51
|
Kimura M, Yokoyama A, Higuchi S. Aldehyde dehydrogenase-2 as a therapeutic target. Expert Opin Ther Targets 2019; 23:955-966. [DOI: 10.1080/14728222.2019.1690454] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Mitsuru Kimura
- National Hospital Organization Kurihama Medical and Addiction Center, Yokosuka, Kanagawa, Japan
| | - Akira Yokoyama
- National Hospital Organization Kurihama Medical and Addiction Center, Yokosuka, Kanagawa, Japan
| | - Susumu Higuchi
- National Hospital Organization Kurihama Medical and Addiction Center, Yokosuka, Kanagawa, Japan
| |
Collapse
|
52
|
Wu NN, Tian H, Chen P, Wang D, Ren J, Zhang Y. Physical Exercise and Selective Autophagy: Benefit and Risk on Cardiovascular Health. Cells 2019; 8:cells8111436. [PMID: 31739509 PMCID: PMC6912418 DOI: 10.3390/cells8111436] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022] Open
Abstract
Physical exercise promotes cardiorespiratory fitness, and is considered the mainstream of non-pharmacological therapies along with lifestyle modification for various chronic diseases, in particular cardiovascular diseases. Physical exercise may positively affect various cardiovascular risk factors including body weight, blood pressure, insulin sensitivity, lipid and glucose metabolism, heart function, endothelial function, and body fat composition. With the ever-rising prevalence of obesity and other types of metabolic diseases, as well as sedentary lifestyle, regular exercise of moderate intensity has been indicated to benefit cardiovascular health and reduce overall disease mortality. Exercise offers a wide cadre of favorable responses in the cardiovascular system such as improved dynamics of the cardiovascular system, reduced prevalence of coronary heart diseases and cardiomyopathies, enhanced cardiac reserve capacity, and autonomic regulation. Ample clinical and experimental evidence has indicated an emerging role for autophagy, a conservative catabolism process to degrade and recycle cellular organelles and nutrients, in exercise training-offered cardiovascular benefits. Regular physical exercise as a unique form of physiological stress is capable of triggering adaptation while autophagy in particular selective autophagy seems to be permissive to such cardiovascular adaptation. Here in this mini-review, we will summarize the role for autophagy in particular mitochondrial selective autophagy namely mitophagy in the benefit versus risk of physical exercise on cardiovascular function.
Collapse
Affiliation(s)
- Ne N. Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Haili Tian
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.T.); (P.C.)
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.T.); (P.C.)
| | - Dan Wang
- School of Physical Education and Sport Training, Shanghai University of Sport, Shanghai 200438, China;
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Correspondence: (J.R.); (Y.Z.)
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Correspondence: (J.R.); (Y.Z.)
| |
Collapse
|
53
|
Han L, Tang Y, Li S, Wu Y, Chen X, Wu Q, Hong K, Li J. Protective mechanism of SIRT1 on Hcy-induced atrial fibrosis mediated by TRPC3. J Cell Mol Med 2019; 24:488-510. [PMID: 31680473 PMCID: PMC6933351 DOI: 10.1111/jcmm.14757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 09/29/2019] [Accepted: 10/04/2019] [Indexed: 12/21/2022] Open
Abstract
High plasma levels of homocysteine (Hcy) are regarded as a risk factor for atrial fibrillation (AF), which is closely associated with the pathological consequence of atrial fibrosis and can lead to heart failure with a high mortality rate; here, we show that atrial fibrosis is mediated by the relationship between canonical transient receptor potential 3 (TRPC3) channels and sirtuin type 1 (SIRT1) under the stimulation of Hcy. The left atrial appendage was obtained from patients with either sinus rhythm (SR) or AF and used to evaluate the relationship between the concentration of Hcy and a potential mechanism of cardiac fibrosis mediated by TRPC3 and SIRT1. We next performed transverse aortic constriction (TAC) in mouse to investigate the relationship. The mechanisms underlying atrial fibrosis involving TRPC3 and SIRT1 proteins were explored by co‐IP, BLI and lentivirus transfection experiments. qPCR and WB were performed to analyse gene and protein expression, respectively. The higher level of atrial fibrosis was observed in the HH mouse group with a high Hcy diet. Such results suggest that AF patients may be more susceptible to atrial fibrosis and possess a high probability of progressing to hyperhomocysteinemia. Moreover, our findings are consistent with the hypothesis that TRPC3 channel up‐regulation leads to abnormal accumulation of collagen, with the down‐regulation of SIRT1 as an aetiological factor of high Hcy, which in turn predisposes to atrial fibrosis and strongly enhances the possibility of AF.
Collapse
Affiliation(s)
- Lu Han
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanhua Tang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shaochuan Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanqing Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoshu Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kui Hong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
54
|
Liu Q, Liu Y, Li F, Gu Z, Liu M, Shao T, Zhang L, Zhou G, Pan C, He L, Cai J, Zhang X, Barve S, McClain CJ, Chen Y, Feng W. Probiotic culture supernatant improves metabolic function through FGF21-adiponectin pathway in mice. J Nutr Biochem 2019; 75:108256. [PMID: 31760308 DOI: 10.1016/j.jnutbio.2019.108256] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/01/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
High-fat/high-fructose diet plus intermittent hypoxia exposure (HFDIH) causes metabolic disorders such as insulin resistance, obesity, nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes. The purpose of this study is to examine the effects and understand the mechanism of action of Lactobacillus rhamnosus GG culture supernatant (LGGs) on HFDIH-induced metabolic dysfunction. Mice were fed high-fat:high-fructose diet for 15 weeks. After 3 weeks of feeding, the mice were exposed to chronic intermittent hypoxia for the next 12 weeks (HFDIH), and LGGs was supplemented over the entire experiment. HFDIH exposure significantly led to metabolic disorders. LGGs treatment showed significant improvements in indices of metabolic disorders including fat mass, energy expenditure, glucose intolerance, insulin resistance, increased hepatic steatosis and liver injury. HFDIH mice markedly increased adipose inflammation and adipocyte size, and reduced circulating adiponectin, which was restored by LGGs treatment. LGGs treatment increased hepatic FGF21 mRNA expression and circulating FGF21 protein levels, which were associated with increased hepatic PPARα expression and fecal butyrate concentration. In addition, HFDIH-induced hepatic fat accumulation and apoptosis were significantly reduced by LGGs supplementation. In summary, LGGs treatment increased energy expenditure and insulin sensitivity and prevented metabolic abnormalities in HFDIH mice, and this is associated with the FGF21-adiponectin signaling pathway. LGGs may be a potential prevention/treatment strategy in subjects with the metabolic syndrome.
Collapse
Affiliation(s)
- Qi Liu
- Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Yunhuan Liu
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Fengyuan Li
- Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Zelin Gu
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Min Liu
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Tuo Shao
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Lihua Zhang
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Guangyao Zhou
- Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Chengwei Pan
- Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liqing He
- Department of Chemistry, University of Louisville, Louisville, KY, USA
| | - Jun Cai
- Department of Pediatrics, University of Louisville, Louisville, KY, USA
| | - Xiang Zhang
- Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA; Department of Chemistry, University of Louisville, Louisville, KY, USA
| | - Shirish Barve
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Craig J McClain
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA; Robley Rex VA medical Center, Louisville, KY, USA
| | - Yiping Chen
- Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Wenke Feng
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
55
|
Castañeda D, Gabani M, Choi SK, Nguyen QM, Chen C, Mapara A, Kassan A, Gonzalez AA, Ait-Aissa K, Kassan M. Targeting Autophagy in Obesity-Associated Heart Disease. Obesity (Silver Spring) 2019; 27:1050-1058. [PMID: 30938942 DOI: 10.1002/oby.22455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/30/2019] [Indexed: 01/18/2023]
Abstract
Over the past three decades, the increasing rates of obesity have led to an alarming obesity epidemic worldwide. Obesity is associated with an increased risk of cardiovascular diseases; thus, it is essential to define the molecular mechanisms by which obesity affects heart function. Individuals with obesity and overweight have shown changes in cardiac structure and function, leading to cardiomyopathy, hypertrophy, atrial fibrillation, and arrhythmia. Autophagy is a highly conserved recycling mechanism that delivers proteins and damaged organelles to lysosomes for degradation. In the hearts of patients and mouse models with obesity, this process is impaired. Furthermore, it has been shown that autophagy flux restoration in obesity models improves cardiac function. Therefore, autophagy may play an important role in mitigating the adverse effects of obesity on the heart. Throughout this review, we will discuss the benefits of autophagy on the heart in obesity and how regulating autophagy might be a therapeutic tool to reduce the risk of obesity-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Diana Castañeda
- Department of Biological Sciences, California State University, Los Angeles, California, USA
| | - Mohanad Gabani
- Cardiovascular Division, Department of Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Soo-Kyoung Choi
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Quynh My Nguyen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California, USA
| | - Cheng Chen
- Department of Emergency and Critical Care, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China, Shanghai
| | - Ayesha Mapara
- Department of Biology, Northeastern Illinois University, Chicago, Illinois, USA
| | - Adam Kassan
- School of Pharmacy, West Coast University, Los Angeles, California, USA
| | - Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Karima Ait-Aissa
- Cardiovascular Division, Department of Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Modar Kassan
- Cardiovascular Division, Department of Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
56
|
Sun M, Tan Y, Rexiati M, Dong M, Guo W. Obesity is a common soil for premature cardiac aging and heart diseases - Role of autophagy. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1898-1904. [DOI: 10.1016/j.bbadis.2018.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/22/2018] [Accepted: 09/04/2018] [Indexed: 12/31/2022]
|
57
|
Wang Y, Yang Z, Zheng G, Yu L, Yin Y, Mu N, Ma H. Metformin promotes autophagy in ischemia/reperfusion myocardium via cytoplasmic AMPKα1 and nuclear AMPKα2 pathways. Life Sci 2019; 225:64-71. [PMID: 30953640 DOI: 10.1016/j.lfs.2019.04.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/25/2019] [Accepted: 04/01/2019] [Indexed: 12/25/2022]
Abstract
AIMS In myocardial ischemia-reperfusion (MI/R) injury, impaired autophagy function worsens cardiomyocyte death. AMP-activated protein kinase (AMPK) is a heterotrimeric protein that plays an important role in cardioprotection and myocardial autophagic function. AMPKα1 and α2 are localized primarily in the cytoplasm and nucleus, respectively, in cardiomyocytes, but the isoform-specific autophagy regulation of AMPK during MI/R remains unclear. MATERIALS AND METHODS An MI/R model was built, and the protein expression of AMPKα1/α2, p-AMPK, mTOR, p-mTOR, TFEB, p-FoxO3a, SKP2, CARM1, TBP, Atg5, LAMP2, LC3B, and p62 during ischemia and reperfusion was determined by western blotting. Recombinant adeno-associated virus (serotype 9) vectors carrying tandem fluorescent-tagged LC3 or mRFP-GFP-LC3/GFP-LC3 were used to evaluate the autophagy status. AMPKα2 knockout mice were used for in vivo studies. KEY FINDINGS Both cytoplasmic AMPKα1 and nuclear α2 subunit expression decreased during the reperfusion period, which led to AMPKα1-mTOR-TFEB and AMPKα2-Skp2-CARM1-TFEB signaling inhibition, respectively. The decreased TFEB level during reperfusion suppressed autophagy. Metformin could activate both the AMPKα1- and α2- mediated pathways, thus restoring autophagy flux during reperfusion. Nevertheless, in AMPKα2 knockout mice, nuclear α2-regulated Skp2-CARM1-TFEB signaling was inhibited, while α1-related signaling was comparatively unaffected, which partially impaired metformin-enhanced autophagy. SIGNIFICANCE Our study suggests that metformin had the dual effects of promoting both cytoplasmic AMPKα1- and nuclear AMPKα2-related signaling to improve autophagic flux and restore cardiac function during MI/R.
Collapse
Affiliation(s)
- Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Zheng Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Guoxu Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China.
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
58
|
Pang J, Peng H, Wang S, Xu X, Xu F, Wang Q, Chen Y, Barton LA, Chen Y, Zhang Y, Ren J. Mitochondrial ALDH2 protects against lipopolysaccharide-induced myocardial contractile dysfunction by suppression of ER stress and autophagy. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1627-1641. [PMID: 30946956 DOI: 10.1016/j.bbadis.2019.03.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/13/2019] [Accepted: 03/28/2019] [Indexed: 12/21/2022]
Abstract
Lipopolysaccharide (LPS), an essential component of outer membrane of the Gram-negative bacteria, plays a pivotal role in myocardial anomalies in sepsis. Recent evidence depicted an essential role for mitochondrial aldehyde dehydrogenase (ALDH2) in cardiac homeostasis. This study examined the effect of ALDH2 on endotoxemia-induced cardiac anomalies. Echocardiographic, cardiac contractile and intracellular Ca2+ properties were examined. Our results indicated that LPS impaired cardiac contractile function (reduced fractional shortening, LV end systolic diameter, peak shortening, maximal velocity of shortening/relengthening, prolonged relengthening duration, oxidation of SERCA, and intracellular Ca2+ mishandling), associated with ER stress, inflammation, O2- production, increased autophagy, CAMKKβ, phosphorylated AMPK and suppressed phosphorylation of mTOR, the effects of which were significantly attenuated or negated by ALDH2. LPS promoted early endosomal formation (as evidenced by RAB4 and RAB5a), apoptosis and necrosis (MTT and LDH) while decreasing late endosomal formation (RAB7 and RAB 9), the effects were reversed by ALDH2. In vitro study revealed that LPS-induced SERCA oxidation, autophagy and cardiac dysfunction were abrogated by ALDH2 activator Alda-1, the ER chaperone TUDCA, the autophagy inhibitor 3-MA, or the AMPK inhibitor Compound C. The beneficial effect of Alda-1 against LPS was nullified by AMPK activator AICAR or rapamycin. CAMKKβ inhibition failed to rescue LPS-induced ER stress. Tunicamycin-induced cardiomyocyte dysfunction was ameliorated by Alda-1 and autophagy inhibition, the effect of which was abolished by rapamycin. These data suggested that ALDH2 protected against LPS-induced cardiac anomalies via suppression of ER stress, autophagy in a CAMKKβ/AMPK/mTOR-dependent manner.
Collapse
Affiliation(s)
- Jiaojiao Pang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Hu Peng
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Shuyi Wang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Xihui Xu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Feng Xu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Qiurong Wang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Yuanzhuo Chen
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Linzi A Barton
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Yuguo Chen
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Yingmei Zhang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Fudan University Zhongshan Hospital, Shanghai 200032, China.
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Fudan University Zhongshan Hospital, Shanghai 200032, China.
| |
Collapse
|
59
|
Xia CL, Chu P, Liu YX, Qu XL, Gao XF, Wang ZM, Dong J, Chen SL, Zhang JX. ALDH2 rs671 polymorphism and the risk of heart failure with preserved ejection fraction (HFpEF) in patients with cardiovascular diseases. J Hum Hypertens 2019; 34:16-23. [PMID: 30846829 DOI: 10.1038/s41371-019-0182-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/31/2019] [Accepted: 02/05/2019] [Indexed: 12/16/2022]
Abstract
Aldehyde dehydrogenase 2 (ALDH2) rs671 polymorphism is an established genetic risk of hypertension, diabetes, and coronary heart diseases in Asian population. Previous experimental data showed ALDH2 regulated inflammation, a potential mechanism of heart failure with preserved ejection fraction (HFpEF). However, clinically, the association between ALDH2 polymorphism and incidence of HFpEF remains unknown. In this prospective cross-sectional study, ALDH2 genotyping was performed in 613 consecutive patients enrolled with cardiovascular diseases (CVDs), including hypertension, coronary heart diseases, and/or diabetes mellitus, with normal left ventricular ejection fraction (LVEF). HFpEF was diagnosed according to symptoms and/or signs of dyspnea, fatigue or ankle swelling, N-terminal pro-B-Type natriuretic peptide (NT pro-BNP ≥ 280 pg/mL), LVEF ≥ 50%, and at least one additional criterion: left atrial enlargement (left atrial diameter > 40 mm), diastolic dysfunction (E/E' ≥ 13 or E'/A' < 1) or concurrently with atrial fibrillation. Finally, of 613 patients with CVD, 379 patients (61.8%) were assigned to the wild-type ALDH2*1/*1 group and 234 patients (38.2%) to the mutation-type ALDH2*2 group according to genotyping results. Sixty-nine patients (11.3%) were diagnosed with HFpEF. In ALDH2*2 group, the occurrence of HFpEF was higher (15.4% vs. 8.7%, p = 0.011) than that in ALDH2*1/*1 group. Leukocyte count, the indicator of systemic inflammation, was significantly higher (6.9 ± 2.4 × 109/L vs. 6.5 ± 1.9 × 109/L, p = 0.010) in ALDH2*2 group compared to ALDH2*1/*1 group. In conclusion, ALDH2*2 variant is associated with the risk of HFpEF in patients with CVD. Increased systemic inflammation probably involved in this disease process.
Collapse
Affiliation(s)
- Chun-Lei Xia
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Chu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yi-Xian Liu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xin-Liang Qu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Fei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhi-Mei Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jing Dong
- Department of Echocardiography, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shao-Liang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Jun-Xia Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
60
|
Cholinergic drugs ameliorate endothelial dysfunction by decreasing O-GlcNAcylation via M3 AChR-AMPK-ER stress signaling. Life Sci 2019; 222:1-12. [PMID: 30786250 DOI: 10.1016/j.lfs.2019.02.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/15/2019] [Accepted: 02/17/2019] [Indexed: 01/22/2023]
Abstract
AIMS Obesity is associated with increased cardiovascular morbidity and mortality. It is accompanied by augmented O-linked β-N-acetylglucosamine (O-GlcNAc) modification of proteins via increasing hexosamine biosynthetic pathway (HBP) flux. However, the changes and regulation of the O-GlcNAc levels induced by obesity are unclear. MAIN METHODS High fat diet (HFD) model was induced obesity in mice with or without the cholinergic drug pyridostigmine (PYR, 3 mg/kg/d) for 22 weeks and in vitro human umbilical vein endothelial cells (HUVECs) was treated with high glucose (HG, 30 mM) with or without acetylcholine (ACh). KEY FINDINGS PYR significantly reduced body weight, blood glucose, and O-GlcNAcylation levels and attenuated vascular endothelial cells detachment in HFD-fed mice. HG addition induced endoplasmic reticulum (ER) stress and increased O-GlcNAcylation levels and apoptosis in HUVECs in a time-dependent manner. Additionally, HG decreased levels of phosphorylated AMP-activated protein kinase (AMPK). Interestingly, ACh significantly blocked damage to HUVECs induced by HG. Furthermore, the effects of ACh on HG-induced ER stress, O-GlcNAcylation, and apoptosis were prevented by treating HUVECs with 4-diphenylacetoxy-N-methylpiperidine methiodide (4-DAMP, a selective M3 AChR antagonist) or compound C (Comp C, an AMPK inhibitor). Treatment with 5-aminoimidazole-4-carboxamide ribose (AICAR, an AMPK activator), 4-phenyl butyric acid (4-PBA, an ER stress inhibitor), and 6-diazo-5-oxonorleucine (DON, a GFAT antagonist) reproduced a similar effect with ACh. SIGNIFICANCE Activation of cholinergic signaling ameliorated endothelium damage, reduced levels of ER stress, O-GlcNAcylation, and apoptosis in mice and HUVECs under obese conditions, which may function through M3 AChR-AMPK signaling.
Collapse
|
61
|
Che DN, Kang HJ, Cho BO, Shin JY, Jang SI. Combined effects of Diospyros lotus leaf and grape stalk extract in high-fat-diet-induced obesity in mice. Food Sci Biotechnol 2019; 28:1207-1215. [PMID: 31275721 DOI: 10.1007/s10068-018-00551-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to investigate the combined effects of Diospyros lotus leaves extracts (DLE) and Muscat Bailey A grape stalk extracts (MGSE) in obesity induced by high-fat diet (HFD) in mice. The mice were fed with HFD and orally administered daily with DLE, MGSE, a mixture of DLE and MGSE, and Garcinia cambogia extract over a period of 16 weeks. The results revealed that daily administration of DLE and MGSE mixtures markedly prevented HFD-induced weight gain, plasma lipid profile, hepatic steatosis, hepatic fibrosis, diabetic symptoms, and the risk of developing cardiovascular diseases. Also, DLE and MGSE mixtures administration greatly prevented oxidative stress and liver toxicity. The combined effects of DLE and MGSE mixtures were higher than effects of the single extracts and of G. cambogia, a plant known for its anti-obesity effects. In summary, these findings demonstrated that DLE and MGSE mixtures, exhibit anti-obesity activity in HFD-fed mice.
Collapse
Affiliation(s)
- Denis Nchang Che
- 1Department of Health Management, Jeonju University, 303 Cheonjam-ro, Wansan-gu, Jeonju, Jeollabuk-do 55069 Republic of Korea
- 2Department of Food Science and Technology, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Hyun Ju Kang
- Research Institute, Ato Q&A Co., LTD, Jeonju, Jeollabuk-do 54840 Republic of Korea
| | - Byoung Ok Cho
- 1Department of Health Management, Jeonju University, 303 Cheonjam-ro, Wansan-gu, Jeonju, Jeollabuk-do 55069 Republic of Korea
- Research Institute, Ato Q&A Co., LTD, Jeonju, Jeollabuk-do 54840 Republic of Korea
| | - Jae Young Shin
- 1Department of Health Management, Jeonju University, 303 Cheonjam-ro, Wansan-gu, Jeonju, Jeollabuk-do 55069 Republic of Korea
| | - Seon Il Jang
- 1Department of Health Management, Jeonju University, 303 Cheonjam-ro, Wansan-gu, Jeonju, Jeollabuk-do 55069 Republic of Korea
- Research Institute, Ato Q&A Co., LTD, Jeonju, Jeollabuk-do 54840 Republic of Korea
| |
Collapse
|
62
|
Aldehyde Dehydrogenases Genetic Polymorphism and Obesity: From Genomics to Behavior and Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1193:135-154. [PMID: 31368102 DOI: 10.1007/978-981-13-6260-6_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obesity is multifactorial and complex. Remarkable progress has been made recently in search for polygenic obesity through genome-wide association study (GWAS), but biology of polygenic effects on obesity is largely poor. This review summarizes the available evidence and provides an overview of the links between ALDH2 variants and adiposity, which were firstly and mainly derived from studies of polygenic obesity and also indirectly investigated by using cell lines and mice. The genetic association studies have observed consistent associations of ALDH2 variants with obesity-related traits including BMI, waist circumference (WC), waist-to-hip ratio (WHR), and visceral fat accumulation. In consideration of ALDH2 variants with enzyme activity and alcohol consumption behavior in physiological mechanism studies, we proposed a model by which the physiological and behavioral consequences of alcohol consumption serve as an intermediary process between polymorphisms in ALDH2 and obesity.
Collapse
|
63
|
Aldehyde Dehydrogenase 2 (ALDH2) and Aging: Is There a Sensible Link? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1193:237-253. [DOI: 10.1007/978-981-13-6260-6_15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
64
|
Wan H, Li J, Liao H, Liao M, Luo L, Xu L, Yuan K, Zeng Y. Nicotinamide induces liver regeneration and improves liver function by activating SIRT1. Mol Med Rep 2018; 19:555-562. [PMID: 30483782 DOI: 10.3892/mmr.2018.9688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 09/21/2018] [Indexed: 02/05/2023] Open
Affiliation(s)
- Hai‑Feng Wan
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jia‑Xin Li
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hao‑Tian Liao
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ming‑Heng Liao
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lin Luo
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lin Xu
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ke‑Fei Yuan
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yong Zeng
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
65
|
Ceylan AF, Wang S, Kandadi MR, Chen J, Hua Y, Pei Z, Nair S, Ren J. Cardiomyocyte-specific knockout of endothelin receptor a attenuates obesity cardiomyopathy. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3339-3352. [DOI: 10.1016/j.bbadis.2018.07.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/14/2018] [Accepted: 07/16/2018] [Indexed: 12/20/2022]
|
66
|
Ren J, Sowers JR, Zhang Y. Metabolic Stress, Autophagy, and Cardiovascular Aging: from Pathophysiology to Therapeutics. Trends Endocrinol Metab 2018; 29:699-711. [PMID: 30145108 PMCID: PMC6151141 DOI: 10.1016/j.tem.2018.08.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/30/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022]
Abstract
Recent advances in health care have improved the management of cardiometabolic disorders, and prolonged lifespan. However, the ever-rising prevalence of metabolic stress related to obesity (insulin resistance, diabetes, hypertension, and dyslipidemia) has greatly challenged geriatric care. The ubiquitin-proteasome system and autophagy-lysosomal pathways represent two major, yet distinct cellular machineries, for degradation and removal of damaged or long-lived proteins and organelles; the function of which declines with aging. To seek new strategies for cardiovascular aging under various metabolic diseases, it is imperative to understand the precise role for metabolic stress and protein quality control, in particular autophagy, in premature cardiovascular aging. Targeting metabolic stress and autophagy may offer exciting new avenues for the management of cardiovascular aging.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| | - James R Sowers
- Diabetes and Dalton Cardiovascular Center and Harry S. Truman Memorial VA Research, University of Missouri-Columbia School of Medicine, Columbia, MO 65212, USA
| | - Yingmei Zhang
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
67
|
Epigenetic processing in cardiometabolic disease. Atherosclerosis 2018; 281:150-158. [PMID: 30290963 DOI: 10.1016/j.atherosclerosis.2018.09.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/24/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
Albeit a consistent body of evidence supports the notion that genes influence cardiometabolic features and outcomes, the "non-genetic regulation" of this process is gaining increasing attention. Plastic chemical changes of DNA/histone complexes - known as epigenetic changes - critically determine gene activity by rapidly modifying chromatin accessibility to transcription factors. In this review, we describe the emerging role of chromatin modifications as fine tuners of gene transcription in adipogenesis, insulin resistance, macrophage polarization, immuno-metabolism, endothelial dysfunction and metabolic cardiomyopathy. Epigenetic processing participates in the dynamic interplay among different organs in the cardiometabolic patient. DNA methylation and post-translational histone modifications in both visceral and subcutaneous adipose tissue enable the transcription of genes implicated in lipo- and adipogenesis, inflammation and insulin resistance. Along the same line, complex networks of chromatin modifying enzymes are responsible for impaired nitric oxide bioavailability and defective insulin signalling in the vasculature, thus leading to reduced capillary recruitment and insulin delivery in the liver, skeletal muscle and adipose tissue. Furthermore, changes in methylation status of IL-4, IFNγ and Forkhead box P3 (Foxp3) gene loci are crucial for the polarization of immune cells, thus leading to adipose tissue inflammation and atherosclerosis. Cell-specific epigenetic information could advance our understanding of cardiometabolic processes, thus leading to individualized risk assessment and personalized therapeutic approaches in patients with cardiometabolic disturbances. The development of new chromatin modifying drugs indicates that targeting epigenetic changes is a promising approach to reduce the burden of cardiovascular disease in this setting.
Collapse
|
68
|
Autophagy in Metabolic Age-Related Human Diseases. Cells 2018; 7:cells7100149. [PMID: 30249977 PMCID: PMC6210409 DOI: 10.3390/cells7100149] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a highly conserved homeostatic cellular mechanism that mediates the degradation of damaged organelles, protein aggregates, and invading pathogens through a lysosome-dependent pathway. Over the last few years, specific functions of autophagy have been discovered in many tissues and organs; however, abnormal upregulation or downregulation of autophagy has been depicted as an attribute of a variety of pathologic conditions. In this review, we will describe the current knowledge on the role of autophagy, from its regulation to its physiological influence, in metabolic age-related disorders. Finally, we propose to discuss the therapeutic potential of pharmacological and nutritional modulators of autophagy to treat metabolic diseases.
Collapse
|
69
|
Sun T, Li MY, Li PF, Cao JM. MicroRNAs in Cardiac Autophagy: Small Molecules and Big Role. Cells 2018; 7:cells7080104. [PMID: 30103495 PMCID: PMC6116024 DOI: 10.3390/cells7080104] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy, which is an evolutionarily conserved process according to the lysosomal degradation of cellular components, plays a critical role in maintaining cell homeostasis. Autophagy and mitochondria autophagy (mitophagy) contribute to the preservation of cardiac homeostasis in physiological settings. However, impaired or excessive autophagy is related to a variety of diseases. Recently, a close link between autophagy and cardiac disorders, including myocardial infarction, cardiac hypertrophy, cardiomyopathy, cardiac fibrosis, and heart failure, has been demonstrated. MicroRNAs (miRNAs) are a class of small non-coding RNAs with a length of approximately 21–22 nucleotides (nt), which are distributed widely in viruses, plants, protists, and animals. They function in mediating the post-transcriptional gene silencing. A growing number of studies have demonstrated that miRNAs regulate cardiac autophagy by suppressing the expression of autophagy-related genes in a targeted manner, which are involved in the pathogenesis of heart diseases. This review summarizes the role of microRNAs in cardiac autophagy and related cardiac disorders. Furthermore, we mainly focused on the autophagy regulation pathways, which consisted of miRNAs and their targeted genes.
Collapse
Affiliation(s)
- Teng Sun
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, China.
| | - Meng-Yang Li
- Institute for Translational Medicine, Qingdao University, Qingdao 266021, China.
| | - Pei-Feng Li
- Institute for Translational Medicine, Qingdao University, Qingdao 266021, China.
| | - Ji-Min Cao
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
70
|
Zhang L, Wei C, Ruan Y, Zhang Y, Zhou Y, Lei D. Serum containing Buyang Huanwu decoction prevents age-associated migration and invasion of human vascular smooth muscle cells by up regulating SIRT1 expression. Biosci Trends 2018; 12:282-290. [PMID: 29952352 DOI: 10.5582/bst.2018.01063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The migration and invasion of vascular smooth muscle cells (VSMCs) caused by advanced aging play an important role in diffuse intimal thickening, facilitate adverse arterial remodeling and contribute to the initiation and progression of cardiovascular diseases. The inhibitory function of Buyang Huanwu decoction (BYHWD) has been found on aortic intimal hyperplasia and VSMC proliferation, but its effect on age-associated migration and invasion remains unknown. Here, we used an in vitro angiotensin II (Ang II)-induced senescence model to study the effects of serum containing BYHWD (BYHWS) on the migratory and invasive capacities, matrix metalloprotease type 2 (MMP-2) expression and modulation of sirtuin1 (SIRT1) signaling in human aorta VSMCs (HA-VAMCs). Our results showed that BYHWS was able to inhibit Ang II-induced migration and invasion, with down-regulation of MMP-2. In addition, manipulation of SIRT1 by either over-expression or siRNA knockdown ameliorated or promoted cellular migration and invasion, respectively. Moreover, BYHWS reversed senescence-mediated decrease of SIRT1 levels and SIRT1 was required for BYHWS regulation on migration and invasion of senescent HA-VAMCs. In summary, our data demonstrated that BYHWS suppressed the migration and invasion of age-associated VSMC via an increase of the SIRT1 level, which provides novel insights for the therapy of age-associated cardiovascular diseases.
Collapse
MESH Headings
- Aging/drug effects
- Aging/physiology
- Angiotensin II/pharmacology
- Aorta/cytology
- Aorta/physiology
- Cardiovascular Diseases/drug therapy
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cellular Senescence/drug effects
- Cellular Senescence/physiology
- Down-Regulation
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Gene Knockdown Techniques
- Humans
- Matrix Metalloproteinase 2/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- RNA, Small Interfering/metabolism
- Signal Transduction/drug effects
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| | - Chunshan Wei
- Department of Liver Disease, Shenzhen Hospital Affiliated to Guangzhou University of Chinese Medicine
| | - Yunjun Ruan
- Department of Cardiology, Guangzhou General Hospital of Guangzhou Military Command
| | - Yanan Zhang
- Veterinary medicine, Northeast Agricultural University
| | - Yuliang Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| | - Da Lei
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| |
Collapse
|
71
|
Abstract
Obesity poses a severe threat to human health, including the increased prevalence of hypertension, insulin resistance, diabetes mellitus, cancer, inflammation, sleep apnoea and other chronic diseases. Current therapies focus mainly on suppressing caloric intake, but the efficacy of this approach remains poor. A better understanding of the pathophysiology of obesity will be essential for the management of obesity and its complications. Knowledge gained over the past three decades regarding the aetiological mechanisms underpinning obesity has provided a framework that emphasizes energy imbalance and neurohormonal dysregulation, which are tightly regulated by autophagy. Accordingly, there is an emerging interest in the role of autophagy, a conserved homeostatic process for cellular quality control through the disposal and recycling of cellular components, in the maintenance of cellular homeostasis and organ function by selectively ridding cells of potentially toxic proteins, lipids and organelles. Indeed, defects in autophagy homeostasis are implicated in metabolic disorders, including obesity, insulin resistance, diabetes mellitus and atherosclerosis. In this Review, the alterations in autophagy that occur in response to nutrient stress, and how these changes alter the course of obesogenesis and obesity-related complications, are discussed. The potential of pharmacological modulation of autophagy for the management of obesity is also addressed.
Collapse
Affiliation(s)
- Yingmei Zhang
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri-Columbia School of Medicine, Columbia, MO, USA
| | - Jun Ren
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| |
Collapse
|