1
|
Ibrahim NA, Buabeid MA, Shaimaa Arafa E, Elmorshedy KE. Zinc's protective role against hydroxychloroquine-induced cardiac effects in adult male albino rats. Saudi J Biol Sci 2023; 30:103733. [PMID: 37521750 PMCID: PMC10374629 DOI: 10.1016/j.sjbs.2023.103733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Background Long exposure to Hydroxychloroquine (HCQ) has been complicated by some dangerous though infrequent cardiotoxicity. Methods A total of 40 normal adult male albino rats dispersed into 4 groups were used. Group 1 (Control group), Group II (HCQ treated group), Group III (zinc [Zn]-treated group), and Group IV (HCQ and Zn treated group). Once the experimentation ended, rats were sacrificed and cardiac soft tissue sections were processed twenty-four hours at the end of the experiment for histological study. Results Cardiac-stained sections revealed that HCQ induced widespread necrosis, dilatation, and vacuolar degeneration. However, the combination of HCQ with Zn ameliorated these damaging effects. Cardiac enzyme parameters were also studied in the 4 groups and revealed CK-MB and troponin were considerably elevated in groups II associated to the control group. Conclusion It was concluded that Zn revealed a protective role against HCQ cardiomyopathy in adult male albino rats. This might signify an appreciated means for Zn-based treatment in the upcoming subsequent clinical records to adjust doses and guarantee patient safeguard.
Collapse
Affiliation(s)
- Nihal A. Ibrahim
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, UAE
- Centre of Medical and Bio-allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, UAE
| | | | - El Shaimaa Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, UAE
- Centre of Medical and Bio-allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, UAE
| | | |
Collapse
|
2
|
Taylor JA, Greenhaff PL, Bartlett DB, Jackson TA, Duggal NA, Lord JM. Multisystem physiological perspective of human frailty and its modulation by physical activity. Physiol Rev 2023; 103:1137-1191. [PMID: 36239451 PMCID: PMC9886361 DOI: 10.1152/physrev.00037.2021] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
"Frailty" is a term used to refer to a state characterized by enhanced vulnerability to, and impaired recovery from, stressors compared with a nonfrail state, which is increasingly viewed as a loss of resilience. With increasing life expectancy and the associated rise in years spent with physical frailty, there is a need to understand the clinical and physiological features of frailty and the factors driving it. We describe the clinical definitions of age-related frailty and their limitations in allowing us to understand the pathogenesis of this prevalent condition. Given that age-related frailty manifests in the form of functional declines such as poor balance, falls, and immobility, as an alternative we view frailty from a physiological viewpoint and describe what is known of the organ-based components of frailty, including adiposity, the brain, and neuromuscular, skeletal muscle, immune, and cardiovascular systems, as individual systems and as components in multisystem dysregulation. By doing so we aim to highlight current understanding of the physiological phenotype of frailty and reveal key knowledge gaps and potential mechanistic drivers of the trajectory to frailty. We also review the studies in humans that have intervened with exercise to reduce frailty. We conclude that more longitudinal and interventional clinical studies are required in older adults. Such observational studies should interrogate the progression from a nonfrail to a frail state, assessing individual elements of frailty to produce a deep physiological phenotype of the syndrome. The findings will identify mechanistic drivers of frailty and allow targeted interventions to diminish frailty progression.
Collapse
Affiliation(s)
- Joseph A Taylor
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Paul L Greenhaff
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - David B Bartlett
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, North Carolina.,Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Thomas A Jackson
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, https://ror.org/03angcq70University of Birmingham, Birmingham, United Kingdom
| | - Niharika A Duggal
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, https://ror.org/03angcq70University of Birmingham, Birmingham, United Kingdom
| | - Janet M Lord
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, https://ror.org/03angcq70University of Birmingham, Birmingham, United Kingdom.,NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
3
|
Guo Z, Valenzuela Ripoll C, Picataggi A, Rawnsley DR, Ozcan M, Chirinos JA, Chendamarai E, Girardi A, Riehl T, Evie H, Diab A, Kovacs A, Hyrc K, Ma X, Asnani A, Shewale SV, Scherrer-Crosbie M, Cowart LA, Parks JS, Zhao L, Gordon D, Ramirez-Valle F, Margulies KB, Cappola TP, Desai AA, Pedersen LN, Bergom C, Stitziel NO, Rettig MP, DiPersio JF, Hajny S, Christoffersen C, Diwan A, Javaheri A. Apolipoprotein M Attenuates Anthracycline Cardiotoxicity and Lysosomal Injury. JACC Basic Transl Sci 2023; 8:340-355. [PMID: 37034289 PMCID: PMC10077122 DOI: 10.1016/j.jacbts.2022.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 01/06/2023]
Abstract
Apolipoprotein M (ApoM) binds sphingosine-1-phosphate (S1P) and is inversely associated with mortality in human heart failure (HF). Here, we show that anthracyclines such as doxorubicin (Dox) reduce circulating ApoM in mice and humans, that ApoM is inversely associated with mortality in patients with anthracycline-induced heart failure, and ApoM heterozygosity in mice increases Dox-induced mortality. In the setting of Dox stress, our studies suggest ApoM can help sustain myocardial autophagic flux in a post-transcriptional manner, attenuate Dox cardiotoxicity, and prevent lysosomal injury.
Collapse
Affiliation(s)
- Zhen Guo
- Washington University School of Medicine, St Louis, Missouri, USA
| | | | | | | | - Mualla Ozcan
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Julio A. Chirinos
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Amanda Girardi
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Terrence Riehl
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Hosannah Evie
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Ahmed Diab
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Attila Kovacs
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Krzysztof Hyrc
- Hope Center, Washington University School of Medicine, St Louis, Missouri, USA
| | - Xiucui Ma
- Washington University School of Medicine, St Louis, Missouri, USA
- John Cochran Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Aarti Asnani
- Beth Israel Deaconess, Harvard Medical School, Boston, Massachusetts, USA
| | - Swapnil V. Shewale
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marielle Scherrer-Crosbie
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lauren Ashley Cowart
- Virginia Commonwealth University, Richmond, Virginia, USA
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, Virginia, USA
| | - John S. Parks
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Lei Zhao
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - David Gordon
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Kenneth B. Margulies
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas P. Cappola
- Perelman School of Medicine, University of Pennsylvania School of Medicine/Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Carmen Bergom
- Washington University School of Medicine, St Louis, Missouri, USA
| | | | | | - John F. DiPersio
- Washington University School of Medicine, St Louis, Missouri, USA
| | - Stefan Hajny
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Abhinav Diwan
- Washington University School of Medicine, St Louis, Missouri, USA
- John Cochran Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Ali Javaheri
- Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
4
|
Liu Y, Xiong Z, Zhou W, Chen Y, Huang Q, Wu Y. Role of apolipoprotein O in autophagy via the p38 mitogen-activated protein kinase signaling pathway in myocardial infarction. Clinics (Sao Paulo) 2022; 77:100046. [PMID: 35588578 PMCID: PMC9120058 DOI: 10.1016/j.clinsp.2022.100046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/10/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To explore the role and possible mechanisms of action of apolipoprotein O (APOO) in autophagy in Myocardial Infarction (MI) in vivo and in vitro. METHODS Differential gene expression and single Gene Set Enrichment Analysis (GSEA) were used to evaluate MI-related candidate genes. Animal and cell MI models were established. Sh-APOO, si-APOO, and SB203580 were used to inhibit the expression of APOO or p38MAPK. Western blot and qRT-PCR were used to analyze the expression levels of the target protein or mRNA. Apoptosis was observed using the TUNEL assay. The plasma concentrations of CK-MB and cTn-I in humans and mice were determined. RESULTS In the GSE23294 dataset, APOO mRNA was highly expressed in the left ventricle of mice with MI; GSEA revealed that APOO was positively correlated with p38MAPK, autophagy, and apoptosis. The plasma concentration of APOO in patients with MI was significantly higher than that in healthy subjects. The expression of APOO, Beclin-1, LC3, and Bax in mouse and AC16 cell MI models increased, while the level of Bcl-2 decreased. After silencing the APOO gene, the expression of APOO was downregulated; meanwhile, changes in autophagy, apoptosis and myocardial cell injury were reversed in vivo and in vitro. Furthermore, autophagy was alleviated after AC16 cells were treated with SB203580. CONCLUSIONS The increased APOO expression in mouse and cell MI models may activate autophagy and apoptosis by regulating the p38MAPK signaling pathway, thus aggravating the myocardial injury.
Collapse
Affiliation(s)
- Yue Liu
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Zhiping Xiong
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Wei Zhou
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Yuxin Chen
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Qing Huang
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Yanqing Wu
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China.
| |
Collapse
|
5
|
Zhang H, Lu J, Liu H, Guan L, Xu S, Wang Z, Qiu Y, Liu H, Peng L, Men X. Ajugol enhances TFEB-mediated lysosome biogenesis and lipophagy to alleviate non-alcoholic fatty liver disease. Pharmacol Res 2021; 174:105964. [PMID: 34732369 DOI: 10.1016/j.phrs.2021.105964] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/24/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022]
Abstract
Lipophagy is the autophagic degradation of lipid droplets. Dysregulated lipophagy has been implicated in the development of non-alcoholic fatty liver disease (NAFLD). Ajugol is an active alkaloid isolated from the root of Rehmannia glutinosa which is commonly used to treat various inflammatory and metabolic diseases. This study aimed to investigate the effect of ajugol on alleviating hepatic steatosis and sought to determine whether its potential mechanism via the key lysosome-mediated process of lipophagy. Our findings showed that ajugol significantly improved high-fat diet-induced hepatic steatosis in mice and inhibited palmitate-induced lipid accumulation in hepatocytes. Further analysis found that hepatic steatosis promoted the expression of LC3-II, an autophagosome marker, but led to autophagic flux blockade due to a lack of lysosomes. Ajugol also enhanced lysosomal biogenesis and promoted the fusion of autophagosome and lysosome to improve impaired autophagic flux and hepatosteatosis. Mechanistically, ajugol inactivated mammalian target of rapamycin and induced nuclear translocation of the transcription factor EB (TFEB), an essential regulator of lysosomal biogenesis. siRNA-mediated knockdown of TFEB significantly abrogated ajugol-induced lysosomal biogenesis as well as autophagosome-lysosome fusion and lipophagy. We conclude that lysosomal deficit is a critical mediator of hepatic steatosis, and ajugol may alleviate NAFLD via promoting the TFEB-mediated autophagy-lysosomal pathway and lipophagy.
Collapse
Affiliation(s)
- Heng Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China; School of Basic Medical Sciences, North China University of Science and Technology, No. 21 Bohai Road, Caofeidian District, Tangshan, Hebei 063210, China
| | - Junfeng Lu
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China; Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Hao Liu
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China
| | - Lingling Guan
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China
| | - Shiqing Xu
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China
| | - Zai Wang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China
| | - Yang Qiu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Honglin Liu
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China
| | - Liang Peng
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China.
| | - Xiuli Men
- School of Basic Medical Sciences, North China University of Science and Technology, No. 21 Bohai Road, Caofeidian District, Tangshan, Hebei 063210, China.
| |
Collapse
|
6
|
Kirk JA, Cheung JY, Feldman AM. Therapeutic targeting of BAG3: considering its complexity in cancer and heart disease. J Clin Invest 2021; 131:e149415. [PMID: 34396980 DOI: 10.1172/jci149415] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Bcl2-associated athanogene-3 (BAG3) is expressed ubiquitously in humans, but its levels are highest in the heart, the skeletal muscle, and the central nervous system; it is also elevated in many cancers. BAG3's diverse functions are supported by its multiple protein-protein binding domains, which couple with small and large heat shock proteins, members of the Bcl2 family, other antiapoptotic proteins, and various sarcomere proteins. In the heart, BAG3 inhibits apoptosis, promotes autophagy, couples the β-adrenergic receptor with the L-type Ca2+ channel, and maintains the structure of the sarcomere. In cancer cells, BAG3 binds to and supports an identical array of prosurvival proteins, and it may represent a therapeutic target. However, the development of strategies to block BAG3 function in cancer cells may be challenging, as they are likely to interfere with the essential roles of BAG3 in the heart. In this Review, we present the current knowledge regarding the biology of this complex protein in the heart and in cancer and suggest several therapeutic options.
Collapse
Affiliation(s)
- Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, USA
| | - Joseph Y Cheung
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Arthur M Feldman
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Hsu CC, Cheng KC, Li Y, Hsu PH, Cheng JT, Niu HS. TGR5 Expression Is Associated with Changes in the Heart and Urinary Bladder of Rats with Metabolic Syndrome. Life (Basel) 2021; 11:695. [PMID: 34357066 PMCID: PMC8306239 DOI: 10.3390/life11070695] [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: 05/25/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 11/19/2022] Open
Abstract
Adipose-derived cytokines may contribute to the inflammation that occurs in metabolic syndrome (MetS). The Takeda G protein-coupled receptor (TGR5) regulates energy expenditure and affects the production of pro-inflammatory biomarkers in metabolic diseases. Etanercept, which acts as a tumor necrosis factor (TNF)-α antagonist, can also block the inflammatory response. Therefore, the interaction between TNF-α and TGR5 expression was investigated in rats with high-fat diet (HFD)-induced obesity. Heart tissues isolated from the HFD-induced MetS rats were analyzed. Changes in TGR5 expression were investigated with lithocholic acid (LCA) as the agonist. Betulinic acid (BA) was used to activate TGR5 in urinary bladders. LCA was more effective in the heart tissues of HFD-fed rats, although etanercept alleviated the function of LCA. STAT3 activation and higher TGR5 expression were observed in the heart tissues collected from HFD-fed rats. Thus, cardiac TGR5 expression is promoted by HFD through STAT3 activation in rats. Moreover, the urinary bladders of female rats fed a HFD showed a low response, which was reversed by etanercept. Relaxation by BA in the bladders was more marked in HFD-fed rats. The high TGR5 expression in HFD-fed rats was characterized using a mRNA assay, and the increased cAMP levels were found to be stimulated by BA in the isolated bladders. Therefore, TGR5 expression increases with a HFD in both the hearts and urinary bladders. Collectively, cytokine-medicated TGR5 activation was observed in the hearts and urinary bladders of rats.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Taoyuan City 33303, Taiwan;
- Department of Otorhinolaryngology, Taipei City Hospital, Taipei City 10341, Taiwan
- Department of Exercise and Health Sciences, University of Taipei, Taipei City 11153, Taiwan
| | - Kai-Chun Cheng
- Department of Pharmacy, College of Pharmacy, Tajen University, Pingtung 90741, Taiwan;
- Pharmacological Department of Herbal Medicine, Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Yingxiao Li
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien, 970302, Taiwan;
| | - Ping-Hao Hsu
- School of Medicine, Chung Shan Medical University, Taichung City 40201, Taiwan;
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Tainan City 71004, Taiwan;
| | - Ho-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien, 970302, Taiwan;
| |
Collapse
|
8
|
Xue Y, Yu X, Zhang X, Yu P, Li Y, Fu W, Yu J, Sui D. Protective effects of ginsenoside Rc against acute cold exposure-induced myocardial injury in rats. J Food Sci 2021; 86:3252-3264. [PMID: 34146399 DOI: 10.1111/1750-3841.15757] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/26/2021] [Accepted: 04/09/2021] [Indexed: 11/30/2022]
Abstract
Ginsenoside Rc is one of the cardinal bioactive components of Panax ginseng. The present study aimed to investigate whether ginsenoside Rc exerted protective effects against acute cold exposure-induced myocardial injury in rats. Forty rats were randomly assigned into four groups: Control, model, ginsenoside Rc 10 mg/kg, and 20 mg/kg groups. Rats were intragastrically administrated with ginsenoside Rc (10, 20 mg/kg) or vehicle daily for 7 days. On the seventh day, all rats except the control group were exposed to low temperature. Cardiac function, myocardial enzyme activities, hemorheology, and inflammatory response were detected. Histopathological examination and apoptosis of cardiac tissues were performed. The expressions of silent information regulator 1 (SIRT1), B-cell lymphoma (Bcl-2), Bcl-2-associated X (Bax), procaspase-3, and the mRNA (messenger RNA) level of SIRT1 were measured by western blot and real-time quantitative polymerase chain reaction (PCR) analysis. Ginsenoside Rc significantly improved cardiac function, diminished the activities of lactate dehydrogenase (LDH), aspartate aminotransferase, and creatine kinase isoenzyme (CK-MB), and regulated abnormal hemorheology in acute cold-exposed rats (p < 0.05 or p < 0.01). Furthermore, ginsenoside Rc could attenuate myocardial histological changes and structural abnormalities, decrease apoptotic cells and reduce the mRNA levels and activity of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 (p < 0.01). In addition, ginsenoside Rc upregulated the expressions of SIRT1, Bcl-2, and procaspase-3 and downregulated that of Bax (p < 0.01). The changes in both the mRNA and protein expression levels of SIRT1 were similar. The results of the current study suggested that ginsenoside Rc could alleviate acute cold exposure-induced myocardial injury in rats by inhibiting cardiomyocyte apoptosis via regulating SIRT1 expression and attenuating the inflammatory responses. PRACTICAL APPLICATION: The current study indicated that ginsenoside Rc could alleviate acute cold exposure-induced myocardial injury in rats. Ginsenoside Rc could be potentially used as a bioactive ingredient in processed functional food products or food supplements to prevent from acute cold exposure-induced myocardial injury.
Collapse
Affiliation(s)
- Yan Xue
- The Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, PR China.,The Department of Burn Surgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Xiaofeng Yu
- The Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, PR China
| | - Xiuhang Zhang
- The Department of Burn Surgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Ping Yu
- The Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, PR China
| | - Yuangeng Li
- The Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, PR China
| | - Wenwen Fu
- The Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, PR China
| | - Jiaao Yu
- The Department of Burn Surgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Dayun Sui
- The Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, PR China
| |
Collapse
|
9
|
Trial J, Diaz Lankenau R, Angelini A, Tovar Perez JE, Taffet GE, Entman ML, Cieslik KA. Treatment with a DC-SIGN ligand reduces macrophage polarization and diastolic dysfunction in the aging female but not male mouse hearts. GeroScience 2021; 43:881-899. [PMID: 32851570 PMCID: PMC8110645 DOI: 10.1007/s11357-020-00255-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022] Open
Abstract
Cardiac diastolic dysfunction in aging arises from increased ventricular stiffness caused by inflammation and interstitial fibrosis. The diastolic dysfunction contributes to heart failure with preserved ejection fraction (HFpEF), which in the aging population is more common in women. This report examines its progression over 12 weeks in aging C57BL/6J mice and correlates its development with changes in macrophage polarization and collagen deposition.Aged C57BL/6J mice were injected with dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) ligand 1 (DCSL1, an anti-inflammatory agent) or saline for 12 weeks. Echo and Doppler measurements were performed before and after 4 and 12 weeks of treatment. DCSL1 prevented the worsening of diastolic dysfunction over time in females but not in males. Cardiac single cell suspensions analyzed by flow cytometry revealed changes in the inflammatory infiltrate: (1) in males, there was an increased total number of leukocytes with an increased pro-inflammatory profile compared with females and they did not respond to DCSL1; (2) by contrast, DCSL1 treatment resulted in a shift in macrophage polarization to an anti-inflammatory phenotype in females. Notably, DCSL1 preferentially targeted tumor necrosis factor-α (TNFα+) pro-inflammatory macrophages. The reduction in pro-inflammatory macrophage polarization was accompanied by a decrease in collagen content in the heart.Age-associated diastolic dysfunction in mice is more severe in females and is associated with unique changes in macrophage polarization in cardiac tissue. Treatment with DCSL1 mitigates the changes in inflammation, cardiac function, and fibrosis. The characteristics of diastolic dysfunction in aging female mice mimic similar changes in aging women.
Collapse
Affiliation(s)
- JoAnn Trial
- Department of Medicine, Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM 620, Houston, TX, 77030, USA
| | - Rodrigo Diaz Lankenau
- Department of Medicine, Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM 620, Houston, TX, 77030, USA
| | - Aude Angelini
- Department of Medicine, Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM 620, Houston, TX, 77030, USA
| | - Jorge E Tovar Perez
- Department of Medicine, Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM 620, Houston, TX, 77030, USA
- Texas A&M University, 2121 W. Holcombe Blvd, Houston, TX, 77030, USA
| | - George E Taffet
- Department of Medicine, Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM 620, Houston, TX, 77030, USA
- The DeBakey Heart Center, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Mark L Entman
- Department of Medicine, Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM 620, Houston, TX, 77030, USA
- The DeBakey Heart Center, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Katarzyna A Cieslik
- Department of Medicine, Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM 620, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Mubagwa K. Cardiac effects and toxicity of chloroquine: a short update. Int J Antimicrob Agents 2020; 56:106057. [PMID: 32565195 PMCID: PMC7303034 DOI: 10.1016/j.ijantimicag.2020.106057] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/07/2020] [Accepted: 06/14/2020] [Indexed: 02/07/2023]
Abstract
There is currently increased interest in the use of the antimalarial drugs chloroquine and hydroxychloroquine for the treatment of other diseases, including cancer and viral infections such as coronavirus disease 2019 (COVID-19). However, the risk of cardiotoxic effects tends to limit their use. In this review, the effects of these drugs on the electrical and mechanical activities of the heart as well as on remodelling of cardiac tissue are presented and the underlying molecular and cellular mechanisms are discussed. The drugs can have proarrhythmic as well as antiarrhythmic actions resulting from their inhibition of ion channels, including voltage-dependent Na+ and Ca2+ channels, background and voltage-dependent K+ channels, and pacemaker channels. The drugs also exert a vagolytic effect due at least in part to a muscarinic receptor antagonist action. They also interfere with normal autophagy flux, an effect that could aggravate ischaemia/reperfusion injury or post-infarct remodelling. Most of the toxic effects occur at high concentrations, following prolonged drug administration or in the context of drug associations.
Collapse
Affiliation(s)
- Kanigula Mubagwa
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Basic Sciences, Faculty of Medicine, Université Catholique de Bukavu, Bukavu, DR Congo.
| |
Collapse
|
11
|
Wang SY, Ni X, Hu KQ, Meng FL, Li M, Ma XL, Meng TT, Wu HH, Ge D, Zhao J, Li Y, Su GH. Cilostazol alleviate nicotine induced cardiomyocytes hypertrophy through modulation of autophagy by CTSB/ROS/p38MAPK/JNK feedback loop. Int J Biol Sci 2020; 16:2001-2013. [PMID: 32398966 PMCID: PMC7211170 DOI: 10.7150/ijbs.43825] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022] Open
Abstract
Nicotine is proved to be an important factor for cardiac hypertrophy. Autophagy is important cell recycling system involved in the regulation of cardiac hypertrophy. Cilostazol, which is often used in the management of peripheral vascular disease. However, the effects of cilostazol on nicotine induced autophagy and cardiac hypertrophy are unclear. Here, we aim to determine the role and molecular mechanism of cilostazol in alleviating nicotine-induced cardiomyocytes hypertrophy through modulating autophagy and the underlying mechanisms. Our results clarified that nicotine stimulation caused cardiomyocytes hypertrophy and autophagy flux impairment significantly in neonatal rat ventricular myocytes (NRVMs), which were evidenced by augments of LC3-II and p62 levels, and impaired autophagosomes clearance. Interestingly, cathepsin B (CTSB) activity decreased dramatically after stimulation with nicotine in NRVMs, which was crucial for substrate degradation in the late stage of autophagy process, and cilostazol could reverse this effect dramatically. Intracellular ROS levels were increased significantly after nicotine exposure. Meanwhile, p38MAPK and JNK were activated after nicotine treatment. By using ROS scavenger N-acetyl-cysteine (NAC) could reverse the effects of nicotine by down-regulation the phosphorylation of p38MAPK and JNK pathways, and pretreatment of specific inhibitors of p38MAPK and JNK could restore the autophagy impairment and cardiomyocytes hypertrophy induced by nicotine. Moreover, CTSB activity of lysosome regained after the treatment with cilostazol. Cilostazol also inhibited the ROS accumulation and the activation of p38MAPK and JNK, which providing novel connection between lysosome CTSB and ROS/p38MAPK/JNK related oxidative stress pathway. This is the first demonstration that cilostazol could alleviate nicotine induced cardiomyocytes hypertrophy through restoration of autophagy flux by activation of CTSB and inhibiting ROS/p38/JNK pathway, exhibiting a feedback loop on regulation of autophagy and cardiomyocytes hypertrophy.
Collapse
Affiliation(s)
- Shu-Ya Wang
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xi Ni
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ke-Qing Hu
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fan-Liang Meng
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Min Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xiao-Li Ma
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ting-Ting Meng
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Hui-Hui Wu
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Di Ge
- School of Biological Science and Technology, University of Jinan, China
| | - Jing Zhao
- Development Biology, School of Life Science, Shandong University, Jinan, China
| | - Ying Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guo-Hai Su
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
12
|
CaMKII Activity in the Inflammatory Response of Cardiac Diseases. Int J Mol Sci 2019; 20:ijms20184374. [PMID: 31489895 PMCID: PMC6770001 DOI: 10.3390/ijms20184374] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022] Open
Abstract
Inflammation is a physiological process by which the body responds to external insults and stress conditions, and it is characterized by the production of pro-inflammatory mediators such as cytokines. The acute inflammatory response is solved by removing the threat. Conversely, a chronic inflammatory state is established due to a prolonged inflammatory response and may lead to tissue damage. Based on the evidence of a reciprocal regulation between inflammation process and calcium unbalance, here we described the involvement of a calcium sensor in cardiac diseases with inflammatory drift. Indeed, the Ca2+/calmodulin-dependent protein kinase II (CaMKII) is activated in several diseases with an inflammatory component, such as myocardial infarction, ischemia/reperfusion injury, pressure overload/hypertrophy, and arrhythmic syndromes, in which it actively regulates pro-inflammatory signaling, among which includes nuclear factor kappa-B (NF-κB), thus contributing to pathological cardiac remodeling. Thus, CaMKII may represent a key target to modulate the severity of the inflammatory-driven degeneration.
Collapse
|
13
|
Bartekova M, Radosinska J, Jelemensky M, Dhalla NS. Role of cytokines and inflammation in heart function during health and disease. Heart Fail Rev 2019; 23:733-758. [PMID: 29862462 DOI: 10.1007/s10741-018-9716-x] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
By virtue of their actions on NF-κB, an inflammatory nuclear transcription factor, various cytokines have been documented to play important regulatory roles in determining cardiac function under both physiological and pathophysiological conditions. Several cytokines including TNF-α, TGF-β, and different interleukins such as IL-1 IL-4, IL-6, IL-8, and IL-18 are involved in the development of various inflammatory cardiac pathologies, namely ischemic heart disease, myocardial infarction, heart failure, and cardiomyopathies. In ischemia-related pathologies, most of the cytokines are released into the circulation and serve as biological markers of inflammation. Furthermore, there is an evidence of their direct role in the pathogenesis of ischemic injury, suggesting cytokines as potential targets for the development of some anti-ischemic therapies. On the other hand, certain cytokines such as IL-2, IL-4, IL-6, IL-8, and IL-10 are involved in the post-ischemic tissue repair and thus are considered to exert beneficial effects on cardiac function. Conflicting reports regarding the role of some cytokines in inducing cardiac dysfunction in heart failure and different types of cardiomyopathies seem to be due to differences in the nature, duration, and degree of heart disease as well as the concentrations of some cytokines in the circulation. In spite of extensive research work in this field of investigation, no satisfactory anti-cytokine therapy for improving cardiac function in any type of heart disease is available in the literature.
Collapse
Affiliation(s)
- Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Jana Radosinska
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Marek Jelemensky
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada. .,Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
14
|
Karlstaedt A, Schiffer W, Taegtmeyer H. Actionable Metabolic Pathways in Heart Failure and Cancer-Lessons From Cancer Cell Metabolism. Front Cardiovasc Med 2018; 5:71. [PMID: 29971237 PMCID: PMC6018530 DOI: 10.3389/fcvm.2018.00071] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/24/2018] [Indexed: 12/21/2022] Open
Abstract
Recent advances in cancer cell metabolism provide unprecedented opportunities for a new understanding of heart metabolism and may offer new approaches for the treatment of heart failure. Key questions driving the cancer field to understand how tumor cells reprogram metabolism and to benefit tumorigenesis are also applicable to the heart. Recent experimental and conceptual advances in cancer cell metabolism provide the cardiovascular field with the unique opportunity to target metabolism. This review compares cancer cell metabolism and cardiac metabolism with an emphasis on strategies of cellular adaptation, and how to exploit metabolic changes for therapeutic benefit.
Collapse
Affiliation(s)
- Anja Karlstaedt
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Walter Schiffer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|