival of HFpEF. In this review, numerous mediators involved in the pathogenesis of HF are summarized. The regional upstream signaling and their diagnostic and therapeutic potential are also discussed. Additionally, the recent challenges and development in cardiac regenerative therapy that hold opportunities for future research and clinical translation are discussed.
Collapse
Affiliation(s)
- Yaching Hsu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| |
Collapse
|
59
|
Mascolo A, di Mauro G, Cappetta D, De Angelis A, Torella D, Urbanek K, Berrino L, Nicoletti GF, Capuano A, Rossi F. Current and future therapeutic perspective in chronic heart failure. Pharmacol Res 2021; 175:106035. [PMID: 34915125 DOI: 10.1016/j.phrs.2021.106035] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/29/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
The incidence of heart failure is primarily flat or declining for a presumably reflecting better management of cardiovascular diseases, but that of heart failure with preserved ejection fraction (HFpEF) is probably increasing for the lack of an established effective treatment. Moreover, there is no specific pharmacological treatment for patients with heart failure with mildly reduced ejection fraction (HFmrEF) since no substantial prospective randomized clinical trial has been performed exclusively in such population. According to the recent 2021 European Society of Cardiology (ESC) guidelines, the triad composed of an Angiotensin Converting Enzyme inhibitor or Angiotensin Receptor-Neprilysin Inhibitor (ARNI), a beta-blocker, and a Mineralcorticoid Receptor Antagonist is the cornerstone therapy for all patients with heart failure with reduced ejection fraction (HFrEF) but a substantial gap exists for patients with HFpEF/HFmrEF. Despite the important role of the Renin-Angiotensin-Aldosterone System (RAAS) in heart failure pathophysiology, RAAS blockers were found ineffective for HFpEF patients. Indeed, even the new drug class of ARNI was found effective only in HFrEF patients. In this regard, a therapeutic alternative may be represented by drug stimulating the non-classic RAAS (ACE2 and A1-7) as well as other emerging drug classes (such as SGLT2 inhibitors). Reflecting on this global health burden and the gap in treatments among heart failure phenotypes, we summarize the leading players of heart failure pathophysiology, the available pharmacological treatments for each heart failure phenotype, and that in future development.
Collapse
Affiliation(s)
- Annamaria Mascolo
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Via Costantinopoli 16, 80138 Naples, Italy; Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy.
| | - Gabriella di Mauro
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Via Costantinopoli 16, 80138 Naples, Italy; Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Konrad Urbanek
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Liberato Berrino
- Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Giovanni Francesco Nicoletti
- Plastic Surgery Unit, University of Campania "Luigi Vanvitelli, Multidisciplinary Department of Medical Surgical and Dental Sciences, Napoli, Italy
| | - Annalisa Capuano
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Via Costantinopoli 16, 80138 Naples, Italy; Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Francesco Rossi
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Via Costantinopoli 16, 80138 Naples, Italy; Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
60
|
Sun J, Wang R, Chao T, Wang C. Long Noncoding RNAs Involved in Cardiomyocyte Apoptosis Triggered by Different Stressors. J Cardiovasc Transl Res 2021; 15:588-603. [PMID: 34855148 DOI: 10.1007/s12265-021-10186-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/04/2021] [Indexed: 12/26/2022]
Abstract
Cardiomyocytes are essential to maintain the normal cardiac function. Ischemia, hypoxia, and drug stimulation can induce pathological apoptosis of cardiomyocytes which eventually leads to heart failure, arrhythmia, and other cardiovascular diseases. Understanding the molecular mechanisms that regulate cardiomyocyte apoptosis is of great significance for the prevention and treatment of cardiovascular diseases. In recent years, more and more evidences reveal that long noncoding RNAs (lncRNAs) play important regulatory roles in myocardial cell apoptosis. They can modulate the expression of apoptosis-related genes at post-transcriptional level by altering the translation efficacy of target mRNAs or functioning as a precursor for miRNAs or competing for miRNA-mediated inhibition. Moreover, reversing the abnormal expression of lncRNAs can attenuate and even reverse the pathological apoptosis of cardiomyocytes. Therefore, apoptosis-related lncRNAs may become a potential new field for studying cardiomyocyte apoptosis and provide new ideas for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jinghui Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ru Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tiantian Chao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenglong Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
61
|
Abstract
Although a broad range of viruses cause myocarditis, the mechanisms that underlie viral myocarditis are poorly understood. Here, we report that the M2 gene is a determinant of reovirus myocarditis. The M2 gene encodes outer capsid protein μ1, which mediates host membrane penetration during reovirus entry. We infected newborn C57BL/6 mice with reovirus strain type 1 Lang (T1L) or a reassortant reovirus in which the M2 gene from strain type 3 Dearing (T3D) was substituted into the T1L genetic background (T1L/T3DM2). T1L was non-lethal in wild-type mice, whereas greater than 90% of mice succumbed to T1L/T3DM2 infection. T1L/T3DM2 produced higher viral loads than T1L at the site of inoculation. In secondary organs, T1L/T3DM2 was detected with more rapid kinetics and reached higher peak titers than T1L. We found that hearts from T1L/T3DM2-infected mice were grossly abnormal, with large lesions indicative of substantial inflammatory infiltrate. Lesions in T1L/T3DM2-infected mice contained necrotic cardiomyocytes with pyknotic debris, and extensive lymphocyte and histiocyte infiltration. In contrast, T1L induced the formation of small purulent lesions in a small subset of animals, consistent with T1L being mildly myocarditic. Finally, more activated caspase-3-positive cells were observed in hearts from animals infected with T1L/T3DM2 compared to T1L. Together, our findings indicate that substitution of the T3D M2 allele into an otherwise T1L genetic background is sufficient to change a non-lethal infection into a lethal infection. Our results further indicate that T3D M2 enhances T1L replication and dissemination in vivo, which potentiates the capacity of reovirus to cause myocarditis. IMPORTANCE Reovirus is a non-enveloped virus with a segmented double-stranded RNA genome that serves as a model for studying viral myocarditis. The mechanisms by which reovirus drives myocarditis development are not fully elucidated. We found that substituting the M2 gene from strain type 3 Dearing (T3D) into an otherwise type 1 Lang (T1L) genetic background (T1L/T3DM2) was sufficient to convert the non-lethal T1L strain into a lethal infection in neonatal C57BL/6 mice. T1L/T3DM2 disseminated more efficiently and reached higher maximum titers than T1L in all organs tested, including the heart. T1L is mildly myocarditic and induced small areas of cardiac inflammation in a subset of mice. In contrast, hearts from mice infected with T1L/T3DM2 contained extensive cardiac inflammatory infiltration and more activated caspase-3-positive cells, which is indicative of apoptosis. Together, our findings identify the reovirus M2 gene as a new determinant of reovirus-induced myocarditis.
Collapse
|
62
|
Chunhacha P, Pinkaew D, Sinthujaroen P, Bowles DE, Fujise K. Fortilin inhibits p53, halts cardiomyocyte apoptosis, and protects the heart against heart failure. Cell Death Discov 2021; 7:310. [PMID: 34689154 PMCID: PMC8542040 DOI: 10.1038/s41420-021-00692-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/22/2021] [Accepted: 09/29/2021] [Indexed: 01/01/2023] Open
Abstract
Heart failure (HF) has reached epidemic proportions in developed countries, affecting over 20 million people worldwide. Despite modern medical and device therapies, 60–70% of HF patients still die within 5 years of diagnosis as it relentlessly progresses through pervasive apoptotic loss of cardiomyocytes. Although fortilin, a 172-amino-acid anti-p53 molecule, is one of the most expressed proteins in the heart, its precise role there has remained unknown. Also unclear is how cardiomyocytes are protected against apoptosis. Here, we report that failing human hearts express less fortilin than do non-failing hearts. We also found that mice lacking fortilin in the heart (fortilinKO-heart) die by 9 weeks of age due to extensive cardiomyocyte apoptosis and severe HF, which suggests that fortilin sustains cardiomyocyte viability. The lack of fortilin is also associated with drastic upregulation of p53 target genes in the hearts. The heart-specific deletion of p53 in fortilinKO-heart mice extends their life spans from 9 to 18 weeks by mitigating cardiomyocyte apoptosis. Our data suggest that fortilin is a novel cardiac p53 inhibitor and that its inadequate expression in failing hearts and subsequent overactivation of the p53 apoptosis pathway in cardiomyocytes exacerbates HF.
Collapse
Affiliation(s)
- Preedakorn Chunhacha
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA.,Department of Biochemistry and Microbiology, and Cell-based Drug and Health Product Development Research Unit (CDD), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Decha Pinkaew
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Patuma Sinthujaroen
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA.,Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Dawn E Bowles
- Division of Surgical Sciences, Department of Surgery, Duke University, Durham, NC, 27710, USA
| | - Ken Fujise
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
63
|
Sun J, Zhang C, Zhang Z. Atorvastatin attenuates cardiac hypertrophy through AMPK/miR-143-3p/Bcl2 axis. Arch Physiol Biochem 2021; 127:390-396. [PMID: 31353965 DOI: 10.1080/13813455.2019.1643377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/02/2019] [Accepted: 07/09/2019] [Indexed: 10/26/2022]
Abstract
Atorvastatin is employed as a lipid lowering agent and its heart protective effect has been recently reported as well. However, the mechanism of atorvastatin in attenuating cardiac hypertrophy and inhibiting cardiac failure is unclear. In our study, cardiac hypertrophy was induced in rats using transverse aortic constriction (TAC) method and in cardiomyocytes using angiotensin II (Ang II). Atorvastatin significantly suppressed TAC-induced heart weight increase and cardiomyocytes apoptosis in rats. At a molecular level, we found that miR-143-3p was significantly up-regulated, and the up-regulation could be inhibited by atorvastatin via activating AMPK pathway. Furthermore, it was validated that Bcl2 was one of the target genes of miR-143-3p. Taken together, the data indicated that miR-143-3p aggravated cardiac hypertrophy by inducing cardiomyocytes apoptosis through inhibiting Bcl2 expression. This study demonstrated the effects of atorvastatin in attenuating cardiac hypertrophy and inhibiting cardiac failure, which is depending on Bcl2 expression via miR-143-3p inhibition by AMPK activation.
Collapse
|
64
|
Marunouchi T, Ito T, Onda S, Kyo L, Takahashi K, Uchida M, Yano E, Tanonaka K. Effects of 17-AAG on the RIP1/RIP3/MLKL pathway during the development of heart failure following myocardial infarction in rats. J Pharmacol Sci 2021; 147:192-199. [PMID: 34384567 DOI: 10.1016/j.jphs.2021.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/21/2022] Open
Abstract
In a previous study, we suggested that the Hsp90 inhibitor 17-AAG prevents cardiac dysfunction in the failing heart following myocardial infarction in rats. Although it is assumed that the RIP1/RIP3/MLKL necroptotic pathway, which comprises client proteins for Hsp90, is involved; however, the relationship between the cardioprotective effects of 17-AAG and the activity of the cardiac RIP1/RIP3/MLKL necrosome-associated proteins in the failing heart following myocardial infarction remained unclear. Therefore, the levels of phosphorylated MLKL after myocardial infarction with or without Hsp90 inhibitor treatment were measured. Myocardial infarction was induced by ligation of the coronary artery (CAL) in Wistar rats. 17-AAG was injected from the 2nd to the 8th week after myocardial infarction. The administration of 17-AAG attenuated the cardiac dysfunction, hypertrophy, and fibrosis at the 8th week after CAL, simultaneously lessening the increases in the expression and phosphorylation levels of RIP1, RIP3, and MLKL in the area of the left ventricular muscle without infarct. These results indicate that the activation of the RIP1/RIP3/MLKL pathway is a common event in the development of chronic heart failure. Furthermore, our findings suggest that the effects of 17-AAG treatment on the improvement of cardiac function in rats after myocardial infarction is related to the attenuation of this RIP1/RIP3/MLKL pathway.
Collapse
Affiliation(s)
- Tetsuro Marunouchi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Takumi Ito
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Sumika Onda
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Lina Kyo
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Kirara Takahashi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Manami Uchida
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Emi Yano
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Kouichi Tanonaka
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
65
|
Myocardial dysfunction caused by abemaciclib: a case report. Int Cancer Conf J 2021; 10:324-328. [PMID: 34567946 DOI: 10.1007/s13691-021-00500-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/13/2021] [Indexed: 02/02/2023] Open
Abstract
A 68-year-old female patient with metastatic breast cancer presented 12 weeks after starting chemotherapy with abemaciclib and fulvestrant with breathlessness, peripheral edema, and weight gain. Brain natriuretic peptide (BNP) and troponin I levels were raised above normal, and chest radiography revealed an increase in the cardiothoracic ratio from 47% before chemotherapy to 55%. The transthoracic echocardiogram showed a reduction in left ventricular ejection fraction from 76% before chemotherapy to 68%. Contrast-enhanced cardiac magnetic resonance imaging (MRI) revealed delayed accumulation in the interventricular septum. Under the diagnosis of abemaciclib-induced myocardial dysfunction and heart failure, abemaciclib was discontinued, and enalapril and furosemide were started. Two months later, imaging revealed a cardiothoracic ratio of 47% with a left ventricular ejection fraction of 73%. A cardiac MRI after three months was normal. This case report demonstrates that cardiac dysfunction caused by abemaciclib is reversible if detected early and treated appropriately.
Collapse
|
66
|
Oxygen Delivery Approaches to Augment Cell Survival After Myocardial Infarction: Progress and Challenges. Cardiovasc Toxicol 2021; 22:207-224. [PMID: 34542796 DOI: 10.1007/s12012-021-09696-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/11/2021] [Indexed: 10/20/2022]
Abstract
Myocardial infarction (MI), triggered by blockage of a coronary artery, remains the most common cause of death worldwide. After MI, the capability of providing sufficient blood and oxygen significantly decreases in the heart. This event leads to depletion of oxygen from cardiac tissue and consequently leads to massive cardiac cell death due to hypoxemia. Over the past few decades, many studies have been carried out to discover acceptable approaches to treat MI. However, very few have addressed the crucial role of efficient oxygen delivery to the injured heart. Thus, various strategies were developed to increase the delivery of oxygen to cardiac tissue and improve its function. Here, we have given an overall discussion of the oxygen delivery mechanisms and how the current technologies are employed to treat patients suffering from MI, including a comprehensive view on three major technical approaches such as oxygen therapy, hemoglobin-based oxygen carriers (HBOCs), and oxygen-releasing biomaterials (ORBs). Although oxygen therapy and HBOCs have shown promising results in several animal and clinical studies, they still have a few drawbacks which limit their effectiveness. More recent studies have investigated the efficacy of ORBs which may play a key role in the future of oxygenation of cardiac tissue. In addition, a summary of conducted studies under each approach and the remaining challenges of these methods are discussed.
Collapse
|
67
|
Kaier TE, Alaour B, Marber M. Cardiac troponin and defining myocardial infarction. Cardiovasc Res 2021; 117:2203-2215. [PMID: 33458742 PMCID: PMC8404461 DOI: 10.1093/cvr/cvaa331] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/12/2020] [Indexed: 12/19/2022] Open
Abstract
The 4th Universal Definition of Myocardial Infarction has stimulated considerable debate since its publication in 2018. The intention was to define the types of myocardial injury through the lens of their underpinning pathophysiology. In this review, we discuss how the 4th Universal Definition of Myocardial Infarction defines infarction and injury and the necessary pragmatic adjustments that appear in clinical guidelines to maximize triage of real-world patients.
Collapse
Affiliation(s)
- Thomas E Kaier
- King’s College London BHF Centre, The Rayne Institute, 4th Floor, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Bashir Alaour
- King’s College London BHF Centre, The Rayne Institute, 4th Floor, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Michael Marber
- King’s College London BHF Centre, The Rayne Institute, 4th Floor, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London SE1 7EH, UK
| |
Collapse
|
68
|
He L, Zhao R, Wang Y, Liu H, Wang X. Research Progress on Catalpol as Treatment for Atherosclerosis. Front Pharmacol 2021; 12:716125. [PMID: 34326774 PMCID: PMC8313760 DOI: 10.3389/fphar.2021.716125] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/05/2021] [Indexed: 12/31/2022] Open
Abstract
Coronary atherosclerotic heart disease, cerebrovascular disease, and peripheral artery disease are common diseases with high morbidity and mortality rates and must be addressed. Their most frequent complications, including myocardial infarction and stroke, are caused by spontaneous thrombotic occlusion and are the most frequent cause of death worldwide. Atherosclerosis (AS) is the most widespread underlying pathological change for the above diseases. Therefore, drugs that interfere with this pathophysiological process must be incorporated in the treatment. Chinese traditional and herbal drugs can effectively treat AS. With the development of traditional Chinese medicine, the active ingredients in common Chinese medicinal materials must be thoroughly purified prior to their application in western medicine. Various proprietary Chinese medicine preparations with remarkable effects have been used in AS treatment. Catalpol, the active component of Rehmannia glutinosa, belongs to iridoid terpene and has anti-inflammatory, antioxidant, insulin resistance improvement, and other related effects. Several reviews have been conducted on this compound and its actions against osteoporosis, neurodegenerative diseases, Alzheimer's disease (AD), Parkinson's disease (PD) and diabetes and its complications. The current review focused on catalpol's effect on atherosclerotic plaque formation in different animal models. The potential mechanisms of catalpol to ameliorate AS were also summarized in terms of oxidative stress, inflammation, cell aging, apoptosis, and activation of the silent information regulator factor 2-related enzyme 1 (SIRT1) pathway.
Collapse
Affiliation(s)
- Lei He
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Rusheng Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Youheng Wang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Huibing Liu
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China.,College of Life Science, Henan Normal University, Xinxiang, China
| | - Xuehui Wang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
69
|
Long Noncoding RNA Taurine-Upregulated Gene 1 Knockdown Protects Cardiomyocytes Against Hypoxia/Reoxygenation-induced Injury Through Regulating miR-532-5p/Sox8 Axis. J Cardiovasc Pharmacol 2021; 76:556-563. [PMID: 32833900 DOI: 10.1097/fjc.0000000000000895] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Long noncoding RNA taurine-upregulated gene 1 (TUG1) has been reported to involve in the processing of cardiac ischemia/reperfusion injury after myocardial infarction. Thus, this study further investigates the underlying mechanisms of TUG1 in hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury in vitro. METHODS Cell viability, apoptosis, and migration and invasion were detected using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, flow cytometry, and transwell assay, respectively. Western blot was used to examine the levels of matrix metallopeptidase 9, matrix metallopeptidase 2, and sex determining region Y-box transcription factor 8 (Sox8) protein. Levels of lactate dehydrogenase, malondialdehyde, superoxide dismutase, and glutathione peroxidase were detected using commercial kits. Levels of TUG1, microRNA-532-5p (miR-532-5p), and Sox8 were detected by quantitative real-time polymerase chain reaction. The interaction between miR-532-5p and Sox8 or TUG1 was confirmed by dual-luciferase reporter and RNA immunoprecipitation assay. RESULTS H/R induced rat cardiomyocyte H9c2 injury by inhibiting cell viability, migration and invasion, promoting cell apoptosis, and stimulating oxidative stress. H/R-induced H9c2 injury upregulated the level of TUG1, and TUG1 knockdown alleviated H/R-induced cardiomyocyte injury. TUG1 directly bound to miR-532-5p, and miR-532-5p inhibition reversed the action of TUG1 knockdown on H/R-induced cardiomyocyte injury. Sox8 was a target of miR-532-5p, and miR-532-5p blunted H/R-induced cardiomyocyte injury by targeting Sox8. In addition, TUG1 knockdown inhibited H/R-induced Sox8 elevation through miR-532-5p in H9c2 cells. CONCLUSION TUG1 silence ameliorated H/R-induced cardiomyocytes injury through regulating miR-532-5p/Sox8 axis, suggesting a potential therapeutic target for preventing myocardial ischemia/reperfusion injury.
Collapse
|
70
|
Westbury B, Bolus D, DeLeon-Pennell KY. Find the stimulus, save the heart: a heroes' story. Am J Physiol Heart Circ Physiol 2021; 320:H2185-H2187. [PMID: 33891514 PMCID: PMC8289356 DOI: 10.1152/ajpheart.00194.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 11/22/2022]
Affiliation(s)
- Baylee Westbury
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dawson Bolus
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
71
|
Baik AH, Oluwole OO, Johnson DB, Shah N, Salem JE, Tsai KK, Moslehi JJ. Mechanisms of Cardiovascular Toxicities Associated With Immunotherapies. Circ Res 2021; 128:1780-1801. [PMID: 33934609 PMCID: PMC8159878 DOI: 10.1161/circresaha.120.315894] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immune-based therapies have revolutionized cancer treatments. Cardiovascular sequelae from these treatments, however, have emerged as critical complications, representing new challenges in cardio-oncology. Immune therapies include a broad range of novel drugs, from antibodies and other biologics, including immune checkpoint inhibitors and bispecific T-cell engagers, to cell-based therapies, such as chimeric-antigen receptor T-cell therapies. The recognition of immunotherapy-associated cardiovascular side effects has also catapulted new research questions revolving around the interactions between the immune and cardiovascular systems, and the signaling cascades affected by T cell activation, cytokine release, and immune system dysregulation. Here, we review the specific mechanisms of immune activation from immunotherapies and the resulting cardiovascular toxicities associated with immune activation and excess cytokine production.
Collapse
Affiliation(s)
- Alan H Baik
- Division of Cardiovascular Medicine, Department of Medicine, UCSF, San Francisco, CA (A.H.B.)
| | - Olalekan O Oluwole
- Division of Oncology (D.B.J., J.J.M., O.O.O.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Douglas B Johnson
- Division of Oncology (D.B.J., J.J.M., O.O.O.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Nina Shah
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA (N.S., K.K.T.)
| | - Joe-Elie Salem
- Department of Pharmacology, Cardio-oncology Program, CIC-1901, APHP.Sorbonne Université, Paris, France (J.-E.S.)
- Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.-E.S., J.J.M.)
| | - Katy K Tsai
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA (N.S., K.K.T.)
| | - Javid J Moslehi
- Division of Cardiovascular Medicine (J.J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Division of Oncology (D.B.J., J.J.M., O.O.O.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.-E.S., J.J.M.)
| |
Collapse
|
72
|
Li K, van Delft MF, Dewson G. Too much death can kill you: inhibiting intrinsic apoptosis to treat disease. EMBO J 2021; 40:e107341. [PMID: 34037273 DOI: 10.15252/embj.2020107341] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
Apoptotic cell death is implicated in both physiological and pathological processes. Since many types of cancerous cells intrinsically evade apoptotic elimination, induction of apoptosis has become an attractive and often necessary cancer therapeutic approach. Conversely, some cells are extremely sensitive to apoptotic stimuli leading to neurodegenerative disease and immune pathologies. However, due to several challenges, pharmacological inhibition of apoptosis is still only a recently emerging strategy to combat pathological cell loss. Here, we describe several key steps in the intrinsic (mitochondrial) apoptosis pathway that represent potential targets for inhibitors in disease contexts. We also discuss the mechanisms of action, advantages and limitations of small-molecule and peptide-based inhibitors that have been developed to date. These inhibitors serve as important research tools to dissect apoptotic signalling and may foster new treatments to reduce unwanted cell loss.
Collapse
Affiliation(s)
- Kaiming Li
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, Australia
| | - Mark F van Delft
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, Australia
| | - Grant Dewson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, Australia
| |
Collapse
|
73
|
Huang J, Zhang W, Zhang CL, Wang L. Interleukin-17 aggravates right ventricular remodeling via activating STAT3 under both normoxia and hypoxia. BMC Cardiovasc Disord 2021; 21:249. [PMID: 34020615 PMCID: PMC8139008 DOI: 10.1186/s12872-021-02069-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/17/2021] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Proinflammatory cytokine interleukin 17 (IL-17) is involved in ventricular remodeling, mainly of the left ventricle. This study was designed to explore the role of IL-17 played in the pathogenesis of right ventricular hypertrophy (RVH), aiming to provide a novel treatment target or diagnostic biomarker options for improving the care of RVH patients. METHODS C57BL/6 mice were maintained in 10% O2 chamber or room air for four weeks. Right ventricular hypertrophy index (RVHI), RV/body weight ratio, pulmonary arteriolar remodeling determined by percent media thickness (%MT), and the cardiomyocyte diameter of RV were evaluated. Mice were treated with exogenous recombinant mouse IL-17 (rmIL-17, 1 μg per dose twice a week) for four weeks. H9c2 cardiomyocytes were cultured and treated with IL-17 (10 ng/mL) and STAT3 inhibitor (10 ng/mL) either under normoxia (21% O2, 5% CO2, 74% N2) or under hypoxia (3% O2, 5% CO2, 92% N2). Cardiomyocyte viability was assessed by Cell counting kit 8 (CCK-8) assay. The mRNA level was detected by RT-PCR, where as the protein expression was measured by Western blot, immunohistochemistry, and immunofluorescent analyses. RESULTS In vivo experiments showed that IL-17 did not affect the pulmonary artery under normoxia, after treatment with rmIL-17, %MT was not changed, while RVHI and the RV/body weight ratio were increased, indicating that IL-17 directly induced right ventricular hypertrophy. In a time-course study, the mice were exposed to hypoxia for 0, 1, 2, 3, 4 weeks, respectively. We found that the expression of IL-17 was gradually upregulated in RV tissue in a time-dependent manner after one week of hypoxia exposure, especially at the third and fourth week. Cardiomyocyte hypertrophy and apoptosis were observed after the exposure of the mice to hypoxia for four weeks, rmIL-17 further aggravated the hypoxia-induced cardiomyocyte hypertrophy and apoptosis. The expression of p-STAT3 in the IL-17-deficient mice was lower than in the wild-type mice. In vitro, IL-17 inhibited cardiomyocyte viability and induced cardiomyocyte apoptosis via STAT3 under both normoxic and hypoxic conditions. CONCLUSIONS These findings support a role for IL-17 as a mediator in the pathogenesis RVH, which might be considered as a potential novel anti-inflammation therapeutic strategy or diagnostic biomarker for RVH.
Collapse
MESH Headings
- Animals
- Cell Hypoxia
- Cell Line
- Disease Models, Animal
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- Hypoxia/metabolism
- Hypoxia/pathology
- Hypoxia/physiopathology
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Interleukin-17/toxicity
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phosphorylation
- Rats
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Ventricular Function, Right/drug effects
- Ventricular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Jing Huang
- Department of Rheumatism and Immunology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Wei Zhang
- Department of Emergency Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Cai-Lian Zhang
- Department of Pulmonary and Critical Care Medicine, Yanan University Affiliated Hospital, Yanan, 716000, People's Republic of China
| | - Lei Wang
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xin Cheng District, Xi'an, 710004, People's Republic of China.
| |
Collapse
|
74
|
Ma X, Zhang Q, Zhu H, Huang K, Pang W, Zhang Q. Establishment and analysis of the lncRNA-miRNA-mRNA network based on competitive endogenous RNA identifies functional genes in heart failure. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:4011-4026. [PMID: 34198423 DOI: 10.3934/mbe.2021201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Heart failure (HF), a common disease in adults, accounts for significantly global morbidity and mortality. Due to population aging, therapeutic progression in acute cardiovascular events, the prevalence of HF is increasing, in spite of the efficacy of multiple therapies for HF patients with decreased ejection fraction. Despite great progress in the underlying molecular mechanisms, it remains incompletely clear of the function of competing endogenous RNA (ceRNA) network in HF pathogenesis. Herein, we established an HF-related ceRNA network on the basis of differentially expressed long noncoding RNAs (lncRNAs), microRNAs (miRNAs) as well as mRNAs from GSE136547 and GSE124401 datasets. In brief, the ceRNA network composed of 58 mRNA nodes, 5 miRNA nodes, 82 lncRNA nodes as well as 252 edges. In addition, three lncRNAs (KCNQ1OT1, XIST and AC010336) with higher node degrees than other lncRNAs were chosen as hub nodes. At the same time, we have established five subnetwork of miR-17-5p, miR-20b-5p, miR-107, miR-125a-5p and miR-140-5p centered ceRNA. Pathway analysis revealed the enrichment of ceRNA network in cell cycle pathways. Collectively, our research sheds new lights on the essential functions of ceRNA network in HF development, which also suggests possible application of these hub nodes as diagnostic biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Xudan Ma
- Cardiothoracic Department, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qijun Zhang
- Cardiothoracic Department, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Haihong Zhu
- Cardiothoracic Surgery Department, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kefeng Huang
- Cardiothoracic Surgery Department, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Weina Pang
- Cardiothoracic Surgery Department, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qin Zhang
- Cardiothoracic Surgery Department, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
75
|
Marunouchi T, Nishiumi C, Iinuma S, Yano E, Tanonaka K. Effects of Hsp90 inhibitor on the RIP1-RIP3-MLKL pathway during the development of heart failure in mice. Eur J Pharmacol 2021; 898:173987. [PMID: 33640405 DOI: 10.1016/j.ejphar.2021.173987] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Abstract
Necroptosis is a programmed form of necrotic cell death. Necroptosis is regulated by the necroptosis-regulating proteins including receptor-interacting protein (RIP) 1, RIP3, and mixed lineage kinase domain-like (MLKL), the activities of which are modulated by the molecular chaperone heat-shock protein (Hsp) 90. Presently, to clarify the relationship between Hsp90 and necroptotic pathway proteins, RIP1, RIP3, and MLKL in the development of heart failure, we examined the effects of Hsp90 inhibitor treatment on the RIP1-RIP3-MLKL pathway in mice following transverse aortic constriction (TAC). In this study, TAC mice showed typical signs of heart failure at the 8th week after the operation. In the failing heart, the levels of these regulatory proteins and those of their phosphorylated forms were increased, suggesting that necroptosis contributed to the development of heart failure in the TAC mice. The increases in RIP1, RIP3, and MLKL after TAC were reversed by the administration of an Hsp90 inhibitor. Furthermore, the rise in the phosphorylation levels of these 3 proteins were attenuated by the Hsp90 inhibitor. Concomitantly, cardiac functions were preserved. We also found that exposure of cultured adult mouse cardiomyocytes to the Hsp90 inhibitor attenuated necrotic cell death induced by tumor necrosis factor-α via suppression of RIP1, RIP3, and MLKL activation in in vitro experiments. Taken together, our findings suggest that inhibition of Hsp90 should have therapeutic effects by reducing the activation of RIP1-RIP3-MLKL pathway in the hypertrophied heart and thus could be a new therapeutic strategy for chronic heart failure.
Collapse
Affiliation(s)
- Tetsuro Marunouchi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Chiharu Nishiumi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Saki Iinuma
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Emi Yano
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Kouichi Tanonaka
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan.
| |
Collapse
|
76
|
Targets identified from exercised heart: killing multiple birds with one stone. NPJ Regen Med 2021; 6:23. [PMID: 33837221 PMCID: PMC8035363 DOI: 10.1038/s41536-021-00128-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are a major cause of mortality worldwide, which are mainly driven by factors such as aging, sedentary lifestyle, and excess alcohol use. Exercise targets several molecules and protects hearts against many of these physiological and pathological stimuli. Accordingly, it is widely recognized as an effective therapeutic strategy for CVD. To investigate the molecular mechanism of exercise in cardiac protection, we identify and describe several crucial targets identified from exercised hearts. These targets include insulin-like growth factor 1 (IGF1)-phosphatidylinositol 3 phosphate kinase (PI3K)/protein kinase B (AKT), transcription factor CCAAT/enhancer-binding protein β (C/EBPβ), cardiac microRNAs (miRNAs, miR-222 and miR-17-3p etc.), exosomal-miRNAs (miR-342, miR-29, etc.), Sirtuin 1 (SIRT1), and nuclear factor erythroid 2‑related factor/metallothioneins (Nrf2/Mts). Targets identified from exercised hearts can alleviate injury via multiple avenues, including: (1) promoting cardiomyocyte proliferation; (2) facilitating cardiomyocyte growth and physiologic hypertrophy; (3) elevating the anti-apoptotic capacity of cardiomyocytes; (4) improving vascular endothelial function; (5) inhibiting pathological remodeling and fibrosis; (6) promoting extracellular vesicles (EVs) production and exosomal-molecules transfer. Exercise is one treatment (‘stone’), which is cardioprotective via multiple avenues (‘birds’), and is considered ‘killing multiple birds with one stone’ in this review. Further, we discuss the potential application of EV cargos in CVD treatment. We provide an outline of targets identified from the exercised heart and their mechanisms, as well as novel ideas for CVD treatment, which may provide novel direction for preclinical trials in cardiac rehabilitation.
Collapse
|
77
|
Karwi QG, Ho KL, Pherwani S, Ketema EB, Sun QY, Lopaschuk GD. Concurrent diabetes and heart failure: interplay and novel therapeutic approaches. Cardiovasc Res 2021; 118:686-715. [PMID: 33783483 DOI: 10.1093/cvr/cvab120] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus increases the risk of developing heart failure, and the co-existence of both diseases worsens cardiovascular outcomes, hospitalization and the progression of heart failure. Despite current advancements on therapeutic strategies to manage hyperglycemia, the likelihood of developing diabetes-induced heart failure is still significant, especially with the accelerating global prevalence of diabetes and an ageing population. This raises the likelihood of other contributing mechanisms beyond hyperglycemia in predisposing diabetic patients to cardiovascular disease risk. There has been considerable interest in understanding the alterations in cardiac structure and function in the diabetic patients, collectively termed as "diabetic cardiomyopathy". However, the factors that contribute to the development of diabetic cardiomyopathies is not fully understood. This review summarizes the main characteristics of diabetic cardiomyopathies, and the basic mechanisms that contribute to its occurrence. This includes perturbations in insulin resistance, fuel preference, reactive oxygen species generation, inflammation, cell death pathways, neurohormonal mechanisms, advanced glycated end-products accumulation, lipotoxicity, glucotoxicity, and posttranslational modifications in the heart of the diabetic. This review also discusses the impact of antihyperglycemic therapies on the development of heart failure, as well as how current heart failure therapies influence glycemic control in diabetic patients. We also highlight the current knowledge gaps in understanding how diabetes induces heart failure.
Collapse
Affiliation(s)
- Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Kim L Ho
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Simran Pherwani
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ezra B Ketema
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qiu Yu Sun
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
78
|
Regulation of cardiomyocyte DNA damage and cell death by the type 2A protein phosphatase regulatory protein alpha4. Sci Rep 2021; 11:6293. [PMID: 33737606 PMCID: PMC7973735 DOI: 10.1038/s41598-021-85616-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 02/03/2021] [Indexed: 12/05/2022] Open
Abstract
The type 2A protein phosphatase regulatory protein alpha4 (α4) constitutes an anti-apoptotic protein in non-cardiac tissue, however it’s anti-apoptotic properties in the heart are poorly defined. To this end, we knocked down α4 protein expression (α4 KD) using siRNA in cultured H9c2 cardiomyocytes and confirmed the lack of DNA damage/cell death by TUNEL staining and MTT assay. However, α4 KD did increase the phosphorylation of p53 and ATM/ATR substrates, decreased the expression of poly ADP-ribose polymerase and associated fragments. Expression of anti-apoptotic proteins Bcl-2 and Bcl-xL was reduced, whereas expression of pro-apoptotic BAX protein did not change. Alpha4 KD reduced basal H2AX Ser139 phosphorylation, whereas adenoviral-mediated re-expression of α4 protein following α4 KD, restored basal H2AX phosphorylation at Ser139. The sensitivity of H9c2 cardiomyocytes to doxorubicin-induced DNA damage and cytotoxicity was augmented by α4 KD. Adenoviral-mediated overexpression of α4 protein in ARVM increased PP2AC expression and augmented H2AX Ser139 phosphorylation in response to doxorubicin. Furthermore, pressure overload-induced heart failure was associated with reduced α4 protein expression, increased ATM/ATR protein kinase activity, increased H2AX expression and Ser139 phosphorylation. Hence, this study describes the significance of altered α4 protein expression in the regulation of DNA damage, cardiomyocyte cell death and heart failure.
Collapse
|
79
|
Lai J, Chen C. The Role of Epoxyeicosatrienoic Acids in Cardiac Remodeling. Front Physiol 2021; 12:642470. [PMID: 33716791 PMCID: PMC7943617 DOI: 10.3389/fphys.2021.642470] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid by cytochrome P450 (CYP) epoxygenases, which include four regioisomers: 5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET. Each of them possesses beneficial effects against inflammation, fibrosis, and apoptosis, which could combat cardiovascular diseases. Numerous studies have demonstrated that elevation of EETs by overexpression of CYP2J2, inhibition of sEH, or treatment with EET analogs showed protective effects in various cardiovascular diseases, including hypertension, myocardial infarction, and heart failure. As is known to all, cardiac remodeling is the major pathogenesis of cardiovascular diseases. This review will begin with the introduction of EETs and their protective effects in cardiovascular diseases. In the following, the roles of EETs in cardiac remodeling, with a particular emphasis on myocardial hypertrophy, apoptosis, fibrosis, inflammation, and angiogenesis, will be summarized. Finally, it is suggested that upregulation of EETs is a potential therapeutic strategy for cardiovascular diseases. The EET-related drug development against cardiac remodeling is also discussed, including the overexpression of CYP2J2, inhibition of sEH, and the analogs of EET.
Collapse
Affiliation(s)
- Jinsheng Lai
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
80
|
Gou D, Zhou J, Song Q, Wang Z, Bai X, Zhang Y, Zuo M, Wang F, Chen A, Yousaf M, Yang Z, Peng H, Li K, Xie W, Tang J, Yao Y, Han M, Ke T, Chen Q, Xu C, Wang Q. Mog1 knockout causes cardiac hypertrophy and heart failure by downregulating tbx5-cryab-hspb2 signalling in zebrafish. Acta Physiol (Oxf) 2021; 231:e13567. [PMID: 33032360 DOI: 10.1111/apha.13567] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/09/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
AIMS MOG1 is a small protein that can bind to small GTPase RAN and regulate transport of RNA and proteins between the cytoplasm and nucleus. However, the in vivo physiological role of mog1 in the heart needs to be fully defined. METHODS Mog1 knockout zebrafish was generated by TALEN. Echocardiography, histological analysis, and electrocardiograms were used to examine cardiac structure and function. RNA sequencing and real-time RT-PCR were used to elucidate the molecular mechanism and to analyse the gene expression. Isoproterenol was used to induce cardiac hypertrophy. Whole-mount in situ hybridization was used to observe cardiac morphogenesis. RESULTS Mog1 knockout zebrafish developed cardiac hypertrophy and heart failure (enlarged pericardium, increased nppa and nppb expression and ventricular wall thickness, and reduced ejection fraction), which was aggravated by isoproterenol. RNAseq and KEGG pathway analyses revealed the effect of mog1 knockout on the pathways of cardiac hypertrophy, dilatation and contraction. Mechanistic studies revealed that mog1 knockout decreased expression of tbx5, which reduced expression of cryab and hspb2, resulting in cardiac hypertrophy and heart failure. Overexpression of cryab, hspb2 and tbx5 rescued the cardiac oedema phenotype of mog1 KO zebrafish. Telemetry electrocardiogram monitoring showed QRS and QTc prolongation and a reduced heart rate in mog1 knockout zebrafish, which was associated with reduced scn1b expression. Moreover, mog1 knockout resulted in abnormal cardiac looping during embryogenesis because of the reduced expression of nkx2.5, gata4 and hand2. CONCLUSION Our data identified an important molecular determinant for cardiac hypertrophy and heart failure, and rhythm maintenance of the heart.
Collapse
Affiliation(s)
- Dongzhi Gou
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Juan Zhou
- School of Basic Medicine Gannan Medical University Ganzhou P. R. China
| | - Qixue Song
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Zhijie Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Xuemei Bai
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Yidan Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Mengxia Zuo
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Fan Wang
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Department of Cardiovascular Medicine Cleveland Clinic Cleveland OH USA
- Department of Molecular Medicine Cleveland Clinic Lerner College of Medicine of CaseWestern Reserve University Cleveland OH USA
| | - Ailan Chen
- Department of Cardiology Guangzhou Medical University Guangzhou P. R. China
| | - Muhammad Yousaf
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Zhongcheng Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Huixing Peng
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Ke Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Wen Xie
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Jingluo Tang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Meng Han
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Tie Ke
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Department of Cardiovascular Medicine Cleveland Clinic Cleveland OH USA
- Department of Molecular Medicine Cleveland Clinic Lerner College of Medicine of CaseWestern Reserve University Cleveland OH USA
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Qing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Department of Cardiovascular Medicine Cleveland Clinic Cleveland OH USA
- Department of Molecular Medicine Cleveland Clinic Lerner College of Medicine of CaseWestern Reserve University Cleveland OH USA
- Department of Genetics and Genome Science Case Western Reserve University School of Medicine Cleveland OH USA
| |
Collapse
|
81
|
Lou T, Ma J, Xie Y, Yao G, Fan Y, Ma S, Zou X. Nuanxin capsule enhances cardiac function by inhibiting oxidative stress-induced mitochondrial dependent apoptosis through AMPK/JNK signaling pathway. Biomed Pharmacother 2021; 135:111188. [PMID: 33418304 DOI: 10.1016/j.biopha.2020.111188] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Oxidative stress and apoptosis play critical roles in the pathogenesis of heart failure (HF).Nuanxin capsule (NX) is a Chinese medicine that has outstanding protective effects on HF. The present study aimed to elucidate whether NX could protect HF against oxidative stress-induced apoptosis through intrinsic mitochondrial pathway. METHODS In vivo, HF was induced by transverse aortic constriction. NX and Compound C (Comp C) were administered to C57BL/6 J mice for over a 4-week period. Cardiac function was assessed with echocardiography. In vitro, H9c2 cells were exposed to H2O2 in the presence or absence of NX and Compound C. Cell viability, cytotoxicity, reactive oxygen species (ROS) production, apoptosis, mitochondrial membrane potential (ΔΨm) and mitochondrial function by oxygen consumption rate (OCR) were detected. The expressions of cytochrome c, BAX, Bcl-2, cleaved caspase-3, AMPK and JNK were evaluated by western blotting. RESULTS The results indicated that NX significantly improved cardiac function and enhanced the cell viability, ΔΨm and mitochondrial respiration. Also NX treatment reduced cell cytotoxicity and ROS production. Moreover, NX inhibited mitochondrial-mediated apoptosis by upregulating AMPK and downregulating JNK both in vivo and in vitro. The protective effects of NX on cardiac function by reducing oxidative stress-induced mitochondrial dependent apoptosis were reversed by Compound C treatment. CONCLUSIONS These findings demonstrated that NX effectively improved cardiac function in TAC mice by reducing oxidative stress-induced mitochondrial dependent apoptosis by activating AMPK/JNK signaling pathway.
Collapse
Affiliation(s)
- Tiantian Lou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Jin Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Yanzheng Xie
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Gengzhen Yao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Ye Fan
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Shiyu Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China.
| | - Xu Zou
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China; Dongguan Kanghua Hospital, Dongguan, 523080, China.
| |
Collapse
|
82
|
Shao M, Lu L, Wang Q, Ma L, Tian X, Li C, Li C, Guo D, Wang Q, Wang W, Wang Y. The multi-faceted role of retinoid X receptor in cardiovascular diseases. Biomed Pharmacother 2021; 137:111264. [PMID: 33761589 DOI: 10.1016/j.biopha.2021.111264] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 01/14/2023] Open
Abstract
Retinoid X receptors (RXRs) are members of ligand-dependent transcription factors whose effects on a diversity of cellular processes, including cellular proliferation, the immune response, and lipid and glucose metabolism. Knock out of RXRα causes a hypoplasia of the myocardium which is lethal during fetal life. In addition, the heart maintains a well-orchestrated balances in utilizing fatty acids (FAs) and other substrates to meet the high energy requirements. As the master transcriptional regulators of lipid metabolism, RXRs become particularly important for the energy needs of the heart. Accumulating evidence suggested that RXRs may exert direct beneficial effects in the heart both through heterodimerization with other nuclear receptors (NRs) and homodimerization, thus standing as suitable targets for treating in cardiovascular diseases. Although compounds that target RXRs are promising drugs, their use is limited by toxicity. A better understanding of the structural biology of RXRs in cardiovascular disease should enable the rational design of more selective nuclear receptor modulators to overcome these problems. Here, this review summarizes a brief overview of RXRs structure and versatility of RXR action in the control of cardiovascular diseases. And we also discussed the therapeutic potential of RXR ligand.
Collapse
Affiliation(s)
- Mingyan Shao
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Linghui Lu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lin Ma
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue Tian
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Changxiang Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chun Li
- Modern Research Center of Traditional Chinese Medicine, School of Traditional Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dongqing Guo
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qiyan Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wei Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yong Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China; College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
83
|
Liu J, Zhong L, Guo R. The Role of Posttranslational Modification and Mitochondrial Quality Control in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6635836. [PMID: 33680284 PMCID: PMC7910068 DOI: 10.1155/2021/6635836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/26/2021] [Accepted: 02/05/2021] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the world. The mechanism behind CVDs has been studied for decades; however, the pathogenesis is still controversial. Mitochondrial homeostasis plays an essential role in maintaining the normal function of the cardiovascular system. The alterations of any protein function in mitochondria may induce abnormal mitochondrial quality control and unexpected mitochondrial dysfunction, leading to CVDs. Posttranslational modifications (PTMs) affect protein function by reversibly changing their conformation. This review summarizes how common and novel PTMs influence the development of CVDs by regulating mitochondrial quality control. It provides not only ideas for future research on the mechanism of some types of CVDs but also ideas for CVD treatments with therapeutic potential.
Collapse
Affiliation(s)
- Jinlin Liu
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| |
Collapse
|
84
|
Xu S, Tao H, Cao W, Cao L, Lin Y, Zhao SM, Xu W, Cao J, Zhao JY. Ketogenic diets inhibit mitochondrial biogenesis and induce cardiac fibrosis. Signal Transduct Target Ther 2021; 6:54. [PMID: 33558457 PMCID: PMC7870678 DOI: 10.1038/s41392-020-00411-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/31/2020] [Accepted: 11/03/2020] [Indexed: 02/05/2023] Open
Abstract
In addition to their use in relieving the symptoms of various diseases, ketogenic diets (KDs) have also been adopted by healthy individuals to prevent being overweight. Herein, we reported that prolonged KD exposure induced cardiac fibrosis. In rats, KD or frequent deep fasting decreased mitochondrial biogenesis, reduced cell respiration, and increased cardiomyocyte apoptosis and cardiac fibrosis. Mechanistically, increased levels of the ketone body β-hydroxybutyrate (β-OHB), an HDAC2 inhibitor, promoted histone acetylation of the Sirt7 promoter and activated Sirt7 transcription. This in turn inhibited the transcription of mitochondrial ribosome-encoding genes and mitochondrial biogenesis, leading to cardiomyocyte apoptosis and cardiac fibrosis. Exogenous β-OHB administration mimicked the effects of a KD in rats. Notably, increased β-OHB levels and SIRT7 expression, decreased mitochondrial biogenesis, and increased cardiac fibrosis were detected in human atrial fibrillation heart tissues. Our results highlighted the unknown detrimental effects of KDs and provided insights into strategies for preventing cardiac fibrosis in patients for whom KDs are medically necessary.
Collapse
Affiliation(s)
- Sha Xu
- Zhongshan Hospital of Fudan University, Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, Key Laboratory of Reproduction Regulation of NPFPC, and Institutes of Biomedical Sciences, Fudan University, 200438, Shanghai, China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Hui Tao
- Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, and Cardiovascular Research Center, Anhui Medical University, 230601, Hefei, China
| | - Wei Cao
- Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, and Cardiovascular Research Center, Anhui Medical University, 230601, Hefei, China
| | - Li Cao
- Zhongshan Hospital of Fudan University, Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, Key Laboratory of Reproduction Regulation of NPFPC, and Institutes of Biomedical Sciences, Fudan University, 200438, Shanghai, China
| | - Yan Lin
- Zhongshan Hospital of Fudan University, Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, Key Laboratory of Reproduction Regulation of NPFPC, and Institutes of Biomedical Sciences, Fudan University, 200438, Shanghai, China
| | - Shi-Min Zhao
- Zhongshan Hospital of Fudan University, Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, Key Laboratory of Reproduction Regulation of NPFPC, and Institutes of Biomedical Sciences, Fudan University, 200438, Shanghai, China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Wei Xu
- Zhongshan Hospital of Fudan University, Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, Key Laboratory of Reproduction Regulation of NPFPC, and Institutes of Biomedical Sciences, Fudan University, 200438, Shanghai, China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Jing Cao
- Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, China
| | - Jian-Yuan Zhao
- Zhongshan Hospital of Fudan University, Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, Key Laboratory of Reproduction Regulation of NPFPC, and Institutes of Biomedical Sciences, Fudan University, 200438, Shanghai, China. .,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China. .,Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, China.
| |
Collapse
|
85
|
Congestive Heart Failure. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00050-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
86
|
Goldblatt ZE, Cirka HA, Billiar KL. Mechanical Regulation of Apoptosis in the Cardiovascular System. Ann Biomed Eng 2021; 49:75-97. [PMID: 33169343 PMCID: PMC7775273 DOI: 10.1007/s10439-020-02659-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022]
Abstract
Apoptosis is a highly conserved physiological process of programmed cell death which is critical for proper organism development, tissue maintenance, and overall organism homeostasis. Proper regulation of cell removal is crucial, as both excessive and reduced apoptotic rates can lead to the onset of a variety of diseases. Apoptosis can be induced in cells in response to biochemical, electrical, and mechanical stimuli. Here, we review literature on specific mechanical stimuli that regulate apoptosis and the current understanding of how mechanotransduction plays a role in apoptotic signaling. We focus on how insufficient or excessive mechanical forces may induce apoptosis in the cardiovascular system and thus contribute to cardiovascular disease. Although studies have demonstrated that a broad range of mechanical stimuli initiate and/or potentiate apoptosis, they are predominantly correlative, and no mechanisms have been established. In this review, we attempt to establish a unifying mechanism for how various mechanical stimuli initiate a single cellular response, i.e. apoptosis. We hypothesize that the cytoskeleton plays a central role in this process as it does in determining myriad cell behaviors in response to mechanical inputs. We also describe potential approaches of using mechanomedicines to treat various diseases by altering apoptotic rates in specific cells. The goal of this review is to summarize the current state of the mechanobiology field and suggest potential avenues where future research can explore.
Collapse
|
87
|
Metabolomic Analysis of the Ameliorative Effect of Enhanced Proline Metabolism on Hypoxia-Induced Injury in Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8866946. [PMID: 33294127 PMCID: PMC7718065 DOI: 10.1155/2020/8866946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/29/2022]
Abstract
Background Coronary heart disease is currently the leading cause of death in humans. Its poor prognosis and high mortality are associated with myocardial ischemia, which leads to metabolic disorder-related cardiomyocyte apoptosis and reactive oxygen species (ROS) production. Previous cardiovascular metabolomics studies in humans and mice have shown that proline metabolism is severely altered after cardiomyocyte hypoxia. Proline dehydrogenase (PRODH) is located on the inner mitochondrial membrane and is an enzyme that catalyzes the first step of proline catabolism, which plays an important role in improving the cellular redox state. In vitro oxygen-glucose deprivation can mimic in vivo myocardial ischemic injury. This study is aimed at investigating whether enhancing proline metabolism by overexpressing PRODH can ameliorate hypoxia-induced injury in cardiomyocytes and to reveal the related altered metabolites and mechanistic pathway via untargeted metabolomics analysis. Methods and Results First, through public database analysis and RT-qPCR and western blot analyses in a cardiomyocyte hypoxia model, we found that the expression of the proline-degrading enzyme PRODH was downregulated after myocardial infarction and hypoxia exposure. Second, LDH assays, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), DHE staining, flow cytometric apoptosis analysis with DCFH and Annexin V-FITC/PI, and western blot analysis were used to assess the injury level in cardiomyocytes. Enhanced proline metabolism induced by PRODH overexpression reduced the levels of reactive oxidative stress and apoptosis, whereas PRODH knockdown had the opposite effects. Third, untargeted metabolomics analysis revealed that the protective effect was associated with significant changes in metabolism linked to sphingolipid signaling pathways, unsaturated fatty acid biosynthesis, phosphocreatine, glutathione disulfide, aminoacyl-tRNA biosynthesis, and ABC transporters. Conclusions Our study demonstrated a protective effect of enhanced proline metabolism in cardiomyocytes under hypoxia, providing a novel strategy for exploring new treatments for coronary heart disease.
Collapse
|
88
|
Abstract
The regenerative capacity of the heart has long fascinated scientists. In contrast to other organs such as liver, skin, and skeletal muscle, the heart possesses only a minimal regenerative capacity. It lacks a progenitor cell population, and cardiomyocytes exit the cell cycle shortly after birth and do not re-enter after injury. Thus, any loss of cardiomyocytes is essentially irreversible and can lead to or exaggerate heart failure, which represents a major public health problem. New therapeutic options are urgently needed, but regenerative therapies have remained an unfulfilled promise in cardiovascular medicine until today. Yet, through a clearer comprehension of signaling pathways that regulate the cardiomyocyte cell cycle and advances in stem cell technology, strategies have evolved that demonstrate the potential to generate new myocytes and thereby fulfill an essential central criterion for heart repair.
Collapse
Affiliation(s)
- Florian Weinberger
- Institute for Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; , .,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Thomas Eschenhagen
- Institute for Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; , .,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
89
|
Crocin ameliorates arsenic trioxide‑induced cardiotoxicity via Keap1-Nrf2/HO-1 pathway: Reducing oxidative stress, inflammation, and apoptosis. Biomed Pharmacother 2020; 131:110713. [PMID: 32920515 DOI: 10.1016/j.biopha.2020.110713] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/16/2020] [Accepted: 08/29/2020] [Indexed: 12/27/2022] Open
Abstract
Arsenic trioxide (ATO) is an excellent therapy for acute promyelocytic leukemia; however, its use is limited due to its cardiotoxicity. Crocin (CRO) possesses abundant pharmacological and biological properties, including antioxidant, anti-inflammatory, and anti-apoptotic. This study examined the cardioprotective effects of crocin and explored their mechanistic involvement in ATO-induced cardiotoxicity. Forty-eight male rats were treated with ATO to induce cardiotoxicity. In combination with ATO, CRO were given to evaluate its cardioprotection. The results demonstrated that CRO administration not only diminished QTc prolongation, myocardial enzymes and Troponin T levels but also improved histopathological results. CRO administration reduced reactive oxygen species generation. However, the CRO administration caused an increase in glutathione, superoxide dismutase, catalase, glutathione peroxidase, glutathione S-transferase and total sulphydryl levels and a decrease in malondialdehyde content, gamma glutamyl transferase and lipid hydroperoxides levels and proinflammatory cytokines. Importantly, immunohistochemical analysis, real time PCR and western blotting showed a reduction in Caspase-3 and Bcl-2-associated X protein expressions and enhancement of B cell lymphoma-2 expression. Real time PCR and western blotting showed a reduction in proinflammatory cytokines. Moreover, CRO caused an activation in nuclear factor erythroid-2 related factor 2, leading to enhanced Kelch-like ECH-associated protein 1, heme oxygenase-1 and nicotinamide adenine dinucleotide quinone dehydrogenase 1 expressions involved in Nrf2 signaling during ATO-induced cardiotoxicity. CRO was shown to ameliorate ATO-induced cardiotoxicity. The mechanisms for CRO amelioration of cardiotoxicity due to inflammation, oxidative damage, and apoptosis may occur via an up-regulated Keap1-Nrf2/HO-1 signaling pathway.
Collapse
|
90
|
Shen Z, Shen A, Chen X, Wu X, Chu J, Cheng Y, Peng M, Chen Y, Weygant N, Wu M, Lin X, Peng J, Chen K. Huoxin pill attenuates myocardial infarction-induced apoptosis and fibrosis via suppression of p53 and TGF-β1/Smad2/3 pathways. Biomed Pharmacother 2020; 130:110618. [PMID: 34321167 DOI: 10.1016/j.biopha.2020.110618] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/27/2020] [Accepted: 08/02/2020] [Indexed: 11/24/2022] Open
Abstract
Huoxin Pill (HXP), a Traditional Chinese Medicine, is used widely to treat patients with coronary heart disease and angina pectoris in China. However, the underlying protective mechanism of HXP on cardiac apoptosis and fibrosis has never been evaluated. Therefore, the aim of this study was to investigate the role of HXP in a myocardial infarction (MI) mouse model. The mice were randomly divided into 3 groups and subjected to surgical ligation of the left anterior descending (LAD) coronary artery or sham surgery (n = 6 for each group) and treated with HXP (50 mg/kg/day) or saline by gavage for 2 weeks. At 2 weeks post MI, we found that HXP significantly enhanced myocardial function and attenuated the increase of heart weight index (HWI) and pathological changes in MI mice. RNA-sequencing and KEGG pathway analyses identified 660 differentially expressed genes and multiple enriched signaling pathways including p53 and TGF-β. In support of these findings, HXP attenuated cardiac apoptosis and decreased p53 and Bax protein expression, while increasing Bcl-2 protein expression in cardiac tissues of MI mice. Furthermore, HXP treatment inhibited cardiac fibrosis and significantly down-regulated TGF-β1 protein expression and Smad2/3 phosphorylation in cardiac tissues. In summary, HXP can improve cardiac function in mice after MI by attenuating cardiac apoptosis and fibrosis partly via supression of the p53/Bax/Bcl-2 and TGF-β1/Smad2/3 pathways.
Collapse
Affiliation(s)
- Zhiqing Shen
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Aling Shen
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Xiaoping Chen
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Xiangyan Wu
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Jianfeng Chu
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Ying Cheng
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Meizhong Peng
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Youqin Chen
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH, 44106, USA.
| | - Nathaniel Weygant
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Meizhu Wu
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Xiaoying Lin
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Jun Peng
- Academy of Integrative Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, Fujian, 350122, China.
| | - Keji Chen
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
91
|
Kianmehr P, Azarbayjani MA, Peeri M, Farzanegi P. Synergic effects of exercise training and octopamine on peroxisome proliferator-activated receptor-gamma coactivator -1a and uncoupling protein 1 mRNA in heart tissue of rat treated with deep frying oil. Biochem Biophys Rep 2020; 22:100735. [PMID: 32140572 PMCID: PMC7047140 DOI: 10.1016/j.bbrep.2020.100735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/18/2020] [Accepted: 01/21/2020] [Indexed: 11/06/2022] Open
Abstract
Octopamine (OCT) have an adverse effect on heart function. One of the positive effects of exercise training is improving cardiac function and cardiomyocytes signaling, which along with herbal supplements can have better effects on the heart tissue. Therefore, the aim of this study was to evaluate the effects of exercise training and OCT on changes of PGC1α and UCP1 expression in heart tissue of rat treated with deep frying oil (DFO). In this study, 45 male wistar rats were divided into 5 groups (n = 9 in each): I) control (Co), II) DFO, III) DFO + exercise, IV) DFO + OCT, and V) DFO + OCT + exercise. The quantification of apoptotic effects of DFO in heart tissue was assessed by TUNEL assay. Masson's trichrome stain applied to study cardiomyocytic fibers. Moreover, PGC1α and UCP1 genes and proteins expression in all groups were investigated using quantitative real-time PCR and immunohistochemical method. A significant increase in apoptotic cells was observed in the DFO-treated group (p < 0.05). In Masson's Trichrome stain study, more cardiomyocytic fibers were observed and some lymphocytic cells were present in some fibers. Also, the expression of PGC1α and UCP1 was significantly increase in DFO + exercise group, DFO + OCT group, and DFO + OCT + exercise group compare to DFO group (p < 0.05). Based on these findings, exercise and octopamine can be considered as factors affecting the expression of PGC1α genes and UCP1 as well as drug poisoning.
Collapse
Affiliation(s)
- Pantea Kianmehr
- -Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Ali Azarbayjani
- -Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maghsoud Peeri
- -Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Parvin Farzanegi
- -Department of Exercise Physiology, Sari Branch, Islamic Azad University, Sari, Iran
| |
Collapse
|
92
|
Qingda granules attenuate hypertensive cardiac remodeling and inflammation in spontaneously hypertensive rats. Biomed Pharmacother 2020; 129:110367. [PMID: 32559624 DOI: 10.1016/j.biopha.2020.110367] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/22/2020] [Accepted: 06/02/2020] [Indexed: 01/08/2023] Open
Abstract
Qingda granules (QDG) are derived from QingXuanJiangYa Decoction (QXJYD) a traditional Chinese medication that has been used to treat hypertension for more than 60 years. QXJYD has been shown to be effective in rat models of hypertension. However, the effects of QDG on hypertension remain largely unknown. In the current study, baicalin was identified as one of the main components of QDG using Ultra Performance Liquid Chromatography (UPLC) analysis. We investigated the effects of QDG on blood pressure, cardiac remodeling, and cardiac inflammation. QDG (0.8 g/kg/day) treatment attenuated the elevated blood pressure in spontaneously hypertensive rats (SHRs). Moreover, QDG treatment reduced the degree of myocardial fiber disarray, degeneration and necrosis of myocardial cells, expression of ANP and BNP, as well as collagen content of SHRs. Moreover, we further assessed the effect of QDG treatment on cardiac inflammation and found that QDG treatment reduced CD68 protein expression, decreased levels of IL-6 and TNF-α in both serum and cardiac tissues, as well as suppressed activation of NF-κB pathway in cardiac tissues of SHRs. Differential expressed metabolites (DEMs) analysis identified 41 increased and 51 decreased metabolites in the cardiac tissues of SHRs after QDG treatment. In summary, QDG treatment of SHRs attenuated the elevated blood pressure and ameliorated cardiac remodeling and inflammation, in part, through suppression of NF-κB pathway and DEMs, which provide a basis for other therapeutic uses of this TCM.
Collapse
|
93
|
Ben-Nun D, Buja LM, Fuentes F. Prevention of heart failure with preserved ejection fraction (HFpEF): reexamining microRNA-21 inhibition in the era of oligonucleotide-based therapeutics. Cardiovasc Pathol 2020; 49:107243. [PMID: 32629211 DOI: 10.1016/j.carpath.2020.107243] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for 50% of cases of heart failure, which is the most common cause of hospitalization in US patients over the age of 65. HFpEF pathogenesis is increasingly believed to be due to pathological hypertrophy and fibrosis of the myocardium that may be a result of systemic inflammation from comorbid conditions such as hypertension, diabetes mellitus, chronic obstructive pulmonary disease, anemia, chronic kidney disease and others. It is believed that oxidative stress triggers a process of pathological hypertrophy and fibrosis in cardiac endothelial cells, which leads to increased left ventricle filling pressures and, eventually, symptoms of heart failure. Numerous recent major clinical trials that have examined various therapies aimed at improving mortality in HFpEF have emerged empty-handed and thus the search for effective management strategies continues. Over the last several years, there have been many new developments in the field of antisense oligonucleotide-based therapeutics, which involves using noncoding nucleic acid particles such as microRNA and small interfering RNA to repress the expression of specific messenger RNA. In this article, we review the concept of using oligonucleotide-based therapeutics to prevent or treat HFpEF by targeting a specific microRNA that has been implicated in the pathogenesis of myocardial fibrosis and hypertrophy, microRNA-21 (miR-21). We review the various evidence that implicates miR-21 in the process of myocardial fibrosis and discuss recent attempts to use antimiR-21 compounds to prevent fibrosis. We also discuss proposed methods for screening patients at high risk for HFpEF for diastolic dysfunction in order to determine which patients.
Collapse
Affiliation(s)
- David Ben-Nun
- Tel Aviv University Sackler Faculty of Medicine, Sackler Faculty of Medicine, NY St..., 69978 Tel Aviv, Israel.
| | - L Maximilian Buja
- The University of Texas Health Science Center at Houston, McGovern Medical School
| | - Francisco Fuentes
- The University of Texas Health Science Center at Houston, McGovern Medical School
| |
Collapse
|
94
|
Szibor M, Schreckenberg R, Gizatullina Z, Dufour E, Wiesnet M, Dhandapani PK, Debska‐Vielhaber G, Heidler J, Wittig I, Nyman TA, Gärtner U, Hall AR, Pell V, Viscomi C, Krieg T, Murphy MP, Braun T, Gellerich FN, Schlüter K, Jacobs HT. Respiratory chain signalling is essential for adaptive remodelling following cardiac ischaemia. J Cell Mol Med 2020; 24:3534-3548. [PMID: 32040259 PMCID: PMC7131948 DOI: 10.1111/jcmm.15043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 01/09/2023] Open
Abstract
Cardiac ischaemia-reperfusion (I/R) injury has been attributed to stress signals arising from an impaired mitochondrial electron transport chain (ETC), which include redox imbalance, metabolic stalling and excessive production of reactive oxygen species (ROS). The alternative oxidase (AOX) is a respiratory enzyme, absent in mammals, that accepts electrons from a reduced quinone pool to reduce oxygen to water, thereby restoring electron flux when impaired and, in the process, blunting ROS production. Hence, AOX represents a natural rescue mechanism from respiratory stress. This study aimed to determine how respiratory restoration through xenotopically expressed AOX affects the re-perfused post-ischaemic mouse heart. As expected, AOX supports ETC function and attenuates the ROS load in post-anoxic heart mitochondria. However, post-ischaemic cardiac remodelling over 3 and 9 weeks was not improved. AOX blunted transcript levels of factors known to be up-regulated upon I/R such as the atrial natriuretic peptide (Anp) whilst expression of pro-fibrotic and pro-apoptotic transcripts were increased. Ex vivo analysis revealed contractile failure at nine but not 3 weeks after ischaemia whilst label-free quantitative proteomics identified an increase in proteins promoting adverse extracellular matrix remodelling. Together, this indicates an essential role for ETC-derived signals during cardiac adaptive remodelling and identified ROS as a possible effector.
Collapse
Affiliation(s)
- Marten Szibor
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
- Department of Cardiothoracic SurgeryJena University HospitalJenaGermany
| | | | | | - Eric Dufour
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Marion Wiesnet
- Department Cardiac Development and RemodellingMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Praveen K. Dhandapani
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| | | | - Juliana Heidler
- Functional ProteomicsFaculty of MedicineGoethe UniversityFrankfurt am MainGermany
| | - Ilka Wittig
- Functional ProteomicsFaculty of MedicineGoethe UniversityFrankfurt am MainGermany
| | - Tuula A. Nyman
- Department of ImmunologyInstitute of Clinical MedicineOslo University HospitalUniversity of OsloOsloNorway
| | - Ulrich Gärtner
- Institute of Anatomy and Cell BiologyJustus‐Liebig‐University GiessenGiessenGermany
| | - Andrew R. Hall
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
| | - Victoria Pell
- Department of MedicineUniversity of CambridgeCambridgeUK
| | - Carlo Viscomi
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Thomas Krieg
- Department of MedicineUniversity of CambridgeCambridgeUK
| | - Michael P. Murphy
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
| | - Thomas Braun
- Department Cardiac Development and RemodellingMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | | | | | - Howard T. Jacobs
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
95
|
Oldfield CJ, Duhamel TA, Dhalla NS. Mechanisms for the transition from physiological to pathological cardiac hypertrophy. Can J Physiol Pharmacol 2020; 98:74-84. [DOI: 10.1139/cjpp-2019-0566] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The heart is capable of responding to stressful situations by increasing muscle mass, which is broadly defined as cardiac hypertrophy. This phenomenon minimizes ventricular wall stress for the heart undergoing a greater than normal workload. At initial stages, cardiac hypertrophy is associated with normal or enhanced cardiac function and is considered to be adaptive or physiological; however, at later stages, if the stimulus is not removed, it is associated with contractile dysfunction and is termed as pathological cardiac hypertrophy. It is during physiological cardiac hypertrophy where the function of subcellular organelles, including the sarcolemma, sarcoplasmic reticulum, mitochondria, and myofibrils, may be upregulated, while pathological cardiac hypertrophy is associated with downregulation of these subcellular activities. The transition of physiological cardiac hypertrophy to pathological cardiac hypertrophy may be due to the reduction in blood supply to hypertrophied myocardium as a consequence of reduced capillary density. Oxidative stress, inflammatory processes, Ca2+-handling abnormalities, and apoptosis in cardiomyocytes are suggested to play a critical role in the depression of contractile function during the development of pathological hypertrophy.
Collapse
Affiliation(s)
- Christopher J. Oldfield
- Faculty of Kinesiology & Recreation Management, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Todd A. Duhamel
- Faculty of Kinesiology & Recreation Management, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
96
|
Mirzadeh Azad F, Arabian M, Maleki M, Malakootian M. Small Molecules with Big Impacts on Cardiovascular Diseases. Biochem Genet 2020; 58:359-383. [PMID: 31997044 DOI: 10.1007/s10528-020-09948-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 01/13/2020] [Indexed: 12/22/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Although in recent years there has been a significant progress in the diagnosis, treatment, and prognosis of CVD, but due to their complex pathobiology, developing novel biomarkers and therapeutic interventions are still in need. MicroRNAs (miRNAs) are a fraction of non-coding RNAs that act as micro-regulators of gene expression. Mounting evidences over the last decade confirmed that microRNAs were deregulated in several CVDs and manipulating their expression could affect homeostasis, differentiation, and function of cardiovascular system. Here, we review the current knowledge concerning the roles of miRNAs in cardiovascular diseases with more details on cardiac remodeling, arrhythmias, and atherosclerosis. In addition, we discuss the latest findings on the potential therapeutic applications of miRNAs in cardiovascular diseases.
Collapse
Affiliation(s)
- Fatemeh Mirzadeh Azad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maedeh Arabian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Malakootian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
97
|
Shati AA. Doxorubicin-induces NFAT/Fas/FasL cardiac apoptosis in rats through activation of calcineurin and P38 MAPK and inhibition of mTOR signalling pathways. Clin Exp Pharmacol Physiol 2020; 47:660-676. [PMID: 31811646 DOI: 10.1111/1440-1681.13225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/28/2019] [Accepted: 12/02/2019] [Indexed: 01/06/2023]
Abstract
This study investigated the role of NFAT/Fas/FasL axis in cardiomyocyte apoptosis following doxorubicin (DOX) treatment in rats and evaluated the involvement and regulation of all NFAT members in cardiac apoptosis. Forty adult male Wistar rats were divided equally into control or DOX-treated groups (15 mg/kg over 2 weeks). Cardiomyocytes were cultured and pre-incubated with various inhibitors and activators (10 μmol/L) prior to DOX exposure (1 μmol/L). In the left ventricles and cultured cells, DOX increased cytoplasmic protein levels of cytochrome C, Bax and increased the activities of caspase-8, caspase3, ERK1/2, JNK, and P38 mitogen-activated protein kinases (MAPKs), reducing levels of Bcl-2 and the activity of mTOR, and inducing cell death. In addition, DOX enhanced mRNA and protein levels of Fas and FasL. Furthermore, the nuclear and cytoplasmic levels of NFAT1 and nuclear accumulation of NFAT2-4were increased with DOX treatment. The inhibition of calcineurin with FK506 significantly inhibited the nuclear levels of NFAT2 and NFAT4 and the inhibition of P38 MAPK with SB203580 inhibited the nuclear and cytoplasmic accumulation of NFAT1. However, the activation of mTOR by IGF-1 significantly lowered NFAT3. In conclusion, NFAT/Fas/FasL-induced cell death in cardiac myocytes of DOX-treated rats is regulated, at least, by the activation of calcineurin and P38 MAPK and inhibition of mTOR.
Collapse
Affiliation(s)
- Ali A Shati
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
98
|
Nazarali P, Dadaei M, Alizadeh R. Effect of an 8-week endurance rehabilitation exercise on apoptosis in cardiac patients. Res Cardiovasc Med 2020. [DOI: 10.4103/rcm.rcm_25_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
99
|
Pervaiz S, Bellot GL, Lemoine A, Brenner C. Redox signaling in the pathogenesis of human disease and the regulatory role of autophagy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 352:189-214. [DOI: 10.1016/bs.ircmb.2020.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
100
|
Kar S, Shahshahan HR, Hackfort BT, Yadav SK, Yadav R, Kambis TN, Lefer DJ, Mishra PK. Exercise Training Promotes Cardiac Hydrogen Sulfide Biosynthesis and Mitigates Pyroptosis to Prevent High-Fat Diet-Induced Diabetic Cardiomyopathy. Antioxidants (Basel) 2019; 8:antiox8120638. [PMID: 31835893 PMCID: PMC6943713 DOI: 10.3390/antiox8120638] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 12/30/2022] Open
Abstract
Obesity increases the risk of developing diabetes and subsequently, diabetic cardiomyopathy (DMCM). Reduced cardioprotective antioxidant hydrogen sulfide (H2S) and increased inflammatory cell death via pyroptosis contribute to adverse cardiac remodeling and DMCM. Although exercise training (EX) has cardioprotective effects, it is unclear whether EX mitigates obesity-induced DMCM by increasing H₂S biosynthesis and mitigating pyroptosis in the heart. C57BL6 mice were fed a high-fat diet (HFD) while undergoing treadmill EX for 20 weeks. HFD mice developed obesity, hyperglycemia, and insulin resistance, which were reduced by EX. Left ventricle pressure-volume measurement revealed that obese mice developed reduced diastolic function with preserved ejection fraction, which was improved by EX. Cardiac dysfunction was accompanied by increased cardiac pyroptosis signaling, structural remodeling, and metabolic remodeling, indicated by accumulation of lipid droplets in the heart. Notably, EX increased cardiac H₂S concentration and expression of H₂S biosynthesis enzymes. HFD-induced obesity led to features of type 2 diabetes (T2DM), and subsequently DMCM. EX during the HFD regimen prevented the development of DMCM, possibly by promoting H₂S-mediated cardioprotection and alleviating pyroptosis. This is the first report of EX modulating H₂S and pyroptotic signaling in the heart.
Collapse
Affiliation(s)
- Sumit Kar
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (H.R.S.); (B.T.H.); (S.K.Y.); (R.Y.); (T.N.K.)
| | - Hamid R. Shahshahan
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (H.R.S.); (B.T.H.); (S.K.Y.); (R.Y.); (T.N.K.)
| | - Bryan T. Hackfort
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (H.R.S.); (B.T.H.); (S.K.Y.); (R.Y.); (T.N.K.)
| | - Santosh K. Yadav
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (H.R.S.); (B.T.H.); (S.K.Y.); (R.Y.); (T.N.K.)
| | - Roopali Yadav
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (H.R.S.); (B.T.H.); (S.K.Y.); (R.Y.); (T.N.K.)
| | - Tyler N. Kambis
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (H.R.S.); (B.T.H.); (S.K.Y.); (R.Y.); (T.N.K.)
| | - David J. Lefer
- Department of Pharmacology and Experimental Therapeutics, Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Paras K. Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (H.R.S.); (B.T.H.); (S.K.Y.); (R.Y.); (T.N.K.)
- Correspondence: ; Tel.: +1-402-559-8524; Fax: +1-402-559-4438
| |
Collapse
|