1
|
Zhang BY, He GL, Wang ZZ, Zhou H, Huang XY, Shen TT, Liu XQ, Liu YS, Luo Z, Li P, Tan YL, Luo X, Yang XS. L-carnitine ameliorates myocardial injury by alleviating endoplasmic reticulum stress via inhibition of PERK pathway in exertional heatstroke rats. Heliyon 2024; 10:e40502. [PMID: 39641050 PMCID: PMC11617224 DOI: 10.1016/j.heliyon.2024.e40502] [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: 08/04/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024] Open
Abstract
Exertional heatstroke (EHS) is a life-threatening condition with potential for tissues and organs injury, including heart. Effective drug strategies to treat patients with EHS are warranted to unlock the therapeutic potential. Considering the cardioprotective effects of L-carnitine (LC), this study aimed to investigate the effects of LC on EHS-induced myocardial injury in rats and to explore the underlying mechanisms. Here, we found that LC exerted a greater protective effect on EHS-induced cardiac dysfunction and mortality, which also significantly attenuated certain negative effects, including increased myocardial apoptosis, pathological changes, and ultrastructural impairment, enhanced activity levels of such serum enzymes as AST, LDH, CK, and CK-MB, reduced BCL-2 expression, increased the expression of cleaved caspase-3 and the critical endoplasmic reticulum stress (ERS) indices like CHOP and GRP78 in EHS rats. Besides, pretreatment of EHS rats with PBA (4-Phenyl butyric acid), a chemical chaperone that attenuates ERS, restored BCL-2 expression, reduced the protein levels of cleaved caspase-3, CHOP, and GRP78. Furthermore, thapsigargin (TG), which induces ERS, enhanced the expression of BAX, cleaved caspase-3, CHOP, and GRP78, attenuated BCL-2 expression, and enhanced mitochondrial impairment in EHS + LC rats. Mechanismly, the protective effects of LC were mediated, at least partly, by inhibiting the activation of PERK pathway against ERS-associated myocardial damage. These results indicate that supplementation of LC might be a potential strategy to reduce myocardial injury by affecting ERS via inhibiting the PERK pathway against EHS.
Collapse
Affiliation(s)
- Bo-Yi Zhang
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Gen-Lin He
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Ze-Ze Wang
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Huan Zhou
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Xue-Yan Huang
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Ting-Ting Shen
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Xiao-Qian Liu
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Yi-Shan Liu
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Zhen Luo
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Ping Li
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Yu-Long Tan
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Xue Luo
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Xue-Sen Yang
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Wu Q, Yao J, Xiao M, Zhang X, Zhang M, Xi X. Targeting Nrf2 signaling pathway: new therapeutic strategy for cardiovascular diseases. J Drug Target 2024; 32:874-883. [PMID: 38753446 DOI: 10.1080/1061186x.2024.2356736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally, with oxidative stress (OS) identified as a primary contributor to their onset and progression. Given the elevated incidence and mortality rates associated with CVDs, there is an imperative need to investigate novel therapeutic strategies. Nuclear factor erythroid 2-related factor 2 (Nrf2), ubiquitously expressed in the cardiovascular system, has emerged as a promising therapeutic target for CVDs due to its role in regulating OS and inflammation. This review aims to delve into the mechanisms and actions of the Nrf2 pathway, highlighting its potential in mitigating the pathogenesis of CVDs.
Collapse
Affiliation(s)
- Qi Wu
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| | - Jiangting Yao
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| | - Mengyun Xiao
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| | - Xiawei Zhang
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| | - Mengxiao Zhang
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Xinting Xi
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| |
Collapse
|
3
|
El Feky W, El-Afify D, Abdelhai D, Elkashlan M, Fakhreldin A, El Amrousy D. L-carnitine decreases myocardial injury in children undergoing open-heart surgery: A randomized controlled trial. Eur J Pediatr 2024; 183:2783-2789. [PMID: 38568244 PMCID: PMC11098932 DOI: 10.1007/s00431-024-05534-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 05/19/2024]
Abstract
Myocardial injury in open-heart surgery is related to several factors including ischemia-reperfusion injury, generation of reactive oxygen species, increased production of inflammatory mediators, and enhancement of apoptosis of cardiomyocytes. The aim of this study was to study the effect of L-carnitine on myocardial injury in children undergoing open-heart surgery. This clinical trial was performed on 60 children with congenital heart disease (CHD) who underwent open-heart surgery. They were randomized into two groups: L-carnitine group who received L-carnitine 50 mg\kg\day once daily for 1 month before cardiac surgery and control group who received placebo for 1 month before cardiac surgery. Left ventricular cardiac function was assessed by conventional echocardiography to measure left ventricular ejection fraction (LVEF) and two-dimensional speckle tracking echocardiography (2D-STE) to determine left ventricular global longitudinal strain (2D-LV GLS). Blood samples were obtained pre-operatively at baseline before the administration of L-carnitine or placebo and 12 h post-operatively to measure the level of malondialdehyde (MDA), superoxide dismutase (SOD), fas, caspase-3, creatinine kinase-MB (CK-MB), and troponin I. L-carnitine group had significantly lower post-operative level of oxidative stress marker (MDA), apoptosis markers (fas and caspase-3), and myocardial injury markers (CK-MB and troponin I), but they had significantly higher SOD post-operative level compared to the control group. In addition, post-operative LVEF and 2D-LVGLS were significantly lower in the control group compared to L-carnitine group. Conclusion: L-carnitine can reduce myocardial injury, improve post-operative left ventricular cardiac function, and may provide myocardium protection in children with CHD who underwent open-heart surgery. Trial registration: The clinical trial was registered at www.pactr.org with registration number PACTR202010570607420 at 29/10/2020 before recruiting the patients. What is Known: • Myocardial injury in open-heart surgery is related to several factors including ischemia-reperfusion injury, generation of reactive oxygen species, increased production of inflammatory mediators, and enhancement of apoptosis of cardiomyocytes. • L-carnitine was reported to have myocardial protective effects in rheumatic valvular surgery and coronary artery bypass graft (CABG) in adults; however, there is no evidence on its effectiveness in children undergoing open-heart surgery. What is New: • L-carnitine significantly lowered the post-operative level of oxidative stress marker (MDA), apoptosis markers (fas and caspase-3), and myocardial injury markers (CK-MB and troponin I) in the treatment group. • L-carnitine can reduce myocardial injury, improve post-operative left ventricular cardiac function, and may provide myocardium protection in children with CHD who underwent open-heart surgery.
Collapse
Affiliation(s)
- Wael El Feky
- Cardiothoracic Surgery Department, Faculty of Medicine, Kafr Elsheikh University, Kafr Elsheikh, Egypt
| | - Dalia El-Afify
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Dina Abdelhai
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed Elkashlan
- Anesthesia Department, Elmenshawy Hospital, Ministry of Health, Tanta, Egypt
| | - Ahmed Fakhreldin
- Pediatric Department, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Doaa El Amrousy
- Pediatric Department, Faculty of Medicine, Tanta University, Tanta, Egypt.
| |
Collapse
|
4
|
Wong CN, Gui XY, Rabkin SW. Myeloperoxidase, carnitine, and derivatives of reactive oxidative metabolites in heart failure with preserved versus reduced ejection fraction: A meta-analysis. Int J Cardiol 2024; 399:131657. [PMID: 38101703 DOI: 10.1016/j.ijcard.2023.131657] [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: 07/12/2023] [Revised: 11/03/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND Understanding the pathophysiology of heart failure (HF) with preserved ejection fraction (HFpEF) continues to be challenging. Several inflammatory and metabolic biomarkers have recently been suggested to be involved in HFpEF. OBJECTIVES The purpose of this review was to synthesize the evidence on non-traditional biomarkers from metabolomic studies that may distinguish HFpEF from heart failure with reduced ejection fraction (HFrEF) and controls without HF. METHODS A systematic search was conducted using Medline and PubMed with search terms such as "HFpEF" and "metabolomics", and a meta-analysis was conducted. RESULTS Myeloperoxidase (MPO) levels were significantly (p < 0.001) higher in HFpEF than controls without HF, but comparable (p = 0.838) between HFpEF and HFrEF. Carnitine levels were significantly (p < 0.0001) higher in HFrEF than HFpEF, but comparable (p = 0.443) between HFpEF and controls without HF. Derivatives of reactive oxidative metabolites (DROMs) were not significantly (p = 0.575) higher in HFpEF than controls without HF. CONCLUSION These data suggest that MPO is operative in HFpEF and HFrEF and may be a biomarker for HF. Furthermore, circulating carnitine levels may distinguish HFrEF from HFpEF.
Collapse
Affiliation(s)
- Chenille N Wong
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Xi Yao Gui
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Simon W Rabkin
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Division of Cardiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
5
|
Farag A, Elfadadny A, Mandour AS, Ngeun SK, Aboubakr M, Kaneda M, Tanaka R. Potential protective effects of L-carnitine against myocardial ischemia/reperfusion injury in a rat model. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:18813-18825. [PMID: 38349499 DOI: 10.1007/s11356-024-32212-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 01/22/2024] [Indexed: 03/09/2024]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a growing concern for global public health. This study seeks to explore the potential protective effects of L-carnitine (LC) against heart ischemia-reperfusion injury in rats. To induce I/R injury, the rat hearts underwent a 30-min ligation of the left anterior descending coronary artery, followed by 24 h of reperfusion. We evaluated cardiac function through electrocardiography and heart rate variability (HRV) and conducted pathological examinations of myocardial structure. Additionally, the study investigated the influence of LC on myocardial apoptosis, inflammation, and oxidative stress in the context of I/R injury. The results show that pretreatment with LC led to improvements in the observed alterations in ECG waveforms and HRV parameters in the nontreated ischemic reperfusion model group, although most of these changes did not reach statistical significance. Similarly, although without a significant difference, LC reduced the levels of proinflammatory cytokines when compared to the values in the nontreated ischemic rat group. Furthermore, LC restored the reduced expressions of SOD1, SOD2, and SOD3. Additionally, LC significantly reduced the elevated Bax expressions and showed a nonsignificant increase in Bcl-2 expression, resulting in a favorable adjustment of the Bcl-2/Bax ratio. We also observed a significant enhancement in the histological appearance of cardiac muscles, a substantial reduction in myocardial fibrosis, and suppressed CD3 + cell proliferation in the ischemic myocardium. This small-scale, experimental, in vivo study indicates that LC was associated with enhancements in the pathological findings in the ischemic myocardium in the context of ischemia/reperfusion injury in this rat model. Although statistical significance was not achieved, LC exhibits potential and beneficial protective effects against I/R injury. It does so by modulating the expression of antioxidative and antiapoptotic genes, inhibiting the inflammatory response, and enhancing autonomic balance, particularly by increasing vagal tone in the heart. Further studies are necessary to confirm and elaborate on these findings.
Collapse
Affiliation(s)
- Ahmed Farag
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan.
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| | - Ahmed Elfadadny
- Department of Animal Internal Medicine, Faculty of Veterinary Medicine, Damanhur University, Damanhur, Egypt
| | - Ahmed S Mandour
- Department of Animal Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Sai Koung Ngeun
- Laboratory of Veterinary Diagnostic Imaging, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Mohamed Aboubakr
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Qaliobiya, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryou Tanaka
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| |
Collapse
|
6
|
Zhang L, Xiu L, Wang T, Zhao D. Effect of L-carnitine in Ameliorating Lipopolysaccharide-Induced Cardiomyocyte Injury via MAPK Signaling. Mol Biotechnol 2024; 66:79-89. [PMID: 37029860 DOI: 10.1007/s12033-023-00731-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/22/2023] [Indexed: 04/09/2023]
Abstract
The present study aimed to elucidate whether L-carnitine (LC) protected H9c2 cells and its underlying mechanisms. Cell counting kit-8 (CCK-8) assay was used to evaluate cell viability. Apoptosis, cell morphology, and lactate dehydrogenase (LDH) assessment were used to prove effects of lipopolysaccharide (LPS) and LC on H9c2 cells. RT-qPCR and western blot assays were hired to evaluate the mRNA and protein expression levels, respectively. ELISA assay was performed to determine the released protein levels. Reactive oxygen species (ROS) level was evaluated by immunofluorescence and flow cytometry. LC was revealed to protect H9c2 cells against LPS-induced injury as indicated by increased cell viability, reduced apoptosis ratio and LDH level. LC treatment also reduced BAX expression as well as up-regulated Bcl-2 expression under LPS treatment. Mechanically, LC reduced oxidative stress and ameliorated the mitochondrial injury through modulating extracellular signal-regulated kinase 1/2 and c-Jun N-terminal protein kinase c-Jun N-terminal protein kinase phosphorylation levels as indicated by decreased membrane potential, increased ATP production and mtDNA expression. We found that LC ameliorates LPS-induced cardiomyocyte injury by abrogating cell apoptosis ratio, ROS levels, as well as mitochondrial dysfunction via mitogen-activated protein kinase signaling. Our findings revealed a potential drug for sepsis or LPS-induced cardiomyocyte injury.
Collapse
Affiliation(s)
- Li Zhang
- Medical College, Internal Medicine Teaching and Research Office, Zhengzhou University of Industry Technology, Zhengzhou, Henan, China
- Internal Medicine-Cardiovascular Department, Xinzheng Huaxin Minsheng Hospital, Zhengzhou, Henan, China
| | - Lei Xiu
- Medical College, Zhengzhou University of Industry Technology, Zhengzhou, Henan, China
| | - Taoli Wang
- Medical College, Zhengzhou University of Industry Technology, Zhengzhou, Henan, China
| | - Duo Zhao
- Radiology Department, Public People's Hospital of Xinzheng, 2000 Meters South of the Intersection of South China Road and Yanhuang Avenue, Xinzheng, 451100, Henan, China.
| |
Collapse
|
7
|
Han X, Wang H, Du F, Zeng X, Guo C. Nrf2 for a key member of redox regulation: A novel insight against myocardial ischemia and reperfusion injuries. Biomed Pharmacother 2023; 168:115855. [PMID: 37939614 DOI: 10.1016/j.biopha.2023.115855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023] Open
Abstract
Nuclear factor erythroid-2 related factor 2 (Nrf2), a nuclear transcription factor, modulates genes responsible for antioxidant responses against toxic and oxidative stress to maintain redox homeostasis and participates in varieties of cellular processes such as metabolism and inflammation during myocardial ischemia and reperfusion injuries (MIRI). The accumulation of reactive oxygen species (ROS) from damaged mitochondria, xanthine oxidase, NADPH oxidases, and inflammation contributes to depraved myocardial ischemia and reperfusion injuries. Considering that Nrf2 played crucial roles in antagonizing oxidative stress, it is reasonable to delve into the up or down-regulated molecular mechanisms of Nrf2 in the progression of MIRI to provide the possibility of new therapeutic medicine targeting Nrf2 in cardiovascular diseases. This review systematically describes the generation of ROS, the regulatory metabolisms of Nrf2 as well as several natural or synthetic compounds activating Nrf2 during MIRI, which might provide novel insights for the anti-oxidative stress and original ideas targeting Nrf2 for the prevention and treatment in cardiovascular diseases.
Collapse
Affiliation(s)
- Xuejie Han
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China
| | - Hongxia Wang
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China
| | - Fenghe Du
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing 100070, PR China
| | - Xiangjun Zeng
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China.
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China.
| |
Collapse
|
8
|
Gökçe Y, Danisman B, Akcay G, Derin N, Yaraş N. L-Carnitine improves mechanical responses of cardiomyocytes and restores Ca 2+ homeostasis during aging. Histochem Cell Biol 2023; 160:341-347. [PMID: 37329457 DOI: 10.1007/s00418-023-02215-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 06/19/2023]
Abstract
L-Carnitine (β-hydroxy-γ-trimethylaminobutyric acid, LC) is a crucial molecule for the mitochondrial oxidation of fatty acids. It facilitates the transport of long-chain fatty acids into the mitochondrial matrix. The reduction in LC levels during the aging process has been linked to numerous cardiovascular disorders, including contractility dysfunction, and disrupted intracellular Ca2+ homeostasis. The aim of this study was to examine the effects of long-term (7 months) LC administration on cardiomyocyte contraction and intracellular Ca2+ transients ([Ca2+]i) in aging rats. Male albino Wistar rats were randomly assigned to either the control or LC-treated groups. LC (50 mg/kg body weight/day) was dissolved in distilled water and orally administered for a period of 7 months. The control group received distilled water alone. Subsequently, ventricular single cardiomyocytes were isolated, and the contractility and Ca2+ transients were recorded in aging (18 months) rats. This study demonstrates, for the first time, a novel inotropic effect of long-term LC treatment on rat ventricular cardiomyocyte contraction. LC increased cardiomyocyte cell shortening and resting sarcomere length. Furthermore, LC supplementation led to a reduction in resting [Ca2+]i level and an increase in the amplitude of [Ca2+]i transients, indicative of enhanced contraction. Consistent with these results, decay time of Ca2+ transients also decreased significantly in the LC-treated group. The long-term administration of LC may help restore the Ca2+ homeostasis altered during aging and could be used as a cardioprotective medication in cases where myocyte contractility is diminished.
Collapse
Affiliation(s)
- Yasin Gökçe
- Faculty of Medicine, Department of Biophysics, Harran University, Sanliurfa, Turkey.
| | - Betul Danisman
- Faculty of Medicine, Department of Biophysics, Ataturk University, Erzurum, Turkey
| | - Guven Akcay
- Faculty of Medicine, Department of Biophysics, Hitit University, Corum, Turkey
| | - Narin Derin
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| | - Nazmi Yaraş
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| |
Collapse
|
9
|
Chen KY, Liu Z, Yi J, Hui YP, Song YN, Lu JH, Chen HJ, Yang SY, Hu XY, Zhang DS, Liang GY. PDHA1 Alleviates Myocardial Ischemia-Reperfusion Injury by Improving Myocardial Insulin Resistance During Cardiopulmonary Bypass Surgery in Rats. Cardiovasc Drugs Ther 2023:10.1007/s10557-023-07501-9. [PMID: 37610688 DOI: 10.1007/s10557-023-07501-9] [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] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVE Cardiopulmonary bypass (CPB) is a requisite technique for thoracotomy in advanced cardiovascular surgery. However, the consequent myocardial ischemia-reperfusion injury (MIRI) is the primary culprit behind cardiac dysfunction and fatal consequences post-operation. Prior research has posited that myocardial insulin resistance (IR) plays a vital role in exacerbating the progression of MIRI. Nonetheless, the exact mechanisms underlying this phenomenon remain obscure. METHODS We constructed pyruvate dehydrogenase E1 α subunit (PDHA1) interference and overexpression rats and used ascending aorta occlusion in an in vivo model of CPB-MIRI. We devised an in vivo model of CPB-MIRI by constructing rat models with both pyruvate dehydrogenase E1α subunit (PDHA1) interference and overexpression through ascending aorta occlusion. We analyzed myocardial glucose metabolism and the degree of myocardial injury using functional monitoring, biochemical assays, and histological analysis. RESULTS We discovered a clear downregulation of glucose transporter 4 (GLUT4) protein content expression in the CPB I/R model. In particular, cardiac-specific PDHA1 interference resulted in exacerbated cardiac dysfunction, significantly increased myocardial infarction area, more pronounced myocardial edema, and markedly increased cardiomyocyte apoptosis. Notably, the opposite effect was observed with PDHA1 overexpression, leading to a mitigated cardiac dysfunction and decreased incidence of myocardial infarction post-global ischemia. Mechanistically, PDHA1 plays a crucial role in regulating the protein content expression of GLUT4 on cardiomyocytes, thereby controlling the uptake and utilization of myocardial glucose, influencing the development of myocardial insulin resistance, and ultimately modulating MIRI. CONCLUSION Overall, our study sheds new light on the pivotal role of PDHA1 in glucose metabolism and the development of myocardial insulin resistance. Our findings hold promising therapeutic potential for addressing the deleterious effects of MIRI in patients.
Collapse
Affiliation(s)
- Kai-Yuan Chen
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Zhou Liu
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Jing Yi
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou Province, China
| | - Yong-Peng Hui
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Ying-Nan Song
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Jun-Hou Lu
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Hong-Jin Chen
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Si-Yuan Yang
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Xuan-Yi Hu
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Deng-Shen Zhang
- Department of Cardiovascular Surgery, the Affiliated Hospital of Zunyi Medical University, Zunyi, 563009, Guizhou Province, China
| | - Gui-You Liang
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China.
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China.
| |
Collapse
|
10
|
Franczyk B, Gluba-Brzózka A, Rysz-Górzyńska M, Rysz J. The Role of Inflammation and Oxidative Stress in Rheumatic Heart Disease. Int J Mol Sci 2022; 23:ijms232415812. [PMID: 36555452 PMCID: PMC9781220 DOI: 10.3390/ijms232415812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Rheumatic heart disease (RHD), an acquired valvular disease, remains an important cause of morbidity and mortality in developing countries. This chronic illness starts from untreated streptococcal throat infection, resulting in acute rheumatic fever (ARF) in susceptible individuals. Repeated infections lead to a chronic phase characterized by the damage of heart valves. Inflammation has been found to play important role in the development of this disease. All the studies presented in this review clearly show the involvement of the inflammatory state in the progression of this disease. However, the exact role of cytokines in inflammation sites remains to be examined, since most studies have so far focused on peripheral blood. Such analysis would provide information on inflammatory mechanisms in situ.
Collapse
Affiliation(s)
- Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
- Correspondence:
| | - Magdalena Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| |
Collapse
|
11
|
Habimana O, Modupe Salami O, Peng J, Yi GH. Therapeutic Implications of Targeting Pyroptosis in Cardiac-related Etiology of Heart Failure. Biochem Pharmacol 2022; 204:115235. [PMID: 36044938 DOI: 10.1016/j.bcp.2022.115235] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022]
Abstract
Heart failure remains a considerable clinical and public health problem, it is the dominant cause of death from cardiovascular diseases, besides, cardiovascular diseases are one of the leading causes of death worldwide. The survival of patients with heart failure continues to be low with 45-60% reported deaths within five years. Apoptosis, necrosis, autophagy, and pyroptosis mediate cardiac cell death. Acute cell death is the hallmark pathogenesis of heart failure and other cardiac pathologies. Inhibition of pyroptosis, autophagy, apoptosis, or necrosis reduces cardiac damage and improves cardiac function in cardiovascular diseases. Pyroptosis is a form of inflammatory deliberate cell death that is characterized by the activation of inflammasomes such as NOD-like receptors (NLR), absent in melanoma 2 (AIM2), interferon-inducible protein 16 (IFI-16), and their downstream effector cytokines: Interleukin IL-1β and IL-18 leading to cell death. Recent studies have shown that pyroptosis is also the dominant cell death process in cardiomyocytes, cardiac fibroblasts, endothelial cells, and immune cells. It plays a crucial role in the pathogenesis of cardiac diseases that contribute to heart failure. This review intends to summarize the therapeutic implications targeting pyroptosis in the main cardiac pathologies preceding heart failure.
Collapse
Affiliation(s)
- Olive Habimana
- International College, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | | | - Jinfu Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Guang-Hui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China.
| |
Collapse
|
12
|
Tanase DM, Valasciuc E, Gosav EM, Floria M, Costea CF, Dima N, Tudorancea I, Maranduca MA, Serban IL. Contribution of Oxidative Stress (OS) in Calcific Aortic Valve Disease (CAVD): From Pathophysiology to Therapeutic Targets. Cells 2022; 11:cells11172663. [PMID: 36078071 PMCID: PMC9454630 DOI: 10.3390/cells11172663] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is a major cause of cardiovascular mortality and morbidity, with increased prevalence and incidence. The underlying mechanisms behind CAVD are complex, and are mainly illustrated by inflammation, mechanical stress (which induces prolonged aortic valve endothelial dysfunction), increased oxidative stress (OS) (which trigger fibrosis), and calcification of valve leaflets. To date, besides aortic valve replacement, there are no specific pharmacological treatments for CAVD. In this review, we describe the mechanisms behind aortic valvular disease, the involvement of OS as a fundamental element in disease progression with predilection in AS, and its two most frequent etiologies (calcific aortic valve disease and bicuspid aortic valve); moreover, we highlight the potential of OS as a future therapeutic target.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Correspondence:
| | - Claudia Florida Costea
- Department of Ophthalmology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- 2nd Ophthalmology Clinic, Prof. Dr. Nicolae Oblu Emergency Clinical Hospital, 700309 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Ionut Tudorancea
- Department of Morpho-Functional Sciences II, Discipline of Physiology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Cardiology Clinic St. Spiridon County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Minela Aida Maranduca
- Internal Medicine Clinic, St. Spiridon County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Department of Morpho-Functional Sciences II, Discipline of Physiology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ionela Lacramioara Serban
- Department of Morpho-Functional Sciences II, Discipline of Physiology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
13
|
Zhao M, Wei H, Li C, Zhan R, Liu C, Gao J, Yi Y, Cui X, Shan W, Ji L, Pan B, Cheng S, Song M, Sun H, Jiang H, Cai J, Garcia-Barrio MT, Chen YE, Meng X, Dong E, Wang DW, Zheng L. Gut microbiota production of trimethyl-5-aminovaleric acid reduces fatty acid oxidation and accelerates cardiac hypertrophy. Nat Commun 2022; 13:1757. [PMID: 35365608 PMCID: PMC8976029 DOI: 10.1038/s41467-022-29060-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/14/2022] [Indexed: 12/31/2022] Open
Abstract
Numerous studies found intestinal microbiota alterations which are thought to affect the development of various diseases through the production of gut-derived metabolites. However, the specific metabolites and their pathophysiological contribution to cardiac hypertrophy or heart failure progression still remain unclear. N,N,N-trimethyl-5-aminovaleric acid (TMAVA), derived from trimethyllysine through the gut microbiota, was elevated with gradually increased risk of cardiac mortality and transplantation in a prospective heart failure cohort (n = 1647). TMAVA treatment aggravated cardiac hypertrophy and dysfunction in high-fat diet-fed mice. Decreased fatty acid oxidation (FAO) is a hallmark of metabolic reprogramming in the diseased heart and contributes to impaired myocardial energetics and contractile dysfunction. Proteomics uncovered that TMAVA disturbed cardiac energy metabolism, leading to inhibition of FAO and myocardial lipid accumulation. TMAVA treatment altered mitochondrial ultrastructure, respiration and FAO and inhibited carnitine metabolism. Mice with γ-butyrobetaine hydroxylase (BBOX) deficiency displayed a similar cardiac hypertrophy phenotype, indicating that TMAVA functions through BBOX. Finally, exogenous carnitine supplementation reversed TMAVA induced cardiac hypertrophy. These data suggest that the gut microbiota-derived TMAVA is a key determinant for the development of cardiac hypertrophy through inhibition of carnitine synthesis and subsequent FAO. Intestinal microbiota alterations may affect heart function through the production of gut-derived metabolites. Here the authors found that gut microbiota-derived TMAVA is a key determinant for the development of cardiac hypertrophy through inhibition of carnitine synthesis and subsequent fatty acid oxidation.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China.,The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Haoran Wei
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chenze Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Rui Zhan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Changjie Liu
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Jianing Gao
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Yaodong Yi
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiao Cui
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenxin Shan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Liang Ji
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Bing Pan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Si Cheng
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, 100050, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Haipeng Sun
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huidi Jiang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jun Cai
- Fuwai Hospital, State Key Laboratory of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minerva T Garcia-Barrio
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Xiangbao Meng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China.,The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China. .,Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
14
|
Abdel-Emam RA, Ali MF. Effect of l-carnitine supplementation on lead acetate-induced liver cell apoptosis and inflammation: role of caspase-3 and glycogen synthase kinase-3β enzymes. Life Sci 2021; 291:120277. [PMID: 34979196 DOI: 10.1016/j.lfs.2021.120277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/11/2021] [Accepted: 12/24/2021] [Indexed: 01/09/2023]
Abstract
AIM The study aimed at studying the hepatoprotective effect of l-carnitine against lead (Pb) acetate-induced hepatocellular injury, emphasizing the role of caspase-3 and glycogen synthase kinase-3β in hepatocellular apoptosis and inflammation. MATERIALS AND METHODS Male Wistar rats were used. The experimental approach involved estimation of the liver enzymes' serum levels. Oxidative and inflammatory biomarkers were measured in hepatic tissue homogenates. Paraffin-embedded hepatic sections were prepared for histopathology and immunohistochemistry. Quantitative determination of the phosphorylated glycogen synthase kinase-3 beta was performed. KEY FINDINGS The serum showed a significant elevation in ALT, AST, and LDH; tissue homogenates showed significant elevation in lipid peroxide and inflammatory biomarkers with significant reduction in reduced glutathione in the Pb acetate-treated group. Co-administration of l-carnitine with Pb acetate produced significant reduction in liver enzymes with significant improvement in oxidant, antioxidant and inflammatory markers. Lead acetate treatment significantly reduced the phosphorylated glycogen synthase kinase-3 beta, while l-carnitine enhanced its phosphorylation. Histopathological examination showed inflammatory reaction around blood vessels with fatty degeneration in hepatocytes of the Pb acetate intoxicated group. l-Carnitine caused a decrease in hepatic damage with minimal vascular alterations in central vein. Caspase-3 expression in hepatocytes was decreased in Pb-treated group supplemented with l-carnitine. SIGNIFICANCE Our study reveals that oxidative stress and inflammation participate in Pb acetate-induced hepatocellular injury. Glycogen synthase kinase-3β and caspase-3 play role in Pb acetate-induced hepatic damage. l-Carnitine shows significant protective effects against hepatocellular apoptosis and inflammation induced by Pb acetate through antioxidant, anti-inflammatory and anti-apoptotic pathways in part mediated by GSK-3β inhibition.
Collapse
Affiliation(s)
- Rania A Abdel-Emam
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut 71526, Egypt.
| | - Marwa F Ali
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt.
| |
Collapse
|
15
|
Xian S, Chen A, Wu Y, Wen H, Lu C, Huang F, Zeng Z. Interference with the expression of S1PR1 or STAT3 attenuates valvular damage due to rheumatic heart disease. Int J Mol Med 2021; 48:179. [PMID: 34296288 PMCID: PMC8354313 DOI: 10.3892/ijmm.2021.5012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 06/25/2021] [Indexed: 11/20/2022] Open
Abstract
Rheumatic heart disease (RHD) affects numerous individuals annually; however, its pathogenesis remains unclear. The sphingosine 1‑phosphate receptor 1 (S1PR1) and signal transducer and activator of transcription 3 (STAT3) have recently been shown to be involved in valvular damage via the promotion of the differentiation of T helper 17 (Th17) cells during the development of RHD‑induced valvular damage. The present study investigated whether altering the expression of S1PR1 or STAT3 attenuates valvular damage due to RHD. Inactivated group A streptococcus (GAS) was used to establish a rat model of RHD. Recombinant adeno‑associated viral vectors carrying an S1PR1 overexpression sequence were used to overexpress S1PR1. STAT3 small interfering RNA (STAT3‑siRNA) was used to inhibit STAT3 expression. Reverse transcription‑quantitative PCR (RT‑qPCR) was performed to detect the mRNA expression of S1PR1, STAT3, collagen type III α1 chain (Col3a1) and fibroblast‑specific protein 1. Western blotting (WB) and immunohistochemistry were used to detect the levels of S1PR1, STAT3, phosphorylated (p‑) STAT3, and retinoic acid‑related orphan receptor γT (RORγt) proteins. Enzyme‑linked immunosorbent assays (ELISAs) and immunohistochemistry were used to detect the levels of interleukin (IL)‑6 and IL‑17. Hematoxylin and eosin (H&E) staining and Sirius Red staining were performed to evaluate the degree of inflammation and fibrosis in the valvular tissues. S1PR1 expression was decreased in the valvular tissues of the rats with RHD. The levels of IL‑6, IL‑17 and p‑STAT3 in the rats with RHD were increased. The degree of valvular inflammation and fibrosis in the rats with RHD was also increased. The overexpression of S1PR1 and the inhibition of STAT3 reduced the total p‑STAT3 level, resulting in decreased levels of IL‑6, IL‑17 and RORγt, and a reduced degree of valvular inflammation and fibrosis. These results suggest that the expression of S1PR1 and STAT3 may be involved in valvular tissue damage due to RHD. Thus, strategies designed to interfere with the expression of S1PR1 or STAT3 may affect the expression of Th17 cell‑related cytokines and may thus attenuate valvular damage due to RHD.
Collapse
Affiliation(s)
- Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Centre for Cardio-Cerebrovascular Diseases, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ang Chen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Centre for Cardio-Cerebrovascular Diseases, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yunjiao Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Centre for Cardio-Cerebrovascular Diseases, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Hong Wen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Centre for Cardio-Cerebrovascular Diseases, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chuanghong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Centre for Cardio-Cerebrovascular Diseases, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Centre for Cardio-Cerebrovascular Diseases, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Centre for Cardio-Cerebrovascular Diseases, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
16
|
Diabetes Mellitus and Cardiovascular Diseases: Nutraceutical Interventions Related to Caloric Restriction. Int J Mol Sci 2021; 22:ijms22157772. [PMID: 34360538 PMCID: PMC8345941 DOI: 10.3390/ijms22157772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/15/2021] [Accepted: 07/18/2021] [Indexed: 12/17/2022] Open
Abstract
Type 2 diabetes (T2DM) and cardiovascular disease (CVD) are closely associated and represent a key public health problem worldwide. An excess of adipose tissue, NAFLD, and gut dysbiosis establish a vicious circle that leads to chronic inflammation and oxidative stress. Caloric restriction (CR) is the most promising nutritional approach capable of improving cardiometabolic health. However, adherence to CR represents a barrier to patients and is the primary cause of therapeutic failure. To overcome this problem, many different nutraceutical strategies have been designed. Based on several data that have shown that CR action is mediated by AMPK/SIRT1 activation, several nutraceutical compounds capable of activating AMPK/SIRT1 signaling have been identified. In this review, we summarize recent data on the possible role of berberine, resveratrol, quercetin, and L-carnitine as CR-related nutrients. Additionally, we discuss the limitations related to the use of these nutrients in the management of T2DM and CVD.
Collapse
|
17
|
Chen W, Wang Y, Pan Z, Chen X, Luo D, Wang H. Protective effects of dexmedetomidine on the ischemic myocardium in patients undergoing rheumatic heart valve replacement surgery. Exp Ther Med 2021; 21:427. [PMID: 33747166 PMCID: PMC7967827 DOI: 10.3892/etm.2021.9844] [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: 01/12/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to compare the effects of two methods of dexmedetomidine (Dex) administration on myocardial injury, inflammation and stress in ischemic myocardium during rheumatic heart valve replacement. In total, 90 patients were included in the present study and were divided into the following three groups: i) Dex group (1.0 µg/kg Dex pre-administered 10 min prior to anesthesia, then 0.5 µg/kg/h Dex for maintenance); ii) Dex pre-conditioning group (Pre-Dex; 1.0 µg/kg Dex administered 10 min prior to anesthesia, then saline for maintenance); and iii) control group (saline 10 min prior to anesthesia and saline during maintenance), with 30 patients in each group. Heart rate (HR) and mean artery pressure (MAP) were recorded at eight time-points: i) T1, pre-medication; ii) T2, 10 min post-medication; iii) T3, immediately post-intubation; iv) T4, upon skin incision; v) T5, upon sawing the sternum; vi) T6, immediately post-cardiopulmonary bypass; vii) T7, immediately post-operation; and viii) T8, 24 h post-operation. The serum cardiac troponin I (cTnI), interleukin (IL)-8, IL-10 and malondialdehyde (MDA) levels were also detected at T1, T6, T7 and T8. Blood glucose levels were detected at T1, T5, T6 and T7. In comparison with the control group, patients in the Dex group exhibited a significant increase in cardiac function, as indicated by an increase in HR, MAP and IL-10 levels, and a significant decrease in cTnI, IL-8, MDA and glucose levels. Both Dex perfusion and Dex preconditioning were able to reduce myocardial injury, inflammation, oxidative stress and stress response in rheumatic heart valve replacement surgery. However, Dex perfusion during the whole surgery was more effective than Dex preconditioning treatment. The study was registered with the Chinese Clinical Trial Registry (ChiCTR; no. ChiCTR-INR-17011955).
Collapse
Affiliation(s)
- Wei Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yan Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Zhiguo Pan
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Xiyuan Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Dihuan Luo
- Department of Anesthesiology, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Haiying Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
18
|
Zheng HL, Zhang HY, Zhu CL, Li HY, Cui S, Jin J, Piao SG, Jiang YJ, Xuan MY, Jin JZ, Jin YS, Lee JP, Chung BH, Choi BS, Yang CW, Li C. L-Carnitine protects against tacrolimus-induced renal injury by attenuating programmed cell death via PI3K/AKT/PTEN signaling. Acta Pharmacol Sin 2021; 42:77-87. [PMID: 32555441 PMCID: PMC7921136 DOI: 10.1038/s41401-020-0449-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/21/2020] [Indexed: 12/20/2022] Open
Abstract
Reducing immunosuppressant-related complications using conventional drugs is an efficient therapeutic strategy. L-carnitine (LC) has been shown to protect against various types of renal injury. In this study, we investigated the renoprotective effects of LC in a rat model of chronic tacrolimus (TAC) nephropathy. SD rats were injected with TAC (1.5 mg · kg-1 · d-1, sc) for 4 weeks. Renoprotective effects of LC were assessed in terms of renal function, histopathology, oxidative stress, expression of inflammatory and fibrotic cytokines, programmed cell death (pyroptosis, apoptosis, and autophagy), mitochondrial function, and PI3K/AKT/PTEN signaling. Chronic TAC nephropathy was characterized by severe renal dysfunction and typical histological features of chronic nephropathy. At a molecular level, TAC markedly increased the expression of inflammatory and fibrotic cytokines in the kidney, induced oxidative stress, and led to mitochondrial dysfunction and programmed cell death through activation of PI3K/AKT and inhibition of PTEN. Coadministration of LC (200 mg · kg-1 · d-1, ip) caused a prominent improvement in renal function and ameliorated histological changes of kidneys in TAC-treated rats. Furthermore, LC exerted anti-inflammatory and antioxidant effects, prevented mitochondrial dysfunction, and modulated the expression of a series of apoptosis- and autophagy-controlling genes to promote cell survival. Human kidney proximal tubular epithelial cells (HK-2 cells) were treated with TAC (50 μg/mL) in vitro, which induced production of intracellular reactive oxygen species and expression of an array of genes controlling programmed cell death (pyroptosis, apoptosis, and autophagy) through interfering with PI3K/AKT/PTEN signaling. The harmful responses of HK-2 cells to TAC were significantly attenuated by cotreatment with LC and the PI3K inhibitor LY294002 (25 μM). In conclusion, LC treatment protects against chronic TAC nephropathy through interfering the PI3K/AKT/PTEN signaling.
Collapse
Affiliation(s)
- Hai-Lan Zheng
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
| | - Hai-Yue Zhang
- College of Chemical and Life Science, Changchun University of Technology, Changchun, 130000, China
| | - Chun-Lian Zhu
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
| | - Hui-Ying Li
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
| | - Sheng Cui
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
- Transplantation Research Center, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jian Jin
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
| | - Shang-Guo Piao
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
| | - Yu-Ji Jiang
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
| | - Mei-Ying Xuan
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
- Department of Health Examination Central, Yanbian University, Yanji, 133000, China
| | - Ji-Zhe Jin
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
| | - Ying-Shun Jin
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China
| | - Jung-Pyo Lee
- Division of Nephrology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 07061, Korea
| | - Byung-Ha Chung
- Transplantation Research Center, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Bum-Soon Choi
- Transplantation Research Center, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Chul-Woo Yang
- Transplantation Research Center, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Can Li
- Department of Nephrology, Yanbian University Hospital, Yanji, 133000, China.
| |
Collapse
|
19
|
Role of Nrf2 and Its Activators in Cardiocerebral Vascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4683943. [PMID: 32831999 PMCID: PMC7428967 DOI: 10.1155/2020/4683943] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/16/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
Cardiocerebral vascular disease (CCVD) is a common disease with high morbidity, disability, and mortality. Oxidative stress (OS) is closely related to the progression of CCVD. Abnormal redox regulation leads to OS and overproduction of reactive oxygen species (ROS), which can cause biomolecular and cellular damage. The Nrf2/antioxidant response element (ARE) signaling pathway is one of the most important defense systems against exogenous and endogenous OS injury, and Nrf2 is regarded as a vital pharmacological target. The complexity of the CCVD pathological process and the current difficulties in conducting clinical trials have hindered the development of therapeutic drugs. Furthermore, little is known about the role of the Nrf2/ARE signaling pathway in CCVD. Clarifying the role of the Nrf2/ARE signaling pathway in CCVD can provide new ideas for drug design. This review details the recent advancements in the regulation of the Nrf2/ARE system and its role and activators in common CCVD development.
Collapse
|
20
|
Zhao T, Chen S, Wang B, Cai D. L-Carnitine Reduces Myocardial Oxidative Stress and Alleviates Myocardial Ischemia-Reperfusion Injury by Activating Nuclear Transcription-Related Factor 2 (Nrf2)/Heme Oxygenase-1 (HO-1) Signaling Pathway. Med Sci Monit 2020; 26:e923251. [PMID: 32452468 PMCID: PMC7271686 DOI: 10.12659/msm.923251] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Myocardial ischemia-reperfusion injury (IRI) is an important injury mechanism of myocardial infarction. The purpose of this study was to explore the effects of L-carnitine (LC) on myocardial IRI and its mechanism. Material/Methods The IRI model was made by ligating the left anterior descending coronary artery. Then, we injected LC intraperitoneally into the rats of the experimental group to assess the effect of LC on IRI rats by use of serum markers, Western blot, and qRT-PCR. H9c2 cells were cultured and then treated with hypoxia-reoxygenation. The effect of LC on oxidative stress, apoptosis, and nuclear transcription-related factor 2/heme oxygenase-1 (Nrf2/HO-1) signaling pathway of H9c2 cells were detected by Western blot, RT-PCR, and flow cytometry. Results LC significantly reduced malondialdehyde (MDA), creatine kinase (CK), and lactate dehydrogenase (LDH) levels in rat myocardial tissue and increased superoxide dismutase (SOD) expression. LC also increased the expression of SOD1/2 and Bcl-2 in rat myocardial tissue and H9c2 cells and decreased the expression of caspase3/8 and Bax. In addition, LC increased the expression of Nrf2/HO-1 signaling pathway-related molecules in H9c2 cells and increased the activity of the Nrf2/HO-1 signaling pathway. Moreover, inhibition of the Nrf2/HO-1 signaling pathway attenuated the protective effect of LC on H9c2 cells. Conclusions LC can activate the Nrf2/HO-1 signaling pathway and reduce oxidative stress and apoptosis in cardiomyocytes, thereby reducing myocardial IRI.
Collapse
Affiliation(s)
- Tana Zhao
- Second Department of Pediatric, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| | - Shangjun Chen
- Department of Geratology, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| | - Bingxin Wang
- Department of Orthopedics, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| | - Dongliang Cai
- Department of Geratology, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| |
Collapse
|
21
|
Ren L, Wang Q, Ma L, Wang D. MicroRNA-760-mediated low expression of DUSP1 impedes the protective effect of NaHS on myocardial ischemia-reperfusion injury. Biochem Cell Biol 2020; 98:378-385. [PMID: 32160475 DOI: 10.1139/bcb-2019-0310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is the leading cause of the poor prognosis for patients undergoing clinical cardiac surgery. Micro-RNAs are involved in MIRI; however, the effect of miR-760 on MIRI and the molecular mechanisms behind it have not yet been described. For our in-vivo experiments, 20 rats were randomly distributed between 2 groups (n = 10): the sham-treatment group and the ischemia-reperfusion (I/R) group. For our in-vitro experiments, H9C2 cells were subjected to hypoxia for 6 h, and then reoxygenated to establish an hypoxia-reoxygenation (H/R) model. High expression levels of of miR-760 were observed in the rats subjected to MIRI and the H9C2 cells subjected to H/R. Further, the levels of lactate dehydrogenase (LDH) and malonaldehyde (MDA) were increased, and the size of the myocardial infarct was notably greater in the rats subjected to MIRI, suggesting that miR-760 worsens the effects of MIRI. The inhibitory effects from NaHS on apoptosis were enhanced, as were the expression levels of cleaved caspase 3 and cleaved PARP in H9C2 cells exposed to H/R, and with low-expression levels of miR-760. TargetScan and dual luciferase reporter assays further confirmed the targeted relationship between dual-specificity protein phosphatase (DUSP1) and miR-760. Additionally, miR-760 overexpression and H/R treatment of H9C2 cells inhibited the expression of DUSP1, which further promoted apoptosis. Furthermore, DUSP1 enhanced the anti-apoptotic effects of NaHS in rats subjected to MIRI. Taken together, these findings suggest that miR-760 inhibits the protective effect of NaHS against MIRI.
Collapse
Affiliation(s)
- Lin Ren
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang City, Hebei Province 050011, China.,Department of Cardiology, First Hospital of Qinhuangdao, Qinhuangdao City, Hebei Province 066000, China
| | - Qian Wang
- Department of Geriatrics, First Hospital of Qinhuangdao, Qinhuangdao City, Hebei Province 066000, China
| | - Lixiang Ma
- Department of Cardiology, First Hospital of Qinhuangdao, Qinhuangdao City, Hebei Province 066000, China
| | - Dongmei Wang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang City, Hebei Province 050011, China
| |
Collapse
|