1
|
Li YY, Chen HR, Yang Y, Pan YJ, Yuan QC, Liu YZ. Murine exosomal miR-30a aggravates cardiac function after acute myocardial infarction via regulating cell fate of cardiomyocytes and cardiac resident macrophages. Int J Cardiol 2024; 414:132395. [PMID: 39074620 DOI: 10.1016/j.ijcard.2024.132395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/02/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
After acute myocardial infarction (AMI), intercellular communication is crucial for maintaining cardiac homeostasis and patient survival. Exosomes secreted by cardiomyocytes serve as carriers for transporting microRNA(miRNAs), participating in intercellular signaling and the regulation of cardiac function. This study aims to investigate the role of exosomal microRNA-30a(miR-30a) during AMI and its underlying mechanisms. AMI was induced by permanent ligation of the left anterior descending (LAD) artery in C57BL/6 mice. The expression of miR-30a in mice was respectively enhanced and inhibited by administering agomiR-30a and antagomiR-30a. Using HL-1 cardiomyocytes and RAW264.7 macrophages for in vitro experiments, HL-1 cardiomyocytes were cultured under hypoxic conditions to induce ischemic injury. Following isolation and injection of exosomals, a variety of validation methods were utilized to assess the expression of miR-30a, and investigate the effects of enriched exosomal miR-30a on the state of cardiomyocytes. After AMI, the level of exosomal miR-30a in the serum of mice significantly increased and was highly enriched in cardiac tissue. Cardiomyocytes treated with agomiR-30a and miR-30a-enriched exosomes exhibited inhibition of cell autophagy, increased cell apoptosis, mice showed an larger myocardial infarct area and poorer cardiac function. Exosomes released from hypoxic cardiomyocytes transferred miR-30a to cardiac resident macrophages, promoting the polarization into pro-inflammatory M1 macrophages. In conclusion, murine exosomal miR-30a exacerbates cardiac dysfunction post-AMI by disrupting the autophagy-apoptosis balance in cardiomyocytes and polarizing cardiac resident macrophages into pro-inflammatory M1 macrophages. Modulating the expression of miR-30a may reduce cardiac damage following AMI, and targeting exosomal miR-30a could be a potential therapeutic approach for AMI.
Collapse
Affiliation(s)
- Ying-Ying Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Hong-Rui Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yan Yang
- Department of General, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ya-Jie Pan
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qing-Chen Yuan
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Yu-Zhou Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
2
|
Kang MC, Deutz NEP, Kirschner SK, Engelen MPKJ. Metabolic kinetics and muscle and brain health markers in older adults, and the role of age and presence of chronic morbidities: A large cross-sectional cohort study. Clin Nutr 2024; 43:36-47. [PMID: 39423760 DOI: 10.1016/j.clnu.2024.10.015] [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: 02/12/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND & AIMS Older adults are at risk for muscle and cognitive function decline during advanced aging, but the underlying metabolic mechanisms and the role of aging-associated chronic morbidities remain unclear. In the present study, we examined whether protein and amino acid kinetics in older adults with and without chronic morbidities are different when 50-70 and 70-90 of age and related to markers of muscle and brain health declines. METHODS In a large cross-sectional observational study, 575 older adults from 12 trials (2014-2022) were stratified based on their age (50-70y vs. 70-95y) and the presence of chronic morbidities. The main outcomes were whole-body production (WBP) and interconversions of amino acids by stable amino acid tracers, body composition, and muscle and cognitive performance. Additionally, the association between metabolic markers and muscle and brain health was assessed. RESULTS Overall lower muscle strength, muscle and fat mass, and cognitive function (p < 0.03), but no mood disturbances, were found in 70-95y compared to 50-70y older adults. Presence of morbidities was associated with lower muscle strength and mass, and cognitive function, but higher visceral adipose tissue, and mood disturbances (p < 0.05). Aging was associated with suppressed WBP of most amino acids, de novo arginine production, and net protein breakdown, but higher myofibrillar protein breakdown (p < 0.007). Presence of morbidities was associated with lower WBP of glutamine, glutamate, histidine, isoleucine, phenylalanine, tyrosine, and net protein breakdown, and higher WBP of valine and taurine (p < 0.04). Age showed significant negative correlations with WBP of nearly all amino acids, de novo arginine production and net protein breakdown (r: [-0.407, -0.136], p < 0.01) but a positive correlation with WBP of myofibrillar protein breakdown (r = 0.133, p = 0.009). Lean mass showed positive correlations with de novo arginine production and net protein breakdown and WBP of all amino acids except for isoleucine (r: [0.16, 0.799], p < 0.005). MoCA showed a positive correlation with WBP of leucine and valine (r: [0.163, 0.2], p < 0.03). Worse cognitive performance was positively associated with WBP of tau-methylhistidine and taurine (r: [0.13, 0.141], p < 0.04), but negatively associated with WBP of glycine and valine, de novo arginine production, and net protein breakdown (r: [-0.222, -0.115], p < 0.05). CONCLUSION Comprehensive phenotyping of a large group of older adults revealed differences in metabolic health in response to advanced aging and chronic morbidities. Poor muscle health accompanied by advanced aging was associated with overall metabolic downregulation, except for enhanced myofibrillar (muscle) protein breakdown. Presence of chronic morbidities was further associated with disturbed muscle health, mood, arginine, and taurine pathways, and higher visceral adipose tissue. Therefore, different phenotypes among older adults need to be considered when evaluating therapeutic approaches to improve muscle and brain health.
Collapse
Affiliation(s)
- Minchae C Kang
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX, USA
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX, USA
| | - Sarah K Kirschner
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX, USA
| | - Mariëlle P K J Engelen
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
3
|
Singh A, Ravendranathan N, Frisbee JC, Singh KK. Complex Interplay between DNA Damage and Autophagy in Disease and Therapy. Biomolecules 2024; 14:922. [PMID: 39199310 PMCID: PMC11352539 DOI: 10.3390/biom14080922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
Cancer, a multifactorial disease characterized by uncontrolled cellular proliferation, remains a global health challenge with significant morbidity and mortality. Genomic and molecular aberrations, coupled with environmental factors, contribute to its heterogeneity and complexity. Chemotherapeutic agents like doxorubicin (Dox) have shown efficacy against various cancers but are hindered by dose-dependent cytotoxicity, particularly on vital organs like the heart and brain. Autophagy, a cellular process involved in self-degradation and recycling, emerges as a promising therapeutic target in cancer therapy and neurodegenerative diseases. Dysregulation of autophagy contributes to cancer progression and drug resistance, while its modulation holds the potential to enhance treatment outcomes and mitigate adverse effects. Additionally, emerging evidence suggests a potential link between autophagy, DNA damage, and caretaker breast cancer genes BRCA1/2, highlighting the interplay between DNA repair mechanisms and cellular homeostasis. This review explores the intricate relationship between cancer, Dox-induced cytotoxicity, autophagy modulation, and the potential implications of autophagy in DNA damage repair pathways, particularly in the context of BRCA1/2 mutations.
Collapse
Affiliation(s)
- Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond Street North, London, ON N6A 5C1, Canada; (A.S.); (N.R.); (J.C.F.)
| | - Naresh Ravendranathan
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond Street North, London, ON N6A 5C1, Canada; (A.S.); (N.R.); (J.C.F.)
| | - Jefferson C. Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond Street North, London, ON N6A 5C1, Canada; (A.S.); (N.R.); (J.C.F.)
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond Street North, London, ON N6A 5C1, Canada; (A.S.); (N.R.); (J.C.F.)
- Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
4
|
Sato R, Vatic M, Peixoto da Fonseca GW, Anker SD, von Haehling S. Biological basis and treatment of frailty and sarcopenia. Cardiovasc Res 2024:cvae073. [PMID: 38828887 DOI: 10.1093/cvr/cvae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/23/2022] [Accepted: 12/20/2022] [Indexed: 06/05/2024] Open
Abstract
In an ageing society, the importance of maintaining healthy life expectancy has been emphasized. As a result of age-related decline in functional reserve, frailty is a state of increased vulnerability and susceptibility to adverse health outcomes with a serious impact on healthy life expectancy. The decline in skeletal muscle mass and function, also known as sarcopenia, is key in the development of physical frailty. Both frailty and sarcopenia are highly prevalent in patients not only with advanced age but also in patients with illnesses that exacerbate their progression like heart failure (HF), cancer, or dementia, with the prevalence of frailty and sarcopenia in HF patients reaching up to 50-75% and 19.5-47.3%, respectively, resulting in 1.5-3 times higher 1-year mortality. The biological mechanisms of frailty and sarcopenia are multifactorial, complex, and not yet fully elucidated, ranging from DNA damage, proteostasis impairment, and epigenetic changes to mitochondrial dysfunction, cellular senescence, and environmental factors, many of which are further linked to cardiac disease. Currently, there is no gold standard for the treatment of frailty and sarcopenia, however, growing evidence supports that a combination of exercise training and nutritional supplement improves skeletal muscle function and frailty, with a variety of other therapies being devised based on the underlying pathophysiology. In this review, we address the involvement of frailty and sarcopenia in cardiac disease and describe the latest insights into their biological mechanisms as well as the potential for intervention through exercise, diet, and specific therapies.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Mirela Vatic
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Guilherme Wesley Peixoto da Fonseca
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Stefan D Anker
- Department of Cardiology (CVK) of German Heart Center Charité; German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| |
Collapse
|
5
|
Zou R, Shi W, Ceylan AF, Dong M, Zhang M, Zou Z, Peng B, Dong F, Turdi S, Lin J, Zhang Y, Wang G, Fan X, Ren J. Cardiomyocyte-specific deletion of endothelin receptor A (ET A) obliterates cardiac aging through regulation of mitophagy and ferroptosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166958. [PMID: 37963542 DOI: 10.1016/j.bbadis.2023.166958] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/23/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023]
Abstract
Advanced aging evokes unfavorable changes in the heart including cardiac remodeling and contractile dysfunction although the underlying mechanism remains elusive. This study was conducted to evaluate the role of endothelin-1 (ET-1) in the pathogenesis of cardiac aging and mechanism involved. Echocardiographic and cardiomyocyte mechanical properties were determined in young (5-6 mo) and aged (26-28 mo) wild-type (WT) and cardiomyocyte-specific ETA receptor knockout (ETAKO) mice. GSEA enrichment identified differentially expressed genes associated with mitochondrial respiration, mitochondrial protein processing and mitochondrial depolarization in cardiac aging. Aging elevated plasma levels of ET-1, Ang II and suppressed serum Fe2+, evoked cardiac remodeling (hypertrophy and interstitial fibrosis), contractile defects (fractional shortening, ejection fraction, cardiomyocyte peak shortening, maximal velocity of shortening/relengthening and prolonged relengthening) and intracellular Ca2+ mishandling (dampened intracellular Ca2+ release and prolonged decay), the effects with the exception of plasma AngII, ET-1 and Fe2+ were mitigated by ETAKO. Advanced age facilitated O2- production, carbonyl protein damage, cardiac hypertrophy (GATA4, ANP, NFATc3), ER stress, ferroptosis, compromised autophagy (LC3B, Beclin-1, Atg7, Atg5 and p62) and mitophagy (parkin and FUNDC1), and deranged intracellular Ca2+ proteins (SERCA2a and phospholamban), the effects of which were reversed by ETA ablation. ET-1 provoked ferroptosis in vitro, the response was nullified by the ETA receptor antagonist BQ123 and mitophagy inducer CsA. ETA but not ETB receptor antagonism reconciled cardiac aging, which was abrogated by inhibition of mitophagy and ferroptosis. These findings collectively denote promises of targeting ETA, mitophagy and ferroptosis in the management of aging-associated cardiac remodeling and contractile defect.
Collapse
Affiliation(s)
- Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, China
| | - Wanting Shi
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Asli F Ceylan
- Ankara Yildirim Beyazit University, Faculty of Medicine, Department of Medical Pharmacology, Bilkent, Ankara, Turkey
| | - Maolong Dong
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Miao Zhang
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Zengxiao Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Bo Peng
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Subat Turdi
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jie Lin
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Ge Wang
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, China.
| | - Xiaoping Fan
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
6
|
Xiao S, Peng K, Li C, Long Y, Yu Q. The role of sphingosine-1-phosphate in autophagy and related disorders. Cell Death Discov 2023; 9:380. [PMID: 37852968 PMCID: PMC10584985 DOI: 10.1038/s41420-023-01681-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/03/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023] Open
Abstract
S1P, also referred to as sphingosine-1-phosphate, is a lipid molecule with bioactive properties involved in numerous cellular processes such as cell growth, movement, programmed cell death, self-degradation, cell specialization, aging, and immune system reactions. Autophagy is a meticulously controlled mechanism in which cells repurpose their elements to maintain cellular balance. There are five stages in autophagy: initiation, nucleation, elongation and maturation, fusion, and degradation. New research has provided insight into the complex connection between S1P and autophagy, uncovering their interaction in both normal and abnormal circumstances. Gaining knowledge about the regulatory mechanism of S1P signaling on autophagy can offer a valuable understanding of its function in well-being and illness, potentially leading to innovative therapeutic concepts for diverse ailments. Hence, this review analyzes the essential stages in mammalian autophagy, with a specific emphasis on recent research exploring the control of each stage by S1P. Additionally, it sheds light on the roles of S1P-induced autophagy in various disorders.
Collapse
Affiliation(s)
- Siqi Xiao
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Kaixin Peng
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Congxin Li
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Yuanyuan Long
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Qin Yu
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China.
| |
Collapse
|
7
|
Pandey AK, Bhatt DL, Pandey A, Marx N, Cosentino F, Pandey A, Verma S. Mechanisms of benefits of sodium-glucose cotransporter 2 inhibitors in heart failure with preserved ejection fraction. Eur Heart J 2023; 44:3640-3651. [PMID: 37674356 DOI: 10.1093/eurheartj/ehad389] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/07/2023] [Accepted: 05/29/2023] [Indexed: 09/08/2023] Open
Abstract
For decades, heart failure with preserved ejection fraction (HFpEF) proved an elusive entity to treat. Sodium-glucose cotransporter 2 (SGLT2) inhibitors have recently been shown to reduce the composite of heart failure hospitalization or cardiovascular death in patients with HFpEF in the landmark DELIVER and EMPEROR-Preserved trials. While improvements in blood sugar, blood pressure, and attenuation of kidney disease progression all may play some role, preclinical and translational research have identified additional mechanisms of these agents. The SGLT2 inhibitors have intriguingly been shown to induce a nutrient-deprivation and hypoxic-like transcriptional paradigm, with increased ketosis, erythropoietin, and autophagic flux in addition to altering iron homeostasis, which may contribute to improved cardiac energetics and function. These agents also reduce epicardial adipose tissue and alter adipokine signalling, which may play a role in the reductions in inflammation and oxidative stress observed with SGLT2 inhibition. Emerging evidence also indicates that these drugs impact cardiomyocyte ionic homeostasis although whether this is through indirect mechanisms or via direct, off-target effects on other ion channels has yet to be clearly characterized. Finally, SGLT2 inhibitors have been shown to reduce myofilament stiffness as well as extracellular matrix remodelling/fibrosis in the heart, improving diastolic function. The SGLT2 inhibitors have established themselves as robust, disease-modifying therapies and as recent trial results are incorporated into clinical guidelines, will likely become foundational in the therapy of HFpEF.
Collapse
Affiliation(s)
- Arjun K Pandey
- Michael G. DeGroote School of Medicine, McMaster University, 90 Main Street West, Hamilton, Ontario L8P 1H6, Canada
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Avinash Pandey
- Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, Ontario K1Y 4W7, Canada
| | - Nikolaus Marx
- Department of Internal Medicine, University Hospital Aachen, RWTH Aachen University, Templergraben 55, 52062 Aachen, Germany
| | - Francesco Cosentino
- Division of Cardiology, Department of Medicine, Solna, Karolinska Institutet, Norrbacka S1:02, Stockholm, SE 17177, Sweden
- Heart, Vascular and Neuro Theme, Department of Cardiology, Karolinska University Hospital, Anna Steckséns gata 41, 171 64 Solna, Sweden
| | - Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Subodh Verma
- Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, 30 Bond Street, Toronto, ON, Canada
| |
Collapse
|
8
|
Zhi X, Shi S, Li Y, Ma M, Long Y, Li C, Hao H, Liu H, Wang X, Wang L. S100a9 inhibits Atg9a transcription and participates in suppression of autophagy in cardiomyocytes induced by β 1-adrenoceptor autoantibodies. Cell Mol Biol Lett 2023; 28:74. [PMID: 37723445 PMCID: PMC10506287 DOI: 10.1186/s11658-023-00486-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/31/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Cardiomyocyte death induced by autophagy inhibition is an important cause of cardiac dysfunction. In-depth exploration of its mechanism may help to improve cardiac dysfunction. In our previous study, we found that β1-adrenergic receptor autoantibodies (β1-AAs) induced a decrease in myocardial autophagy and caused cardiomyocyte death, thus resulting in cardiac dysfunction. Through tandem mass tag (TMT)-based quantitative proteomics, autophagy-related S100a9 protein was found to be significantly upregulated in the myocardial tissue of actively immunized mice. However, whether S100a9 affects the cardiac function in the presence of β1-AAs through autophagy and the specific mechanism are currently unclear. METHODS In this study, the active immunity method was used to establish a β1-AA-induced mouse cardiac dysfunction model, and RT-PCR and western blot were used to detect changes in gene and protein expression in cardiomyocytes. We used siRNA to knockdown S100a9 in cardiomyocytes. An autophagy PCR array was performed to screen differentially expressed autophagy-related genes in cells transfected with S100a9 siRNA and negative control siRNA. Cytoplasmic nuclear separation, co-immunoprecipitation (Co-IP), and immunofluorescence were used to detect the binding of S100a9 and hypoxia inducible factor-1α (HIF-1α). Finally, AAV9-S100a9-RNAi was injected into mice via the tail vein to knockdown S100a9 in cardiomyocytes. Cardiac function was detected via ultrasonography. RESULTS The results showed that β1-AAs induced S100a9 expression. The PCR array indicated that Atg9a changed significantly in S100a9siRNA cells and that β1-AAs increased the binding of S100a9 and HIF-1α in cytoplasm. Knockdown of S100a9 significantly improved autophagy levels and cardiac dysfunction. CONCLUSION Our research showed that β1-AAs increased S100a9 expression in cardiomyocytes and that S100a9 interacted with HIF-1α, which prevented HIF-1α from entering the nucleus normally, thus inhibiting the transcription of Atg9a. This resulted in autophagy inhibition and cardiac dysfunction.
Collapse
Affiliation(s)
- Xiaoyan Zhi
- Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Shu Shi
- Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yang Li
- Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Mingxia Ma
- Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yaolin Long
- Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Chen Li
- Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Haihu Hao
- Department of Orthopaedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xiaohui Wang
- Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi, 030001, People's Republic of China.
| |
Collapse
|
9
|
Korover N, Etzion S, Cherniak A, Rabinski T, Levitas A, Etzion Y, Ofir R, Parvari R, Cohen S. Functional defects in hiPSCs-derived cardiomyocytes from patients with a PLEKHM2-mutation associated with dilated cardiomyopathy and left ventricular non-compaction. Biol Res 2023; 56:34. [PMID: 37349842 PMCID: PMC10288792 DOI: 10.1186/s40659-023-00442-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/17/2023] [Indexed: 06/24/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a primary myocardial disease, leading to heart failure and excessive risk of sudden cardiac death with rather poorly understood pathophysiology. In 2015, Parvari's group identified a recessive mutation in the autophagy regulator, PLEKHM2 gene, in a family with severe recessive DCM and left ventricular non-compaction (LVNC). Fibroblasts isolated from these patients exhibited abnormal subcellular distribution of endosomes, Golgi apparatus, lysosomes and had impaired autophagy flux. To better understand the effect of mutated PLEKHM2 on cardiac tissue, we generated and characterized induced pluripotent stem cells-derived cardiomyocytes (iPSC-CMs) from two patients and a healthy control from the same family. The patient iPSC-CMs showed low expression levels of genes encoding for contractile functional proteins (α and β-myosin heavy chains and 2v and 2a-myosin light chains), structural proteins integral to heart contraction (Troponin C, T and I) and proteins participating in Ca2+ pumping action (SERCA2 and Calsequestrin 2) compared to their levels in control iPSC-derived CMs. Furthermore, the sarcomeres of the patient iPSC-CMs were less oriented and aligned compared to control cells and generated slowly beating foci with lower intracellular calcium amplitude and abnormal calcium transient kinetics, measured by IonOptix system and MuscleMotion software. Autophagy in patient's iPSC-CMs was impaired as determined from a decrease in the accumulation of autophagosomes in response to chloroquine and rapamycin treatment, compared to control iPSC-CMs. Impairment in autophagy together with the deficiency in the expression of NKX2.5, MHC, MLC, Troponins and CASQ2 genes, which are related to contraction-relaxation coupling and intracellular Ca2+ signaling, may contribute to the defective function of the patient CMs and possibly affect cell maturation and cardiac failure with time.
Collapse
Affiliation(s)
- Nataly Korover
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel.
| | - Sharon Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Alexander Cherniak
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Tatiana Rabinski
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Aviva Levitas
- Department of Pediatric Cardiology, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Yoram Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Rivka Ofir
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Dead Sea & Arava Science Center, 8691000, Masada, Israel
| | - Ruti Parvari
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Smadar Cohen
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| |
Collapse
|
10
|
Jiang B, Zhou X, Yang T, Wang L, Feng L, Wang Z, Xu J, Jing W, Wang T, Su H, Yang G, Zhang Z. The role of autophagy in cardiovascular disease: Cross-interference of signaling pathways and underlying therapeutic targets. Front Cardiovasc Med 2023; 10:1088575. [PMID: 37063954 PMCID: PMC10090687 DOI: 10.3389/fcvm.2023.1088575] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Autophagy is a conserved lysosomal pathway for the degradation of cytoplasmic proteins and organelles, which realizes the metabolic needs of cells and the renewal of organelles. Autophagy-related genes (ATGs) are the main molecular mechanisms controlling autophagy, and their functions can coordinate the whole autophagic process. Autophagy can also play a role in cardiovascular disease through several key signaling pathways, including PI3K/Akt/mTOR, IGF/EGF, AMPK/mTOR, MAPKs, p53, Nrf2/p62, Wnt/β-catenin and NF-κB pathways. In this paper, we reviewed the signaling pathway of cross-interference between autophagy and cardiovascular diseases, and analyzed the development status of novel cardiovascular disease treatment by targeting the core molecular mechanism of autophagy as well as the critical signaling pathway. Induction or inhibition of autophagy through molecular mechanisms and signaling pathways can provide therapeutic benefits for patients. Meanwhile, we hope to provide a unique insight into cardiovascular treatment strategies by understanding the molecular mechanism and signaling pathway of crosstalk between autophagy and cardiovascular diseases.
Collapse
Affiliation(s)
- Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xuan Zhou
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Yang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linlin Wang
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Longfei Feng
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zheng Wang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jin Xu
- Department of First Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Wang
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Haixiang Su
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - GuoWei Yang
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| | - Zheng Zhang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
11
|
Hu GF, Wang X, Meng LB, Li JY, Xu HX, Wu DS, Shan MJ, Chen YH, Xu JP, Gong T, Chen Z, Li YJ, Liu DP. SGLT1/2 as the potential biomarkers of renal damage under Apoe−/− and chronic stress via the BP neural network model and support vector machine. Front Cardiovasc Med 2022; 9:948909. [PMID: 36035950 PMCID: PMC9405420 DOI: 10.3389/fcvm.2022.948909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundChronic stress (CS) could produce negative emotions. The molecular mechanism of SGLT1 and SGLT2 in kidney injury caused by chronic stress combined with atherosclerosis remains unclear.MethodsIn total, 60 C57BL/6J mice were randomly divided into four groups, namely, control (CON, n = 15), control diet + chronic stress (CON+CS, n = 15), high-fat diet + Apoe−/− (HF + Apoe−/−, n = 15), and high-fat diet + Apoe−/− + chronic stress (HF+Apoe−/− + CS, n = 15) groups. The elevated plus maze and open field tests were performed to examine the effect of chronic stress. The expression of SGLT1 and SGLT2 in the kidney was detected. The support vector machine (SVM) and back propagation (BP) neural network model were constructed to explore the predictive value of the expression of SGLT1/2 on the renal pathological changes. The receiver operating characteristic (ROC) curve analysis was used.ResultsA chronic stress model and atherosclerosis model were constructed successfully. Edema, broken reticular fiber, and increased glycogen in the kidney would be obvious in the HF + Apoe−/− + CS group. Compared with the CON group, the expression of SGLT1/2 in the kidney was upregulated in the HF + Apoe−/− + CS group (P < 0.05). There existed positive correlations among edema, glycogen, reticular fiber, expression of SGLT1/2 in the kidney. There were higher sensitivity and specificity of diagnosis of SGLT1/2 for edema, reticular fiber, and glycogen in the kidney. The result of the SVM and BP neural network model showed better predictive values of SGLT1 and SGLT2 for edema and glycogen in the kidney.ConclusionIn conclusion, SGLT1/2 might be potential biomarkers of renal damage under Apoe−/− and chronic stress, which provided a potential research direction for future related explorations into this mechanism.
Collapse
Affiliation(s)
- Gai-feng Hu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiang Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ling-bing Meng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-yi Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-xuan Xu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Di-shan Wu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng-jie Shan
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yu-hui Chen
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jia-pei Xu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Gong
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Tao Gong
| | - Zuoguan Chen
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Zuoguan Chen
| | - Yong-jun Li
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Yong-jun Li
| | - De-ping Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: De-ping Liu
| |
Collapse
|
12
|
Induction of apoptosis and autosis in cardiomyocytes by the combination of homocysteine and copper via NOX-mediated p62 expression. Cell Death Dis 2022; 8:75. [PMID: 35190552 PMCID: PMC8860999 DOI: 10.1038/s41420-022-00870-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/12/2022] [Accepted: 02/01/2022] [Indexed: 01/01/2023]
Abstract
High levels of homocysteine (Hcy) associated with cardiovascular events are accompanied by increased copper (Cu) concentrations in the blood. Hcy has been shown to promote endothelial dysfunction, whereas the effect of Hcy on cardiomyocytes and the role of Cu in the pathogenesis remain less understood. In the present study, it is demonstrated that the combination of Hcy and Cu2+-induced apoptosis and autosis of cardiomyocytes simultaneously, and thus led to cardiac dysfunction in hyperhomocysteinemic rats. These effects were associated with p22phox activation and NADPH oxidase (NOX)-mediated p62 upregulation. Inhibition of the expression of p22phox or p62 in cardiomyocytes significantly attenuated Hcy and Cu2+-mediated reactive oxygen species (ROS) generation and cell death. Furthermore, interrupting the NOX-p62 axis prevented diastolic dysfunction in hyperhomocysteinemic rats (HcyR). These findings establish that the induction of apoptosis and autosis of cardiomyocytes through stimulating the NOX-p62-signaling pathway constitutes a novel mechanism of Hcy and Cu-induced cardiac dysfunction.
Collapse
|
13
|
Liao M, Xie Q, Zhao Y, Yang C, Lin C, Wang G, Liu B, Zhu L. Main active components of Si-Miao-Yong-An decoction (SMYAD) attenuate autophagy and apoptosis via the PDE5A-AKT and TLR4-NOX4 pathways in isoproterenol (ISO)-induced heart failure models. Pharmacol Res 2022; 176:106077. [PMID: 35026404 DOI: 10.1016/j.phrs.2022.106077] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/24/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023]
Abstract
Heart failure (HF), the main cause of death in patients with many cardiovascular diseases, has been reported to be closely related to the complicated pathogenesis of autophagy, apoptosis, and inflammation. Notably, Si-Miao-Yong-An decoction (SMYAD) is a traditional Chinese medicine (TCM) used to treat cardiovascular disease; however, the main active components and their relevant mechanisms remain to be discovered. Based on our previous ultra-performance liquid chromatography coupled to quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF-MS) results, we identified angoriside C (AC) and 3,5-dicaffeoylquinic acid (3,5-DiCQA) as the main active components of SMYAD. In vivo results showed that AC and 3,5-DiCQA effectively improved cardiac function, reduced the fibrotic area, and alleviated isoproterenol (ISO)-induced myocarditis in rats. Moreover, AC and 3,5-DiCQA inhibited ISO-induced autophagic cell death by inhibiting the PDE5A/AKT/mTOR/ULK1 pathway and inhibited ISO-induced apoptosis by inhibiting the TLR4/NOX4/BAX pathway. In addition, the autophagy inhibitor 3-MA was shown to reduce ISO-induced apoptosis, indicating that ISO-induced autophagic cell death leads to excess apoptosis. Taken together, the main active components AC and 3,5-DiCQA of SMYAD inhibit the excessive autophagic cell death and apoptosis induced by ISO by inhibiting the PDE5A-AKT and TLR4-NOX4 pathways, thereby reducing myocardial inflammation and improving heart function to alleviate and treat a rat ISO-induced heart failure model and cell heart failure models. More importantly, the main active components of SMYAD will provide new insights into a promising strategy that will promote the discovery of more main active components of SMYAD for therapeutic purposes in the future.
Collapse
Affiliation(s)
- Minru Liao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuqian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; School of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chengcan Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lingjuan Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; School of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
14
|
Autophagy facilitates age-related cell apoptosis-a new insight from senile cataract. Cell Death Dis 2022; 13:37. [PMID: 35013122 PMCID: PMC8748728 DOI: 10.1038/s41419-021-04489-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/23/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022]
Abstract
Age-related cell loss underpins many senescence-associated diseases. Apoptosis of lens epithelial cells (LECs) is the important cellular basis of senile cataract resulted from prolonged exposure to oxidative stress, although the specific mechanisms remain elusive. Our data indicated the concomitance of high autophagy activity, low SQSTM1/p62 protein level and apoptosis in the same LEC from senile cataract patients. Meanwhile, in primary cultured LECs model, more durable autophagy activation and more obvious p62 degradation under oxidative stress were observed in LECs from elder healthy donors, compared with that from young healthy donors. Using autophagy-deficiency HLE-B3 cell line, autophagy adaptor p62 was identified as the critical scaffold protein sustaining the pro-survival signaling PKCι-IKK-NF-κB cascades, which antagonized the pro-apoptotic signaling. Moreover, the pharmacological inhibitor of autophagy, 3-MA, significantly inhibited p62 degradation and rescued oxidative stress-induced apoptosis in elder LECs. Collectively, this study demonstrated that durable activation of autophagy promoted age-related cell death in LECs. Our work contributes to better understanding the pathogenesis of senescence-associated diseases.
Collapse
|
15
|
Ala M, Eftekhar SP. Target Sestrin2 to Rescue the Damaged Organ: Mechanistic Insight into Its Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8790369. [PMID: 34765085 PMCID: PMC8577929 DOI: 10.1155/2021/8790369] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022]
Abstract
Sestrin2 is a stress-inducible metabolic regulator and a conserved antioxidant protein which has been implicated in the pathogenesis of several diseases. Sestrin2 can protect against atherosclerosis, heart failure, hypertension, myocardial infarction, stroke, spinal cord injury neurodegeneration, nonalcoholic fatty liver disease (NAFLD), liver fibrosis, acute kidney injury (AKI), chronic kidney disease (CKD), and pulmonary inflammation. Oxidative stress and cellular damage signals can alter the expression of Sestrin2 to compensate for organ damage. Different stress signals such as those mediated by P53, Nrf2/ARE, HIF-1α, NF-κB, JNK/c-Jun, and TGF-β/Smad signaling pathways can induce Sestrin2 expression. Subsequently, Sestrin2 activates Nrf2 and AMPK. Furthermore, Sestrin2 is a major negative regulator of mTORC1. Sestrin2 indirectly regulates the expression of several genes and reprograms intracellular signaling pathways to attenuate oxidative stress and modulate a large number of cellular events such as protein synthesis, cell energy homeostasis, mitochondrial biogenesis, autophagy, mitophagy, endoplasmic reticulum (ER) stress, apoptosis, fibrogenesis, and lipogenesis. Sestrin2 vigorously enhances M2 macrophage polarization, attenuates inflammation, and prevents cell death. These alterations in molecular and cellular levels improve the clinical presentation of several diseases. This review will shed light on the beneficial effects of Sestrin2 on several diseases with an emphasis on underlying pathophysiological effects.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Seyed Parsa Eftekhar
- Student Research Committee, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
16
|
Sun C, Lu J, Long Y, Guo S, Jia W, Ning N, Hao H, Wang X, Bian Y, Liu H, Wang L. Adiponectin up-regulates the decrease of myocardial autophagic flux induced by β 1 -adrenergic receptor autoantibody partly dependent on AMPK. J Cell Mol Med 2021; 25:8464-8478. [PMID: 34322993 PMCID: PMC8419161 DOI: 10.1111/jcmm.16807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiomyocytes autophagy is essential for maintaining cardiac function. Our previous studies have found that β1‐adrenergic receptor autoantibody (β1‐AA) induced the decreased myocardial autophagic flux, which resulted in cardiomyocyte death and cardiac dysfunction. And other studies demonstrated that β1‐AA induced the decrease of AMPK phosphorylation, the key hub of autophagy pathway, while adiponectin up‐regulated autophagic flux mediated by AMPK. However, it is not clear whether adiponectin improves the inhibition of myocardial autophagic flux induced by β1‐AA by up‐regulating the level of AMPK phosphorylation. In this study, it has been confirmed that β1‐AA induced the decrease of AMPK phosphorylation level in both vivo and vitro. Moreover, pretreatment of cardiomyocytes with AMPK inhibitor Compound C could further reduce the autophagic flux induced by β1‐AA. Adiponectin deficiency could aggravate the decrease of myocardial AMPK phosphorylation level, autophagic flux and cardiac function induced by β1‐AA. Further, exogenous adiponectin could reverse the decline of AMPK phosphorylation level and autophagic flux induced by β1‐AA and even reduce cardiomyocyte death. While pretreated with the Compound C, the adiponectin treatment did not improve the decreased autophagosome formation, but still improved the decreased autophagosome clearance induced by β1‐AA in cardiomyocytes. This study is the first time to confirm that β1‐AA could inhibit myocardial autophagic flux by down‐regulating AMPK phosphorylation level. Adiponectin could improve the inhibition of myocardial autophagic flux induced by β1‐AA partly dependent on AMPK, so as to provide an experimental basis for the treatment of patients with β1‐AA‐positive cardiac dysfunction.
Collapse
Affiliation(s)
- Cong Sun
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China.,Department of Pathology, Linfen Central Hospital, Linfen, China
| | - Jiebei Lu
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Yaolin Long
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Shuai Guo
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Weiwei Jia
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Na Ning
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Haihu Hao
- Department of Orthopedics, Shanxi Bethune Hospital & Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiaohui Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| | - Yunfei Bian
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| |
Collapse
|
17
|
Sophoricoside ameliorates cardiac hypertrophy by activating AMPK/mTORC1-mediated autophagy. Biosci Rep 2021; 40:226492. [PMID: 32964914 PMCID: PMC7677750 DOI: 10.1042/bsr20200661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/20/2020] [Accepted: 08/03/2020] [Indexed: 02/01/2023] Open
Abstract
Aim: The study aims to evaluate protective effects of sophoricoside (Sop) on cardiac hypertrophy. Meanwhile, the potential and significance of Sop should be broadened and it should be considered as an attractive drug for the treatment of pathological cardiac hypertrophy and heart failure. Methods: Using the phenylephrine (PE)-induced neonatal rat cardiomyocytes (NRCMs) enlargement model, the potent protection of Sop against cardiomyocytes enlargement was evaluated. The function of Sop was validated in mice received transverse aortic coarctation (TAC) or sham surgery. At 1 week after TAC surgery, mice were treated with Sop for the following 4 weeks, the hearts were harvested after echocardiography examination. Results: Our study revealed that Sop significantly mitigated TAC-induced heart dysfunction, cardiomyocyte hypertrophy and cardiac fibrosis. Mechanistically, Sop treatment induced a remarkable activation of AMPK/mTORC1-autophagy cascade following sustained hypertrophic stimulation. Importantly, the protective effect of Sop was largely abolished by the AMPKα inhibitor Compound C, suggesting an AMPK activation-dependent manner of Sop function on suppressing pathological cardiac hypertrophy. Conclusion: Sop ameliorates cardiac hypertrophy by activating AMPK/mTORC1-mediated autophagy. Hence, Sop might be an attractive candidate for the treatment of pathological cardiac hypertrophy and heart failure.
Collapse
|
18
|
Ott C, Jung T, Brix S, John C, Betz IR, Foryst-Ludwig A, Deubel S, Kuebler WM, Grune T, Kintscher U, Grune J. Hypertrophy-Reduced Autophagy Causes Cardiac Dysfunction by Directly Impacting Cardiomyocyte Contractility. Cells 2021; 10:805. [PMID: 33916597 PMCID: PMC8065800 DOI: 10.3390/cells10040805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 12/22/2022] Open
Abstract
Cardiac remodeling and contractile dysfunction are leading causes in hypertrophy-associated heart failure (HF), increasing with a population's rising age. A hallmark of aged and diseased hearts is the accumulation of modified proteins caused by an impaired autophagy-lysosomal-pathway. Although, autophagy inducer rapamycin has been described to exert cardioprotective effects, it remains to be shown whether these effects can be attributed to improved cardiomyocyte autophagy and contractility. In vivo hypertrophy was induced by transverse aortic constriction (TAC), with mice receiving daily rapamycin injections beginning six weeks after surgery for four weeks. Echocardiographic analysis demonstrated TAC-induced HF and protein analyses showed abundance of modified proteins in TAC-hearts after 10 weeks, both reduced by rapamycin. In vitro, cardiomyocyte hypertrophy was mimicked by endothelin 1 (ET-1) and autophagy manipulated by silencing Atg5 in neonatal cardiomyocytes. ET-1 and siAtg5 decreased Atg5-Atg12 and LC3-II, increased natriuretic peptides, and decreased amplitude and early phase of contraction in cardiomyocytes, the latter two evaluated using ImageJ macro Myocyter recently developed by us. ET-1 further decreased cell contractility in control but not in siAtg5 cells. In conclusion, ET-1 decreased autophagy and cardiomyocyte contractility, in line with siAtg5-treated cells and the results of TAC-mice demonstrating a crucial role for autophagy in cardiomyocyte contractility and cardiac performance.
Collapse
Affiliation(s)
- Christiane Ott
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (T.J.); (C.J.); (S.D.); (T.G.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (T.J.); (C.J.); (S.D.); (T.G.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
| | - Sarah Brix
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitaetsmedizin, 10115 Berlin, Germany
| | - Cathleen John
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (T.J.); (C.J.); (S.D.); (T.G.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
| | - Iris R. Betz
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitaetsmedizin, 10115 Berlin, Germany
| | - Anna Foryst-Ludwig
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitaetsmedizin, 10115 Berlin, Germany
| | - Stefanie Deubel
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (T.J.); (C.J.); (S.D.); (T.G.)
| | - Wolfgang M. Kuebler
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
- Institute of Physiology, Charité-Universitaetsmedizin, 10115 Berlin, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (T.J.); (C.J.); (S.D.); (T.G.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Ulrich Kintscher
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitaetsmedizin, 10115 Berlin, Germany
| | - Jana Grune
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany; (S.B.); (I.R.B.); (A.F.-L.); (W.M.K.); (U.K.); (J.G.)
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitaetsmedizin, 10115 Berlin, Germany
- Institute of Physiology, Charité-Universitaetsmedizin, 10115 Berlin, Germany
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
19
|
How Can Malnutrition Affect Autophagy in Chronic Heart Failure? Focus and Perspectives. Int J Mol Sci 2021; 22:ijms22073332. [PMID: 33805128 PMCID: PMC8036550 DOI: 10.3390/ijms22073332] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/15/2021] [Accepted: 03/21/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic heart failure (CHF) is a disease with important clinical and socio-economic ramifications. Malnutrition and severe alteration of the protein components of the body (protein disarrangements), common conditions in CHF patients, are independent correlates of heart dysfunction, disease progression, and mortality. Autophagy, a prominent occurrence in the heart of patients with advanced CHF, is a self-digestive process that prolongs myocardial cell lifespan by the removal of cytosolic components, such as aging organelles and proteins, and recycles the constituent elements for new protein synthesis. However, in specific conditions, excessive activation of autophagy can lead to the destruction of molecules and organelles essential to cell survival, ultimately leading to organ failure and patient death. In this review, we aim to describe the experimental and clinical evidence supporting a pathophysiological role of nutrition and autophagy in the progression of CHF. The understanding of the mechanisms underlying the interplay between nutrition and autophagy may have important clinical implications by providing molecular targets for innovative therapeutic strategies in CHF patients.
Collapse
|
20
|
Yang M, Wang S, Fu S, Wu NN, Xu X, Sun S, Zhang Y, Ren J. Deletion of the E3 ubiquitin ligase, Parkin, exacerbates chronic alcohol intake-induced cardiomyopathy through an Ambra1-dependent mechanism. Br J Pharmacol 2021; 178:964-982. [PMID: 33300167 DOI: 10.1111/bph.15340] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/17/2020] [Accepted: 09/01/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Chronic alcohol consumption contributes to contractile dysfunction and unfavourable geometric changes in myocardium, accompanied by altered autophagy and disturbed mitochondrial homeostasis. The E3 ubiquitin ligase Parkin encoded by PARK2 gene maintains a fundamental role in regulating mitophagy and mitochondrial homeostasis, although little is known of its role in the aetiology of alcoholic cardiomyopathy. Here we assessed the effects of Parkin deletion in chronic alcohol-evoked cardiotoxicity. EXPERIMENTAL APPROACH Following alcohol (4%) or control diet intake for 8 weeks, adult male wild-type (WT) and PARK2 knockout (Parkin-/- ) mice were examined using echocardiography. Cardiomyocyte mechanical properties, morphology of myocardium, and mitochondrial damage were also evaluated. Autophagy and mitophagy levels were assessed by LC3B and GFP-LC3 puncta, and lysosome-dependent autophagic flux was scrutinized using GFP-mRFP-LC3 puncta and Bafilomycin A1 treatment. KEY RESULTS Chronic alcohol exposure provoked unfavourable geometric changes in myocardium and led to mitochondrial dysfunction and cardiac contractile defects, effects further exacerbated by Parkin knockout. Chronic alcohol exposure provoked autophagy and PINK1/Parkin-mediated mitophagy without affecting lysosome-dependent autophagic flux, the effects of which were diminished by Parkin deletion. Parkin adenovirus infection in neonatal rat cardiomyocytes further increased autophagy and protected against alcohol-induced myocardial injury, effects blocked by siRNA for Ambra1 (Autophagy and Beclin1 regulator 1). Immunofluorescence staining and co-immunoprecipitation assays showed interactions between Parkin and Ambra1. CONCLUSIONS AND IMPLICATIONS Parkin was essential for cardiac homeostasis in alcohol challenge, accompanied by increased autophagy/mitophagy and maintenance of mitochondrial integrity through its interaction with Ambra1.
Collapse
Affiliation(s)
- Mingjie Yang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Fudan University Zhongshan Hospital, Shanghai, China
| | - Shuyi Wang
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shouzhi Fu
- Department of ICU/Emergency Wuhan Third Hospital, Wuhan University, Wuhan, China
| | - Ne N Wu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Fudan University Zhongshan Hospital, Shanghai, China
| | - Xihui Xu
- Cytokinetics Inc, South San Francisco, California, USA
| | - Shiqun Sun
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Fudan University Zhongshan Hospital, Shanghai, China
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Fudan University Zhongshan Hospital, Shanghai, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Fudan University Zhongshan Hospital, Shanghai, China
| |
Collapse
|
21
|
Izzo C, Vitillo P, Di Pietro P, Visco V, Strianese A, Virtuoso N, Ciccarelli M, Galasso G, Carrizzo A, Vecchione C. The Role of Oxidative Stress in Cardiovascular Aging and Cardiovascular Diseases. Life (Basel) 2021; 11:60. [PMID: 33467601 PMCID: PMC7829951 DOI: 10.3390/life11010060] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Aging can be seen as process characterized by accumulation of oxidative stress induced damage. Oxidative stress derives from different endogenous and exogenous processes, all of which ultimately lead to progressive loss in tissue and organ structure and functions. The oxidative stress theory of aging expresses itself in age-related diseases. Aging is in fact a primary risk factor for many diseases and in particular for cardiovascular diseases and its derived morbidity and mortality. Here we highlight the role of oxidative stress in age-related cardiovascular aging and diseases. We take into consideration the molecular mechanisms, the structural and functional alterations, and the diseases accompanied to the cardiovascular aging process.
Collapse
Affiliation(s)
- Carmine Izzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paolo Vitillo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Valeria Visco
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Andrea Strianese
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Nicola Virtuoso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| |
Collapse
|
22
|
Chen Y, Maejima Y, Shirakabe A, Yamamoto T, Ikeda Y, Sadoshima J, Zhai P. Ser9 phosphorylation of GSK-3β promotes aging in the heart through suppression of autophagy. THE JOURNAL OF CARDIOVASCULAR AGING 2021; 1:9. [PMID: 34778891 PMCID: PMC8589323 DOI: 10.20517/jca.2021.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Glycogen synthase kinase-3β (GSK-3β) is a serine/threonine kinase and a negative regulator of cardiac hypertrophy. Phosphorylation of GSK-3β at Ser9 negatively regulates its kinase activity. The role of GSK-3β in cardiac aging remains poorly understood. AIM The study aimed to elucidate the role of GSK-3β Ser9 phosphorylation in mediating cardiac aging and the underlying mechanism. METHODS AND RESULTS Phosphorylation of GSK-3β at Ser9 and the levels of β-catenin and Mcl-1 were increased in the mouse heart during aging, suggesting that GSK-3β is inactivated during aging in the heart. Age-induced cardiac hypertrophy, fibrosis, left ventricular dysfunction, and increases in cardiomyocyte apoptosis and senescence were all attenuated in constitutively active GSK-3βS9A knock-in (KI) mice compared to littermate wild type mice. Although autophagy is inhibited in the heart during aging, KI of GSK-3βS9A reversed the age-associated decline in autophagy in the mouse heart. GSK-3β directly phosphorylates Ulk1, a regulator of autophagy, at Ser913, thereby stimulating autophagy in cardiomyocytes. Ulk1Ser913A KI mice exhibited decreased autophagic flux and increased senescence in cardiomyocytes. CONCLUSION Our results suggest that GSK-3β is inactivated during aging through Ser9 phosphorylation, which in turn plays an important role in mediating cardiac aging. GSK-3β promotes autophagy through phosphorylation of Ulk1 at Ser913, which in turn prevents aging in the heart.
Collapse
Affiliation(s)
- Yanbin Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Soochow 215000, Jiangsu, China
| | - Yasuhiro Maejima
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Akihiro Shirakabe
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital, Chiba 270-1694, Japan
| | - Takanobu Yamamoto
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Yoshiyuki Ikeda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medicine, Kagoshima University, Kagoshima 890-8580, Japan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
23
|
Peng P, Nie Z, Sun F, Peng H. Glucocorticoids induce femoral head necrosis in rats through the ROS/JNK/c-Jun pathway. FEBS Open Bio 2020; 11:312-321. [PMID: 33190410 PMCID: PMC7780117 DOI: 10.1002/2211-5463.13037] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/23/2020] [Accepted: 11/11/2020] [Indexed: 01/01/2023] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a common clinical disease with a high disability rate. Apoptosis of osteoblasts caused by high‐dose short‐term or low‐dose long‐term glucocorticoid (GC) administration is the biological basis of steroid‐induced avascular necrosis of the femoral head (SANFH). The pathogenesis of SANFH has not yet been fully elucidated, and there is currently a lack of effective clinical treatments. Here, we investigated the role of the reactive oxygen species (ROS)/JNK/c‐Jun signaling pathway in SANFH. Dexamethasone (Dex) was used to induce apoptosis in osteoblasts, and this resulted in a significant increase in levels of p‐JNK, p‐c‐Jun, Bax, caspase‐3, caspase‐9, cytochrome C, Beclin‐1, and LC3, and a decrease in levels of P62 and Bcl‐2. In addition, intracellular ROS levels were increased and mitochondrial membrane potential was decreased. Administration of 3‐MA, an autophagy inhibitor, attenuated Dex‐mediated changes in autophagy and apoptosis. A rat model of ONFH exhibited severe bone trabecular hollow bone pits along with a significant increase in femoral head cell apoptosis compared with the control group. Additionally, micro‐CT analysis showed that both bone tissue content and femoral head integrity were significantly reduced in the ONFH group. Furthermore, 3‐MA treatment decreased the effect of Dex on GC‐induced ONFH and osteoblast apoptosis in rats and could counteract microstructure destruction due to femoral head necrosis. In summary, our data suggest that GC can induce osteoblast apoptosis and autophagy through the ROS/JNK/c‐Jun signaling pathway, which contributes to ONFH.
Collapse
Affiliation(s)
- Puji Peng
- Department of Orthopedics, Renmin Hospital of Wuhan University, China
| | - Zhigang Nie
- Department of Orthopedics, Renmin Hospital of Wuhan University, China
| | - Fei Sun
- Department of Orthopedics, Renmin Hospital of Wuhan University, China
| | - Hao Peng
- Department of Orthopedics, Renmin Hospital of Wuhan University, China
| |
Collapse
|
24
|
Lichý M, Szobi A, Hrdlička J, Neckář J, Kolář F, Adameová A. Programmed Cell Death in the Left and Right Ventricle of the Late Phase of Post-Infarction Heart Failure. Int J Mol Sci 2020; 21:E7782. [PMID: 33096720 PMCID: PMC7589581 DOI: 10.3390/ijms21207782] [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: 09/09/2020] [Revised: 09/30/2020] [Accepted: 10/19/2020] [Indexed: 01/03/2023] Open
Abstract
While necroptosis has been shown to contribute to the pathogenesis of post-infarction heart failure (HF), the role of autophagy remains unclear. Likewise, linkage between these two cell death modalities has not been sufficiently investigated. HF was induced by 60-min left coronary occlusion in adult Wistar rats and heart function was assessed 6 weeks later followed by immunoblotting analysis of necroptotic and autophagic proteins in both the left (LV) and right ventricle (RV). HF had no effect on RIP1 and RIP3 expression. PhosphoSer229-RIP3, acting as a pro-necroptotic signal, was increased in LV while deceased in RV of failing hearts. Total MLKL was elevated in RV only. Decrease in pSer555-ULK1, increase in pSer473-Akt and no significant elevation in beclin-1 and LC3-II/I ratio indicated rather a lowered rate of autophagy in LV. No beclin-1 upregulation and decreased LC3 processing also suggested the inhibition of both autophagosome formation and maturation in RV of failing hearts. In contrast, p89 PARP1 fragment, a marker of executed apoptosis, was increased in RV only. This is the first study showing a different signaling in ventricles of the late phase of post-infarction HF, highlighting necroptosis itself rather than its linkage with autophagy in LV, and apoptosis in RV.
Collapse
Affiliation(s)
- Martin Lichý
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Kalinčiakova 8, 83232 Bratislava, Slovakia; (M.L.); (A.S.)
| | - Adrián Szobi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Kalinčiakova 8, 83232 Bratislava, Slovakia; (M.L.); (A.S.)
| | - Jaroslav Hrdlička
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (J.H.); (J.N.); (F.K.)
| | - Jan Neckář
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (J.H.); (J.N.); (F.K.)
| | - František Kolář
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (J.H.); (J.N.); (F.K.)
| | - Adriana Adameová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Kalinčiakova 8, 83232 Bratislava, Slovakia; (M.L.); (A.S.)
| |
Collapse
|
25
|
Exercise as a Therapeutic Strategy for Sarcopenia in Heart Failure: Insights into Underlying Mechanisms. Cells 2020; 9:cells9102284. [PMID: 33066240 PMCID: PMC7602002 DOI: 10.3390/cells9102284] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia, a syndrome commonly seen in elderly populations, is often characterized by a gradual loss of skeletal muscle, leading to the decline of muscle strength and physical performance. Growing evidence suggests that the prevalence of sarcopenia increases in patients with heart failure (HF), which is a dominant pathogenesis in the aging heart. HF causes diverse metabolic complications that may result in sarcopenia. Therefore, sarcopenia may act as a strong predictor of frailty, disability, and mortality associated with HF. Currently, standard treatments for slowing muscle loss in patients with HF are not available. Therefore, here, we review the pathophysiological mechanisms underlying sarcopenia in HF as well as current knowledge regarding the beneficial effects of exercise on sarcopenia in HF and related mechanisms, including hormonal changes, myostatin, oxidative stress, inflammation, apoptosis, autophagy, the ubiquitin-proteasome system, and insulin resistance.
Collapse
|
26
|
George SA, Kiss A, Obaid SN, Venegas A, Talapatra T, Wei C, Efimova T, Efimov IR. p38δ genetic ablation protects female mice from anthracycline cardiotoxicity. Am J Physiol Heart Circ Physiol 2020; 319:H775-H786. [PMID: 32822209 PMCID: PMC11018268 DOI: 10.1152/ajpheart.00415.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The efficacy of an anthracycline antibiotic doxorubicin (DOX) as a chemotherapeutic agent is limited by dose-dependent cardiotoxicity. DOX is associated with activation of intracellular stress signaling pathways including p38 MAPKs. While previous studies have implicated p38 MAPK signaling in DOX-induced cardiac injury, the roles of the individual p38 isoforms, specifically, of the alternative isoforms p38γ and p38δ, remain uncharacterized. We aimed to determine the potential cardioprotective effects of p38γ and p38δ genetic deletion in mice subjected to acute DOX treatment. Male and female wild-type (WT), p38γ-/-, p38δ-/-, and p38γ-/-δ-/- mice were injected with 30 mg/kg DOX and their survival was tracked for 10 days. During this period, cardiac function was assessed by echocardiography and electrocardiography and fibrosis by Picro Sirius Red staining. Immunoblotting was performed to assess the expression of signaling proteins and markers linked to autophagy. Significantly improved survival was observed in p38δ-/- female mice post-DOX relative to WT females, but not in p38γ-/- or p38γ-/-δ-/- male or female mice. The improved survival in DOX-treated p38δ-/- females was associated with decreased fibrosis, increased cardiac output and LV diameter relative to DOX-treated WT females, and similar to saline-treated controls. Structural and echocardiographic parameters were either unchanged or worsened in all other groups. Increased autophagy, as suggested by increased LC3-II level, and decreased mammalian target of rapamycin activation was also observed in DOX-treated p38δ-/- females. p38δ plays a crucial role in promoting DOX-induced cardiotoxicity in female mice by inhibiting autophagy. Therefore, p38δ targeting could be a potential cardioprotective strategy in anthracycline chemotherapy.NEW & NOTEWORTHY This study for the first time identifies the sex-specific roles of the alternative p38γ and p38δ MAPK isoforms in promoting doxorubicin (DOX) cardiotoxicity. We show that p38δ and p38γ/δ systemic deletion was cardioprotective in female but not in male mice. Cardiac structure and function were preserved in DOX-treated p38δ-/- females and autophagy marker was increased.
Collapse
Affiliation(s)
- Sharon A George
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Alexi Kiss
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
- The George Washington Cancer Center, Washington, District of Columbia
| | - Sofian N Obaid
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Aileen Venegas
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Trisha Talapatra
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Chapman Wei
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Tatiana Efimova
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
- The George Washington Cancer Center, Washington, District of Columbia
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
- The George Washington Cancer Center, Washington, District of Columbia
| |
Collapse
|
27
|
Wang HN, Li JL, Xu T, Yao HQ, Chen GH, Hu J. Effects of Sirt3‑autophagy and resveratrol activation on myocardial hypertrophy and energy metabolism. Mol Med Rep 2020; 22:1342-1350. [PMID: 32468001 PMCID: PMC7339626 DOI: 10.3892/mmr.2020.11195] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/10/2018] [Indexed: 02/05/2023] Open
Abstract
The aim of the present study was to examine the role of sirtuin 3 (Sirt3)‑autophagy in regulating myocardial energy metabolism and inhibiting myocardial hypertrophy in angiotensin (Ang) II‑induced myocardial cell hypertrophy. The primary cultured myocardial cells of neonatal Sprague Dawley rats were used to construct a myocardial hypertrophy model induced with Ang II. Following the activation of Sirt3 by resveratrol (Res), Sirt3 was silenced using small interfering (si)RNA‑Sirt3, and the morphology of the myocardial cells was observed under an optical microscope. Reverse transcription‑polymerase chain reaction was used to detect the mRNA expression of the following myocardial hypertrophy markers; atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), Sirt3, medium‑chain acyl‑CoA dehydrogenase (MCAD) and pyruvate kinase (PK). Western blot analysis was used to detect the protein expression of Sirt3, light chain 3 (LC3) and Beclin1. Ang II may inhibit the protein expression of Sirt3, LC3 and Beclin1. Res, an agonist of Sirt3, may promote the protein expression of Sirt3, LC3 and Beclin1. Res inhibited the mRNA expression of ANP and BNP, and reversed the Ang II‑induced myocardial cell hypertrophy. The addition of siRNA‑Sirt3 decreased the protein expression of Sirt3, LC3 and Beclin1, increased the mRNA expression of ANP and BNP, and weakened the inhibitory effect of Res on myocardial cell hypertrophy. Res promoted the mRNA expression of MCAD, inhibited the mRNA expression of PK, and reversed the influence of Ang II on myocardial energy metabolism. siRNA‑Sirt3 intervention significantly decreased the effect of Res in eliminating abnormal myocardial energy metabolism. In conclusion, Sirt3 may inhibit Ang II‑induced myocardial hypertrophy and reverse the Ang II‑caused abnormal myocardial energy metabolism through activation of autophagy.
Collapse
Affiliation(s)
- Hai-Ning Wang
- The First Affiliated Hospital of Shantou University Medical College Cardiac Care Unit, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Dr Hai-Ning Wang, The First Affiliated Hospital of Shantou University Medical College Cardiac Care Unit, 57 Changping Road, Shantou, Guangdong 515041, P. R. China, E-mail:
| | - Ji-Lin Li
- The First Affiliated Hospital of Shantou University Medical College Cardiac Care Unit, Shantou, Guangdong 515041, P.R. China
| | - Tan Xu
- The First Affiliated Hospital of Shantou University Medical College Cardiac Care Unit, Shantou, Guangdong 515041, P.R. China
| | - Huai-Qi Yao
- The First Affiliated Hospital of Shantou University Medical College Cardiac Care Unit, Shantou, Guangdong 515041, P.R. China
| | - Gui-Hua Chen
- The First Affiliated Hospital of Shantou University Medical College Cardiac Care Unit, Shantou, Guangdong 515041, P.R. China
| | - Jing Hu
- The First Affiliated Hospital of Shantou University Medical College Cardiac Care Unit, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
28
|
Hughes WE, Beyer AM, Gutterman DD. Vascular autophagy in health and disease. Basic Res Cardiol 2020; 115:41. [PMID: 32506214 DOI: 10.1007/s00395-020-0802-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Homeostasis is maintained within organisms through the physiological recycling process of autophagy, a catabolic process that is intricately involved in the mobilization of nutrients during starvation, recycling of cellular cargo, as well as initiation of cellular death pathways. Specific to the cardiovascular system, autophagy responds to both chemical (e.g. free radicals) and mechanical stressors (e.g. shear stress). It is imperative to note that autophagy is not a static process, and measurement of autophagic flux provides a more comprehensive investigation into the role of autophagy. The overarching themes emerging from decades of autophagy research are that basal levels of autophagic flux are critical, physiological stressors may increase or decrease autophagic flux, and more importantly, aberrant deviations from basal autophagy may elicit detrimental effects. Autophagy has predominantly been examined within cardiac or vascular smooth muscle tissue within the context of disease development and progression. Autophagic flux within the endothelium holds an important role in maintaining vascular function, demonstrated by the necessary role for intact autophagic flux for shear-induced release of nitric oxide however the underlying mechanisms have yet to be elucidated. Within this review, we theorize that autophagy itself does not solely control vascular homeostasis, rather, it works in concert with mitochondria, telomerase, and lipids to maintain physiological function. The primary emphasis of this review is on the role of autophagy within the human vasculature, and the integrative effects with physiological processes and diseases as they relate to the vascular structure and function.
Collapse
Affiliation(s)
- William E Hughes
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA.
| | - Andreas M Beyer
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA
| | - David D Gutterman
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA
| |
Collapse
|
29
|
Examining Cardiomyocyte Dysfunction Using Acute Chemical Induction of an Ageing Phenotype. Int J Mol Sci 2019; 21:ijms21010197. [PMID: 31892165 PMCID: PMC6982016 DOI: 10.3390/ijms21010197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/21/2019] [Accepted: 12/23/2019] [Indexed: 11/18/2022] Open
Abstract
Much effort is focussed on understanding the structural and functional changes in the heart that underlie age-dependent deterioration of cardiac performance. Longitudinal studies, using aged animals, have pinpointed changes occurring to the contractile myocytes within the heart. However, whilst longitudinal studies are important, other experimental approaches are being advanced that can recapitulate the phenotypic changes seen during ageing. This study investigated the induction of an ageing cardiomyocyte phenotypic change by incubation of cells with hydroxyurea for several days ex vivo. Hydroxyurea incubation has been demonstrated to phenocopy age- and senescence-induced changes in neurons, but its utility for ageing studies with cardiac cells has not been examined. Incubation of neonatal rat ventricular myocytes with hydroxyurea for up to 7 days replicated specific aspects of cardiac ageing including reduced systolic calcium responses, increased alternans and a lesser ability of the cells to follow electrical pacing. Additional functional and structural changes were observed within the myocytes that pointed to ageing-like remodelling, including lipofuscin granule accumulation, reduced mitochondrial membrane potential, increased production of reactive oxygen species, and altered ultrastructure, such as mitochondria with disrupted cristae and disorganised myofibres. These data highlight the utility of alternative approaches for exploring cellular ageing whilst avoiding the costs and co-morbid factors that can affect longitudinal studies.
Collapse
|
30
|
Bo-Htay C, Shwe T, Higgins L, Palee S, Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. Aging induced by D-galactose aggravates cardiac dysfunction via exacerbating mitochondrial dysfunction in obese insulin-resistant rats. GeroScience 2019; 42:233-249. [PMID: 31768765 DOI: 10.1007/s11357-019-00132-9] [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: 07/04/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022] Open
Abstract
The prevalence of obesity and an aging population are increasing worldwide. Both obesity and aging are independently known to be associated with cardiac dysfunction. However, in obese insulin-resistant subjects, the effects of aging on metabolic status and cardiac and mitochondrial functions are not completely understood. We hypothesized that in the obese insulin-resistant condition, aging induced by D-galactose increases cardiac senescence markers and aggravates the impairment of metabolic parameters, cardiac and mitochondrial function, and increases oxidative stress, inflammation, apoptosis, and autophagy. Sixty-four male Wistar rats were fed with either normal diet (ND) or high-fat diet (HFD) for 12 weeks. Then, rats were divided into vehicle groups (0.9% NSS, subcutaneous injection (SC)) or D-galactose groups (150 mg/kg/day, SC). After 0.9%NSS or D-galactose treatment for 4 weeks and 8 weeks, metabolic and cardiac functions were determined. The heart was then removed to determine mitochondrial functions and enable biochemical studies. After 4 weeks of D-galactose injection, ND rats treated with D-galactose (NDD4), HFD rats treated with vehicle (HFV4), and HFD rats treated with D-galactose (HFD4) had reduced cardiac function, impaired cardiac mitochondrial function and autophagy, and increased oxidative stress, inflammation, and apoptosis. Interestingly, after 8 weeks, HFD rats treated with D-galactose (HFD8) had the worst impairment of cardiac and mitochondrial function, autophagy, and apoptosis in comparison to the other groups. Aging induced by D-galactose aggravated cardiac dysfunction in obese insulin-resistant rats through the worsening of cardiac mitochondrial function, autophagy, and increased apoptosis in a time-dependent manner.
Collapse
Affiliation(s)
- Cherry Bo-Htay
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Thazin Shwe
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Louis Higgins
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siripong Palee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Krekwit Shinlapawittayatorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
31
|
Abstract
Patients with diabetes mellitus have >2× the risk for developing heart failure (HF; HF with reduced ejection fraction and HF with preserved ejection fraction). Cardiovascular outcomes, hospitalization, and prognosis are worse for patients with diabetes mellitus relative to those without. Beyond the structural and functional changes that characterize diabetic cardiomyopathy, a complex underlying, and interrelated pathophysiology exists. Despite the success of many commonly used antihyperglycemic therapies to lower hyperglycemia in type 2 diabetes mellitus the high prevalence of HF persists. This, therefore, raises the possibility that additional factors beyond glycemia might contribute to the increased HF risk in diabetes mellitus. This review summarizes the state of knowledge about the impact of existing antihyperglycemic therapies on HF and discusses potential mechanisms for beneficial or deleterious effects. Second, we review currently approved pharmacological therapies for HF and review evidence that addresses their efficacy in the context of diabetes mellitus. Dysregulation of many cellular mechanisms in multiple models of diabetic cardiomyopathy and in human hearts have been described. These include oxidative stress, inflammation, endoplasmic reticulum stress, aberrant insulin signaling, accumulation of advanced glycated end-products, altered autophagy, changes in myocardial substrate metabolism and mitochondrial bioenergetics, lipotoxicity, and altered signal transduction such as GRK (g-protein receptor kinase) signaling, renin angiotensin aldosterone signaling and β-2 adrenergic receptor signaling. These pathophysiological pathways might be amenable to pharmacological therapy to reduce the risk of HF in the context of type 2 diabetes mellitus. Successful targeting of these pathways could alter the prognosis and risk of HF beyond what is currently achieved using existing antihyperglycemic and HF therapeutics.
Collapse
Affiliation(s)
- Helena C Kenny
- From the Fraternal Order of Eagles Diabetes Research Center, and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City
| | - E Dale Abel
- From the Fraternal Order of Eagles Diabetes Research Center, and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City
| |
Collapse
|
32
|
Tomin T, Schittmayer M, Honeder S, Heininger C, Birner-Gruenberger R. Irreversible oxidative post-translational modifications in heart disease. Expert Rev Proteomics 2019; 16:681-693. [PMID: 31361162 PMCID: PMC6816499 DOI: 10.1080/14789450.2019.1645602] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Development of specific biomarkers aiding early diagnosis of heart failure is an ongoing challenge. Biomarkers commonly used in clinical routine usually act as readouts of an already existing acute condition rather than disease initiation. Functional decline of cardiac muscle is greatly aggravated by increased oxidative stress and damage of proteins. Oxidative post-translational modifications occur already at early stages of tissue damage and are thus regarded as potential up-coming disease markers. Areas covered: Clinical practice regarding commonly used biomarkers for heart disease is briefly summarized. The types of oxidative post-translational modification in cardiac pathologies are discussed with a special focus on available quantitative techniques and characteristics of individual modifications with regard to their stability and analytical accessibility. As irreversible oxidative modifications trigger protein degradation pathways or cause protein aggregation, both influencing biomarker abundance, a chapter is dedicated to their regulation in the heart.
Collapse
Affiliation(s)
- Tamara Tomin
- Institute of Pathology, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz , Graz , Austria.,Omics Center Graz, BioTechMed-Graz , Graz , Austria.,Institute of Chemical Technologies and Analytics, Vienna University of Technology , Vienna , Austria
| | - Matthias Schittmayer
- Institute of Pathology, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz , Graz , Austria.,Omics Center Graz, BioTechMed-Graz , Graz , Austria.,Institute of Chemical Technologies and Analytics, Vienna University of Technology , Vienna , Austria
| | - Sophie Honeder
- Institute of Pathology, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz , Graz , Austria.,Omics Center Graz, BioTechMed-Graz , Graz , Austria
| | - Christoph Heininger
- Institute of Pathology, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz , Graz , Austria.,Omics Center Graz, BioTechMed-Graz , Graz , Austria
| | - Ruth Birner-Gruenberger
- Institute of Pathology, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz , Graz , Austria.,Omics Center Graz, BioTechMed-Graz , Graz , Austria.,Institute of Chemical Technologies and Analytics, Vienna University of Technology , Vienna , Austria
| |
Collapse
|
33
|
Sun M, Tan Y, Rexiati M, Dong M, Guo W. Obesity is a common soil for premature cardiac aging and heart diseases - Role of autophagy. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1898-1904. [DOI: 10.1016/j.bbadis.2018.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/22/2018] [Accepted: 09/04/2018] [Indexed: 12/31/2022]
|
34
|
Long Noncoding Competing Endogenous RNA Networks in Age-Associated Cardiovascular Diseases. Int J Mol Sci 2019; 20:ijms20123079. [PMID: 31238513 PMCID: PMC6627372 DOI: 10.3390/ijms20123079] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the most serious health problem in the world, displaying high rates of morbidity and mortality. One of the main risk factors for CVDs is age. Indeed, several mechanisms are at play during aging, determining the functional decline of the cardiovascular system. Aging cells and tissues are characterized by diminished autophagy, causing the accumulation of damaged proteins and mitochondria, as well as by increased levels of oxidative stress, apoptosis, senescence and inflammation. These processes can induce a rapid deterioration of cellular quality-control systems. However, the molecular mechanisms of age-associated CVDs are only partially known, hampering the development of novel therapeutic strategies. Evidence has emerged indicating that noncoding RNAs (ncRNAs), such as long ncRNAs (lncRNAs) and micro RNAs (miRNAs), are implicated in most patho-physiological mechanisms. Specifically, lncRNAs can bind miRNAs and act as competing endogenous-RNAs (ceRNAs), therefore modulating the levels of the mRNAs targeted by the sponged miRNA. These complex lncRNA/miRNA/mRNA networks, by regulating autophagy, apoptosis, necrosis, senescence and inflammation, play a crucial role in the development of age-dependent CVDs. In this review, the emerging knowledge on lncRNA/miRNA/mRNA networks will be summarized and the way in which they influence age-related CVDs development will be discussed.
Collapse
|
35
|
Zhang D, Bhatnagar A, Baba SP. Inhibition of aldose reductase activity stimulates starvation induced autophagy and clears aldehyde protein adducts. Chem Biol Interact 2019; 306:104-109. [DOI: 10.1016/j.cbi.2019.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/14/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
|
36
|
Puerarin Decreases Collagen Secretion in AngII-Induced Atrial Fibroblasts Through Inhibiting Autophagy Via the JNK–Akt–mTOR Signaling Pathway. J Cardiovasc Pharmacol 2019; 73:373-382. [DOI: 10.1097/fjc.0000000000000672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
37
|
Yin J, Lu X, Qian Z, Xu W, Zhou X. New insights into the pathogenesis and treatment of sarcopenia in chronic heart failure. Am J Cancer Res 2019; 9:4019-4029. [PMID: 31281529 PMCID: PMC6592172 DOI: 10.7150/thno.33000] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
Sarcopenia is an age-related geriatric syndrome that is characterized by a progressive loss of muscle mass, strength and function. Chronic heart failure (CHF), the final stage of various cardiovascular diseases, may be closely correlated with the occurrence of sarcopenia. Accumulating evidence has demonstrated that CHF can promote the development of sarcopenia through multiple pathophysiological mechanisms, including malnutrition, inflammation, hormonal changes, oxidative stress, autophagy, and apoptosis. Additionally, CHF can aggravate the adverse outcomes associated with sarcopenia, including falls, osteoporosis, frailty, cachexia, hospitalization, and mortality. Sarcopenia and CHF are mutually interacting clinical syndromes. Patients with these two syndromes seem to endure a double burden, with no particularly effective way to hinder their progression. However, the combination of physical exercise, nutritional supplements, and drug therapy may counteract the development of these maladies. In this review, we will summarize the latest progress in the pathogenesis and treatment of sarcopenia in patients with CHF.
Collapse
|
38
|
Corsetti G, Chen-Scarabelli C, Romano C, Pasini E, Dioguardi FS, Onorati F, Knight R, Patel H, Saravolatz L, Faggian G, Scarabelli TM. Autophagy and Oncosis/Necroptosis Are Enhanced in Cardiomyocytes from Heart Failure Patients. Med Sci Monit Basic Res 2019; 25:33-44. [PMID: 30713336 PMCID: PMC6373236 DOI: 10.12659/msmbr.913436] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Although originally described as a survival mechanism, it is unknown whether and to what extent autophagy is implicated in the terminal stages of heart failure. Here, we studied magnitude and evolution of autophagy in patients with intractable heart failure. Material/Methods Myocardial samples were obtained from 22 patients with ischemic cardiomyopathy and idiopathic dilated cardiomyopathy who were undergoing cardiac transplantation. Hearts from 11 patients who died from non-cardiac causes were used as control samples. Autophagy was evaluated by immunostaining with a monoclonal microtubule associated protein light chain 3 (LC3)-II antibody, while the relationship of autophagy with apoptosis and oncosis was assessed by double staining with TUNEL (terminal deoxynucleotidyl transferase – mediated deoxyuridine triphosphate nick end labeling) assay and complement 9 (C9) immunological staining, respectively. In addition, several necroptotic markers, including RIP1 and RIP3 (receptor interacting protein kinase 1 and 3), anti-C3 (cleaved-caspase-3), and anti-NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) were assessed by immunohistochemistry. Results Anti-LC3-II staining was detected in 8.7±1.6% of the heart failure patient heart samples and in 1.2±0.3% of control patient heart samples. Vacuole formation started at one nuclear pole, before becoming bipolar and involving the cytosol. Subsequently, the autophagic process extended also to the nuclei, which underwent a progressive vacuolization and disintegration, assuming a peculiar “strawberry like appearance”. Myocytes with extensive vacuole formation exhibited nuclear degeneration, which was associated with TUNEL, C3, C9, RIP1, and RIP3 positive staining. Conversely, myocytes with less extensive vacuole formation showed RIP1 and NF-κB positive staining, though not positivity for other cell death markers. Conclusions Autophagy was extensively detected in end-stage heart failure and its progression, resulted in secondary cell death, with occurrence of oncosis and necroptosis exceeding that of apoptosis. Conversely, activation of the RIP1/NF-κB pathway was associated with cell survival.
Collapse
Affiliation(s)
- Giovanni Corsetti
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Carol Chen-Scarabelli
- Center for Heart and Vessel Preclinical Studies, Department of Internal Medicine, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, USA
| | - Claudia Romano
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Evasio Pasini
- Scientific Clinical Institutes Maugeri, Cardiac Rehabilitation Lumezzane Institute, Brescia, Italy
| | | | - Francesco Onorati
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Richard Knight
- Medical Research Council (MRC) Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Hemang Patel
- General Medical Education, Department of Internal Medicine, Ascension St. John Hospital, Detroit, MI, USA
| | - Louis Saravolatz
- Department of Medicine, Ascension St John Hospital and Wayne State University School of Medicine, Detroit, MI, USA
| | - Giuseppe Faggian
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Tiziano M Scarabelli
- Center for Heart and Vessel Preclinical Studies, Department of Internal Medicine, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, USA
| |
Collapse
|
39
|
Esterline RL, Vaag A, Oscarsson J, Vora J. MECHANISMS IN ENDOCRINOLOGY: SGLT2 inhibitors: clinical benefits by restoration of normal diurnal metabolism? Eur J Endocrinol 2018; 178:R113-R125. [PMID: 29371333 DOI: 10.1530/eje-17-0832] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/19/2018] [Indexed: 12/26/2022]
Abstract
Type 2 diabetes (T2D) is associated with inhibition of autophagic and lysosomal housekeeping processes that detrimentally affect key organ functioning; a process likely to be exacerbated by conventional insulin-driven anabolic therapies. We propose that the cardio-renal benefits demonstrated with sodium-glucose cotransporter-2 inhibitor (SGLT2i) treatment in T2D partly may be explained by their ability to drive consistent, overnight periods of increased catabolism brought about by constant glucosuria. Key steps driving this catabolic mechanism include: a raised glucagon/insulin ratio initially depleting glycogen in the liver and ultimately activating gluconeogenesis utilizing circulating amino acids (AAs); a general fuel switch from glucose to free fatty acids (accompanied by a change in mitochondrial morphology from a fission to a sustained fusion state driven by a decrease in AA levels); a decrease in circulating AAs and insulin driving inhibition of mammalian target of rapamycin complex 1 (mTORC1), which enhances autophagy/lysosomal degradation of dysfunctional organelles, eventually causing a change in mitochondrial morphology from a fission to a sustained fusion state. Resumption of eating in the morning restores anabolic biogenesis of new and fully functional organelles and proteins. Restoration of diurnal metabolic rhythms and flexibility by SGLT2is may have therapeutic implications beyond those already demonstrated for the cardio-renal axis and may therefore affect other non-diabetes disease states.
Collapse
Affiliation(s)
| | - Allan Vaag
- Cardiovascular and Metabolic Disease (CVMD) Translational Medicine Unit, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Jan Oscarsson
- AstraZeneca Pharmaceuticals, Gaithersburg, Maryland, USA
| | - Jiten Vora
- Royal Liverpool University Hospital, University of Liverpool, Liverpool, UK
| |
Collapse
|
40
|
Li Y, Yu QH, Chu Y, Wu WM, Song JX, Zhu XB, Wang Q. Blockage of AKAP12 accelerates angiotensin II (Ang II)-induced cardiac injury in mice by regulating the transforming growth factor β1 (TGF-β1) pathway. Biochem Biophys Res Commun 2018; 499:128-135. [PMID: 29501491 DOI: 10.1016/j.bbrc.2018.02.200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 02/27/2018] [Indexed: 02/06/2023]
Abstract
Hypertension is a multifactorial chronic inflammatory disease that leads to cardiac remodeling. A-kinase anchor protein 12 (AKAP12) is a scaffolding protein that has multiple functions in various biological events, including the regulation of vessel integrity and differentiation of neural barriers in blood. However, the role of AKAP12 in angiotensin II (Ang II)-induced cardiac injury remains unclear. In the present study, Ang II infusion reduced AKAP12 expressions in the hearts of wild-type (WT) mice, and AKAP12 knockout (KO) enhanced the infiltration of inflammatory cells. In addition, AKAP12 deletion accelerated Ang II-induced cardiac histologic alterations and dysfunction. Further, AKAP12-/- aggravated heart failure by promoting the inflammation, oxidative stress, cellular apoptosis, and autophagy induced by Ang II. Furthermore, AKAP12 KO elevated Ang II-induced cardiac fibrosis, as indicated by the following: (1) Masson trichrome staining showed that Ang II infusion markedly increased fibrotic areas of the WT mouse heart, which was greatly accelerated in AKAP12-/- mice; (2) immunohistochemistry analysis showed increased expression of transforming growth factor β1 (TGF-β1) and α-smooth muscle actin (α-SMA) in the AKAP12-/- mouse heart; (3) reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) analysis showed increased expression of fibrosis-related molecules in the AKAP12-deficient mouse heart; and (4) Western blot analysis indicated significantly higher upregulation of p-SMAD2/3 in the AKAP12-/- mouse heart. In vitro, AKAP12 knockdown in HL-1 cells was responsible for TGF-β1-induced inflammation, the generation of reactive oxygen species (ROS), apoptosis, autophagy, and fibrosis. Furthermore, overexpression of AKAP12 reduced fibrosis triggered by TGF-β1 in cells. Overall, our study suggests that fibrosis induced by Ang II may be alleviated by AKAP12 expression through inactivation of the TGF-β1 pathway.
Collapse
Affiliation(s)
- Yong Li
- Department of Cardiology, Wujin People's Hospital of Changzhou, Changzhou 213017, China
| | - Qiu-Hua Yu
- Department of Cardio-Thoracic, Wujin People's Hospital of Changzhou, Changzhou 213017, China
| | - Ying Chu
- Central Laboratory, Wujin People's Hospital of Changzhou, Changzhou 213017, China
| | - Wei-Min Wu
- Department of Cardio-Thoracic, Wujin People's Hospital of Changzhou, Changzhou 213017, China
| | - Jian-Xiang Song
- Department of Cardiac Surgery, The Third Hospital of Yancheng, Yancheng 224000, China
| | - Xiao-Bo Zhu
- Department of Cardio-Thoracic, Wujin People's Hospital of Changzhou, Changzhou 213017, China
| | - Qiang Wang
- Department of Cardio-Thoracic, Wujin People's Hospital of Changzhou, Changzhou 213017, China.
| |
Collapse
|
41
|
Martín-Fernández B, Gredilla R. Mitochondrial oxidative stress and cardiac ageing. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2018; 30:74-83. [PMID: 29398015 DOI: 10.1016/j.arteri.2017.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/11/2017] [Accepted: 12/14/2017] [Indexed: 10/18/2022]
Abstract
According with different international organizations, cardiovascular diseases are becoming the first cause of death in western countries. Although exposure to different risk factors, particularly those related to lifestyle, contribute to the etiopathogenesis of cardiac disorders, the increase in average lifespan and aging are considered major determinants of cardiac diseases events. Mitochondria and oxidative stress have been pointed out as relevant factors both in heart aging and in the development of cardiac diseases such as heart failure, cardiac hypertrophy and diabetic cardiomyopathy. During aging, cellular processes related with mitochondrial function, such as bioenergetics, apoptosis and inflammation are altered leading to cardiac dysfunction. Increasing our knowledge about the mitochondrial mechanisms related with the aging process, will provide new strategies in order to improve this process, particularly the cardiovascular ones.
Collapse
Affiliation(s)
- Beatriz Martín-Fernández
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España.
| | - Ricardo Gredilla
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España
| |
Collapse
|
42
|
Zhang Z, Li H, Chen S, Li Y, Cui Z, Ma J. Knockdown of MicroRNA-122 Protects H9c2 Cardiomyocytes from Hypoxia-Induced Apoptosis and Promotes Autophagy. Med Sci Monit 2017; 23:4284-4290. [PMID: 28871076 PMCID: PMC5597036 DOI: 10.12659/msm.902936] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a severe disease causing heart failure and sudden death. Studies indicate that microRNAs (miRNAs) are involved in the pathophysiology of AMI. In the present study, we carefully explored the effects of miR-122 on myocardial hypoxia injury and its possible underlying mechanism. MATERIAL AND METHODS miR-122 expression was analyzed in H9c2 cardiomyocytes after being transfected with miR-122 mimic, ASO-miR-122, or negative control. Cell viability and apoptosis were investigated by CCK-8 assays and flow cytometry analysis, respectively. Cell migration was analyzed using wound-healing assays. Western blotting was performed to analyze the expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN)/phosphatidylinositol 3-hydroxy kinase (PI3K)/AKT and LC3-II/LC3-I. RESULTS Hypoxia exposure significantly inhibited H9c2 cell viability (P<0.01). miR-122 overexpression promoted the hypoxia-induced H9c2 cell proliferation and migration loss (P<0.05), and cell apoptosis was increased (P<0.05). miR-122 knockdown enhanced cell viability and decreased cell apoptosis (P<0.05). Knockdown of miR-122 enhanced PTEN/PI3K/AKT activation and cell autophagy. Overexpression of miR-122 inhibited the PTEN/PI3K/AKT pathway and cell autophagy pathway. CONCLUSIONS The expression of miR-122 is involved in hypoxia-induced H9c2 cardiomyocyte injury. Knockdown of miR-122 protects H9c2 cells from hypoxia-induced apoptosis and enhances cell viability.
Collapse
Affiliation(s)
- Zaiwei Zhang
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Hu Li
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Shasha Chen
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Ying Li
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Zhiyuan Cui
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Jie Ma
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|
43
|
Noordali H, Loudon BL, Frenneaux MP, Madhani M. Cardiac metabolism - A promising therapeutic target for heart failure. Pharmacol Ther 2017; 182:95-114. [PMID: 28821397 DOI: 10.1016/j.pharmthera.2017.08.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Both heart failure with reduced ejection fraction (HFrEF) and with preserved ejection fraction (HFpEF) are associated with high morbidity and mortality. Although many established pharmacological interventions exist for HFrEF, hospitalization and death rates remain high, and for those with HFpEF (approximately half of all heart failure patients), there are no effective therapies. Recently, the role of impaired cardiac energetic status in heart failure has gained increasing recognition with the identification of reduced capacity for both fatty acid and carbohydrate oxidation, impaired function of the electron transport chain, reduced capacity to transfer ATP to the cytosol, and inefficient utilization of the energy produced. These nodes in the genesis of cardiac energetic impairment provide potential therapeutic targets, and there is promising data from recent experimental and early-phase clinical studies evaluating modulators such as carnitine palmitoyltransferase 1 inhibitors, partial fatty acid oxidation inhibitors and mitochondrial-targeted antioxidants. Metabolic modulation may provide significant symptomatic and prognostic benefit for patients suffering from heart failure above and beyond guideline-directed therapy, but further clinical trials are needed.
Collapse
Affiliation(s)
- Hannah Noordali
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Brodie L Loudon
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | - Melanie Madhani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
44
|
Liu H, Lei H, Shi Y, Wang JJ, Chen N, Li ZH, Chen YF, Ye QF, Yang Y. Autophagy inhibitor 3-methyladenine alleviates overload-exercise-induced cardiac injury in rats. Acta Pharmacol Sin 2017; 38:990-997. [PMID: 28260802 DOI: 10.1038/aps.2016.169] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/09/2016] [Indexed: 12/14/2022] Open
Abstract
Overload-exercise (OE) causes myocardial injury through inducing autophagy and apoptosis. In this study we examined whether an autophagy inhibitor 3-methyladenine (3-MA) could alleviate OE-induced cardiac injury. Rats were injected with 3-MA (15 mg/kg, iv) or saline before subjected to various intensities of OE, including no swim (control), 2 h swim (mild-intensity exercise, MIE), 2 h swim with 2.5% body weight overload (moderate OE; MOE), 5% overload (intensive OE; IOE) or 2.5% overload until exhausted (exhaustive OE; EOE). After OE, the hearts were harvested for morphological and biochemiacal analysis. The cardiac morphology, autophagosomes and apoptosis were examined with H&E staining, transmission electron microscopy and TUNEL analysis, respectively. Autophagy-related proteins to (LC3-II/I and Beclin-1) and apoptosis-related proteins (Bcl-2/Bax) were assessed using Western blotting. Our results showed that compared with the control, MIE did not change the morphological structures of the heart tissues that exhibited intact myocardial fibers and neatly arranged cardiomyocytes. However, IOE resulted in irregular arrangement of cardiomyocytes and significantly increased width of cardiomyocytes, whereas EOE caused more swollen and even disrupted cardiomyocytes. In parallel with the increased OE intensity (MOE, IOE, EOE), cardiomyocyte autophagy and apoptosis became more and more prominent, evidenced by the increasing number of autophagosomes and expression levels of LC3-II/I and Beclin-1 as well as the increasing apoptotic cells and decreasing Bcl-2/Bax ratio. 3-MA administration significantly attenuated OE-induced morphological changes of cardiomyocytes as well as all the autophagy- and apoptosis-related abnormalities in MOE, IOE and EOE rats. Thus, the autophagy inhibitor 3-MA could alleviate OE-induced heart injury in rats.
Collapse
|
45
|
The therapeutic hope for HDAC6 inhibitors in malignancy and chronic disease. Clin Sci (Lond) 2017; 130:987-1003. [PMID: 27154743 DOI: 10.1042/cs20160084] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/01/2016] [Indexed: 12/12/2022]
Abstract
Recent years have witnessed an emergence of a new class of therapeutic agents, termed histone deacetylase 6 (HDAC6) inhibitors. HDAC6 is one isoform of a family of HDAC enzymes that catalyse the removal of functional acetyl groups from proteins. It stands out from its cousins in almost exclusively deacetylating cytoplasmic proteins, in exerting deacetylation-independent effects and in the success that has been achieved in developing relatively isoform-specific inhibitors of its enzymatic action that have reached clinical trial. HDAC6 plays a pivotal role in the removal of misfolded proteins and it is this role that has been most successfully targeted to date. HDAC6 inhibitors are being investigated for use in combination with proteasome inhibitors for the treatment of lymphoid malignancies, whereby HDAC6-dependent protein disposal currently limits the cytotoxic effectiveness of the latter. Similarly, numerous recent studies have linked altered HDAC6 activity to the pathogenesis of neurodegenerative diseases that are characterized by misfolded protein accumulation. It seems likely though that the function of HDAC6 is not limited to malignancy and neurodegeneration, the deacetylase being implicated in a number of other cellular processes and diseases including in cardiovascular disease, inflammation, renal fibrosis and cystogenesis. Here, we review the unique features of HDAC6 that make it so appealing as a drug target and its currently understood role in health and disease. Whether HDAC6 inhibition will ultimately find a clinical niche in the treatment of malignancy or prevalent complex chronic diseases remains to be determined.
Collapse
|
46
|
Bartlett JJ, Trivedi PC, Pulinilkunnil T. Autophagic dysregulation in doxorubicin cardiomyopathy. J Mol Cell Cardiol 2017; 104:1-8. [DOI: 10.1016/j.yjmcc.2017.01.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 01/12/2017] [Accepted: 01/16/2017] [Indexed: 12/22/2022]
|
47
|
Preclinical Development of a MicroRNA-Based Therapy for Elderly Patients With Myocardial Infarction. J Am Coll Cardiol 2016; 68:1557-71. [DOI: 10.1016/j.jacc.2016.07.739] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/21/2016] [Accepted: 07/05/2016] [Indexed: 11/22/2022]
|
48
|
Martín-Fernández B, Gredilla R. Mitochondria and oxidative stress in heart aging. AGE (DORDRECHT, NETHERLANDS) 2016; 38:225-238. [PMID: 27449187 PMCID: PMC5061683 DOI: 10.1007/s11357-016-9933-y] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 07/12/2016] [Indexed: 05/06/2023]
Abstract
As average lifespan of humans increases in western countries, cardiac diseases become the first cause of death. Aging is among the most important risk factors that increase susceptibility for developing cardiovascular diseases. The heart has very aerobic metabolism, and is highly dependent on mitochondrial function, since mitochondria generate more than 90 % of the intracellular ATP consumed by cardiomyocytes. In the last few decades, several investigations have supported the relevance of mitochondria and oxidative stress both in heart aging and in the development of cardiac diseases such as heart failure, cardiac hypertrophy, and diabetic cardiomyopathy. In the current review, we compile different studies corroborating this role. Increased mitochondria DNA instability, impaired bioenergetic efficiency, enhanced apoptosis, and inflammation processes are some of the events related to mitochondria that occur in aging heart, leading to reduced cellular survival and cardiac dysfunction. Knowing the mitochondrial mechanisms involved in the aging process will provide a better understanding of them and allow finding approaches to more efficiently improve this process.
Collapse
Affiliation(s)
- Beatriz Martín-Fernández
- Department of Physiology, Faculty of Medicine, Complutense University, Plaza Ramon y Cajal s/n, 28040, Madrid, Spain.
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Plaza Ramon y Cajal s/n, 28040, Madrid, Spain.
| |
Collapse
|
49
|
Li B, Chi RF, Qin FZ, Guo XF. Distinct changes of myocyte autophagy during myocardial hypertrophy and heart failure: association with oxidative stress. Exp Physiol 2016; 101:1050-63. [PMID: 27219474 DOI: 10.1113/ep085586] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 05/19/2016] [Indexed: 01/04/2023]
Abstract
NEW FINDINGS What is the central question of this study? We investigated the changes of myocyte autophagy during the stages of myocardial hypertrophy and failure and the relationship between autophagy and oxidative stress. What is the main findings and its importance? Myocyte autophagy is reduced during myocardial hypertrophy and increased during heart failure. Reduced autophagy is correlated with myocyte hypertrophy, and increased autophagy is correlated with myocyte apoptosis. The distinct alterations are associated with oxidative stress. Hydrogen peroxide causes distinct, concentration-dependent changes in autophagy in cultured cardiomyocytes. Oxidative stress may mediate the distinct alterations of myocyte autophagy during cardiac hypertrophy and failure. Myocyte autophagy occurs at basal levels in the heart in normal conditions and increases in heart failure. However, the changes of myocyte autophagy during the stages of myocardial hypertrophy and failure are not fully understood. Little is known about the relationship among myocyte autophagy, hypertrophy, apoptosis and oxidative stress. In the present study, we first examined the changes of myocyte autophagy in mice with chronic pressure overload and the relationships between myocyte autophagy and hypertrophy, apoptosis and oxidative stress. Second, we determined the direct role of hydrogen peroxide on autophagy in cultured cardiomyocytes. Eight-week-old male C57BL/6J mice underwent transverse aortic constriction (TAC) or sham operation. In TAC mice, left ventricular wall thickness was increased at 1 week and increased further at 9 weeks. Left ventricular end-diastolic dimension showed no change at 1 week, but increased at 9 weeks in association with systolic dysfunction. Myocyte autophagy was decreased at 1 week after TAC, and the decrease was correlated with increased myocyte size. Myocyte autophagy was increased at 9 weeks after TAC, and the increase was correlated with increased myocyte apoptosis. The alterations in autophagy after TAC were associated with myocardial oxidative stress. Hydrogen peroxide caused distinct, concentration-dependent changes in autophagy in cultured cardiomyocytes. In conclusion, myocyte autophagy was decreased during myocardial hypertrophy and increased during heart failure. The distinct changes were associated with myocyte hypertrophy, apoptosis and oxidative stress. These findings suggest that oxidative stress may mediate the distinct alterations of myocyte autophagy during myocardial hypertrophy and heart failure.
Collapse
Affiliation(s)
- Bao Li
- The Affiliated Cardiovascular Hospital of Shanxi Medical University, Taiyuan, 030024, Shanxi, PR China.,Shanxi Province Cardiovascular Hospital, Taiyuan, 030024, Shanxi, PR China.,Shanxi Province Cardiovascular Institute, Taiyuan, 030024, Shanxi, PR China
| | - Rui-Fang Chi
- The Affiliated Cardiovascular Hospital of Shanxi Medical University, Taiyuan, 030024, Shanxi, PR China.,Shanxi Province Cardiovascular Hospital, Taiyuan, 030024, Shanxi, PR China.,Shanxi Province Cardiovascular Institute, Taiyuan, 030024, Shanxi, PR China.,Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Fu-Zhong Qin
- The Affiliated Cardiovascular Hospital of Shanxi Medical University, Taiyuan, 030024, Shanxi, PR China.,Shanxi Province Cardiovascular Hospital, Taiyuan, 030024, Shanxi, PR China.,Shanxi Province Cardiovascular Institute, Taiyuan, 030024, Shanxi, PR China.,Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Xiao-Fei Guo
- The Affiliated Cardiovascular Hospital of Shanxi Medical University, Taiyuan, 030024, Shanxi, PR China.,Shanxi Province Cardiovascular Hospital, Taiyuan, 030024, Shanxi, PR China.,Shanxi Province Cardiovascular Institute, Taiyuan, 030024, Shanxi, PR China.,Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| |
Collapse
|
50
|
Yang Y, Yang X, Dong Y, Chen N, Xiao X, Liu H, Li Z, Chen Y. Transcutaneous electrical acupoint stimulation alleviates adverse cardiac remodeling induced by overload training in rats. J Appl Physiol (1985) 2016; 120:1269-76. [PMID: 27032900 DOI: 10.1152/japplphysiol.00077.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/28/2016] [Indexed: 02/06/2023] Open
Abstract
Electroacupuncture has been shown previously to alleviate cardiac ischemia-reperfusion injury. Overload training (OT) exercise can result in profound cardiac damage and remodeling. In this study, we aimed to examine whether transcutaneous electrical acupoint stimulation (TEAS), a novel noninvasive and low-risk alternative to electroacupuncture, could counteract short-term OT-induced cardiac remodeling, fibrosis, autophagy, and apoptosis. Sixty rats were randomly divided into eight groups (n = 7 or 8/group): control, regular exercise, OT, OT plus low-, moderate- or high-frequency TEAS preconditioning, OT plus moderate-frequency TEAS postconditioning, or transcutaneous electrical nonacupoint stimulation (TENAS) preconditioning. The cardiac weight index (heart weight/body weight) was determined. Left ventricular morphology was examined by hematoxylin and eosin staining. Cardiac fibrosis and apoptosis were determined by Masson's trichrome and TUNEL staining, respectively. The presence of autophagosomes was observed by transmission electron microcopy. The expressions of autophagic markers (LC3 II/I and Beclin-1) were determined by Western blot. The results showed that 1) OT induced adverse cardiac structure changes but did not affect the cardiac weight index; 2) OT increased cardiac fibrosis and apoptosis and induced autophagosome formation with upregulated LC3 II/I and Beclin-1 expression; 3) TEAS preconditioning effectively alleviated OT-induced cardiac structure changes, fibrosis, apoptosis, and autophagy; 4) TEAS preconditioning produced better protective effects than TEAS postconditioning or TENAS preconditioning. Our results demonstrate that TEAS preconditioning protects the heart from OT-induced cardiac injury/remodeling, probably by inhibition of fibrosis, autophagy, and apoptosis.
Collapse
Affiliation(s)
- Yi Yang
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Xi Yang
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Yuchen Dong
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Ning Chen
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Xiang Xiao
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio
| | - Hua Liu
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Zhanghua Li
- Tongren Hospital of Wuhan University, Wuhan, China; and
| | - Yanfang Chen
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio
| |
Collapse
|