1
|
Meulendijks ER, Janssen-Telders C, Hulsman EL, Lobe N, Zappala P, Terpstra MM, Wesselink R, de Vries TAC, Al-Shama RF, van Veen RN, de Castro SMM, de Vries CEE, Nijland LMG, Planken RN, Krul SPJ, de Groot JR. The change of epicardial adipose tissue characteristics and vulnerability for atrial fibrillation upon drastic weight loss. Adipocyte 2024; 13:2395565. [PMID: 39248109 PMCID: PMC11385166 DOI: 10.1080/21623945.2024.2395565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Obesity increases the risk of atrial fibrillation (AF). We hypothesize that 'obese' epicardial adipose tissue (EAT) is, regardless of comorbidities, associated with markers of AF vulnerability. METHODS Patients >40y of age undergoing bariatric surgery and using <2 antihypertensive drugs and no insulin were prospectively included. Study investigations were conducted before and 1y after surgery. Heart rhythm and p-wave duration were measured through ECGs and 7-d-holters. EAT-volume and attenuation were determined on non-enhanced CT scans. Serum markers were quantified by ELISA. RESULTS Thirty-seven patients underwent surgery (age: 52.1 ± 5.9y; 27 women; no AF). Increased p-wave duration correlated with higher BMI, larger EAT volumes, and lower EAT attenuations (p < 0.05). Post-surgery, p-wave duration decreased from 109 ± 11 to 102 ± 11ms. Concurrently, EAT volume decreased from 132 ± 49 to 87 ± 52ml, BMI from 43.2 ± 5.2 to 28.9 ± 4.6kg/m2, and EAT attenuation increased from -76.1 ± 4.0 to -71.7 ± 4.4HU (p <0.001). Adiponectin increased from 8.7 ± 0.8 to 14.2 ± 1.0 μg/ml (p <0.001). However, decreased p-wave durations were not related to changed EAT characteristics, BMI or adiponectin. CONCLUSION In this explorative study, longer p-wave durations related to higher BMIs, larger EAT volume, and lower EAT attenuations. P-wave duration and EAT volume decreased, and EAT attenuation increased upon drastic weightloss. However, there was no relation between decreased p-wave duration and changed BMI or EAT characteristics.
Collapse
Affiliation(s)
- Eva R Meulendijks
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Carolina Janssen-Telders
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Departments of Cardiology, Heart Centre, Vrije Universiteit, Amsterdam UMC, Amsterdam, The Netherlands
| | - Elise L Hulsman
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nick Lobe
- Department of Radiology and Nuclear Medicine, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Pietro Zappala
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Marc M Terpstra
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Robin Wesselink
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Tim A C de Vries
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Departments of Cardiology, Heart Centre, Vrije Universiteit, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Cardiology, Rijnstate Hospital, Arnhem, The Netherlands
| | - Rushd F Al-Shama
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Ruben N van Veen
- Department of Surgery, Onze Lieve Vrouwe Gasthuis, locatie West, Amsterdam, The Netherlands
| | - Steve M M de Castro
- Department of Surgery, Onze Lieve Vrouwe Gasthuis, locatie West, Amsterdam, The Netherlands
| | - Claire E E de Vries
- Department of Surgery, Onze Lieve Vrouwe Gasthuis, locatie West, Amsterdam, The Netherlands
- Department of Plastic surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Leontien M G Nijland
- Department of Surgery, Onze Lieve Vrouwe Gasthuis, locatie West, Amsterdam, The Netherlands
| | - R Nils Planken
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Joris R de Groot
- Departments of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Centre, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Heart Failure and Arrhythmias, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Han R, Huang H, Zhu J, Jin X, Wang Y, Xu Y, Xia Z. Adipokines and their potential impacts on susceptibility to myocardial ischemia/reperfusion injury in diabetes. Lipids Health Dis 2024; 23:372. [PMID: 39538244 DOI: 10.1186/s12944-024-02357-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Coronary artery disease has a high mortality rate and is a striking public health concern, affecting a substantial portion of the global population. On the early onset of myocardial ischemia, thrombolytic therapy and coronary revascularization could promptly restore the bloodstream and nutrient supply to the ischemic tissue, efficiently preserving less severely injured myocardium. However, the abrupt re-establishment of blood flow triggers the significant discharge of previously accumulated oxidative substances and inflammatory cytokines, leading to further harm referred to as ischemia/reperfusion (I/R) injury. Diabetes significantly raises the vulnerability of the heart to I/R injury due to disrupted glucose and lipid processing, impaired insulin sensitivity and metabolic signaling, and increased inflammatory responses. Numerous studies have indicated that adipokines are crucial in the etiology and pathogenesis of obesity, diabetes, hyperlipidemia, hypertension, and coronary artery disease. Adipokines such as adiponectin, adipsin, visfatin, chemerin, omentin, and apelin, which possess protective properties against inflammatory activity and insulin resistance, have been shown to confer myocardial protection in conditions such as atherosclerosis, myocardial hypertrophy, myocardial I/R injury, and diabetic complications. On the other hand, adipokines such as leptin and resistin, known for their pro-inflammatory characteristics, have been linked to elevated cardiac lipid deposition, insulin resistance, and fibrosis. Meteorin-like (metrnl) exhibits opposite effects in various pathological conditions. However, the data on adipokines in myocardial I/R, especially in diabetes, is still incomplete and controversial. This review focuses on recent research regarding the categorization and function of adipokines in the heart muscle, and the identification of different signaling pathways involved in myocardial I/R injury under diabetic conditions, aiming to facilitate the exploration of therapeutic strategies against myocardial I/R injury in diabetes.
Collapse
Affiliation(s)
- Ronghui Han
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Hemeng Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Jianyu Zhu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Xiaogao Jin
- Department of Anesthesiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Yongyan Wang
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China.
- Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, Hengqin, Zhuhai, People's Republic of China.
- Faculty of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China.
| | - Zhengyuan Xia
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China.
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Wang H, Li C, Zhu L, Liu Z, Li N, Zheng Z, Liang S, Yan J. Adiponectin attenuates H2O2-induced apoptosis in chicken skeletal myoblasts through the lysosomal-mitochondrial axis. In Vitro Cell Dev Biol Anim 2024; 60:805-814. [PMID: 38427138 DOI: 10.1007/s11626-024-00857-8] [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: 09/13/2023] [Accepted: 11/20/2023] [Indexed: 03/02/2024]
Abstract
Adiponectin has previously been investigated for exerting its protective effect against myocardial injury through anti-apoptotic and anti-oxidative actions. Therefore, the present study aimed to investigate the nature and mechanism of adiponectin inhibition of H2O2-induced apoptosis in chicken skeletal myoblasts. Skeletal muscle satellite cells were differentiated and assigned into three groups. Group C was on the blank control group, group H was stimulated with the H2O2 (500 μmol/L, 4 h) alone group, group A + H was pre-treated with adiponectin (10 μg/mL, 24 h) and stimulated with the H2O2 (500 μmol/L, 4 h) group. Cytotoxicity inhibited by adiponectin was evaluated by the CCK-8 assay. The degree of apoptosis and oxidative damage was investigated by the TdT-mediated dUTP nick end labeling (TUNEL) and reactive oxygen species (ROS) staining assays. Oxidative stress was assessed by evaluating lipid peroxidation, superoxide dismutase, and reduced glutathione. Acridine orange (AO) staining detected lysosomal membrane permeability. The changes in mitochondrial membrane potential (MMP) were analyzed using 5,5,6,6'-tetrachloro-1,1,3,3-tetraethylimidacarbocyanine iodide (JC-1) dye under a fluorescence microscope. The lysosomal function, mitochondrial function, and apoptosis-related mRNA and protein expression levels were quantified by real-time quantitative PCR and western blot, respectively. The results suggested that adiponectin treatment attenuated H2O2-induced cytotoxicity and oxidative stress in skeletal myoblasts. Compared with H2O2 treatment, TUNEL and ROS staining demonstrated lower apoptosis upon adiponectin treatment. AO staining confirmed the amelioration of lysosomal membrane damage, and JC-1 staining revealed an increase in mitochondrial membrane potential after adiponectin treatment. At the molecular level, adiponectin treatment inhibited the expression of the lysosomal apoptotic factors cathepsin B, chymotrypsin B, and the mitochondrial apoptotic pathway cytochrome-c (cyt-c) and caspase-8; decreased the apoptotic marker gene Bax; and increased the expression of the anti-apoptotic marker gene Bcl-2. Adiponectin treatment attenuated H2O2-induced apoptosis in skeletal myoblasts, possibly by inhibiting oxidative stress and apoptosis through the lysosomal-mitochondrial axis.
Collapse
Affiliation(s)
- Han Wang
- Tianjin Key Laboratory of Animal Molecular Breeding and BiotechnologyTianjin Engineering Research Center of Animal Healthy FarmingInstitute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chi Li
- Tianjin Key Laboratory of Animal Molecular Breeding and BiotechnologyTianjin Engineering Research Center of Animal Healthy FarmingInstitute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Longbo Zhu
- Tianjin Key Laboratory of Animal Molecular Breeding and BiotechnologyTianjin Engineering Research Center of Animal Healthy FarmingInstitute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhengqun Liu
- Tianjin Key Laboratory of Animal Molecular Breeding and BiotechnologyTianjin Engineering Research Center of Animal Healthy FarmingInstitute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China.
| | - Ning Li
- Tianjin Key Laboratory of Animal Molecular Breeding and BiotechnologyTianjin Engineering Research Center of Animal Healthy FarmingInstitute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China
| | - Zi Zheng
- Tianjin Key Laboratory of Animal Molecular Breeding and BiotechnologyTianjin Engineering Research Center of Animal Healthy FarmingInstitute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China
| | - Shiyue Liang
- Tianjin Key Laboratory of Animal Molecular Breeding and BiotechnologyTianjin Engineering Research Center of Animal Healthy FarmingInstitute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China
| | - Jun Yan
- Tianjin Key Laboratory of Animal Molecular Breeding and BiotechnologyTianjin Engineering Research Center of Animal Healthy FarmingInstitute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China.
| |
Collapse
|
4
|
Fu L, Du J, Furkert D, Shipton ML, Liu X, Aguirre T, Chin AC, Riley AM, Potter BVL, Fiedler D, Zhang X, Zhu Y, Fu C. Depleting inositol pyrophosphate 5-InsP7 protected the heart against ischaemia-reperfusion injury by elevating plasma adiponectin. Cardiovasc Res 2024; 120:954-970. [PMID: 38252884 PMCID: PMC11218692 DOI: 10.1093/cvr/cvae017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 01/24/2024] Open
Abstract
AIMS Adiponectin is an adipocyte-derived circulating protein that exerts cardiovascular and metabolic protection. Due to the futile degradation of endogenous adiponectin and the challenges of exogenous administration, regulatory mechanisms of adiponectin biosynthesis are of significant pharmacological interest. METHODS AND RESULTS Here, we report that 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7) generated by inositol hexakisphosphate kinase 1 (IP6K1) governed circulating adiponectin levels via thiol-mediated protein quality control in the secretory pathway. IP6K1 bound to adiponectin and DsbA-L and generated 5-InsP7 to stabilize adiponectin/ERp44 and DsbA-L/Ero1-Lα interactions, driving adiponectin intracellular degradation. Depleting 5-InsP7 by either IP6K1 deletion or pharmacological inhibition blocked intracellular adiponectin degradation. Whole-body and adipocyte-specific deletion of IP6K1 boosted plasma adiponectin levels, especially its high molecular weight forms, and activated AMPK-mediated protection against myocardial ischaemia-reperfusion injury. Pharmacological inhibition of 5-InsP7 biosynthesis in wild-type but not adiponectin knockout mice attenuated myocardial ischaemia-reperfusion injury. CONCLUSION Our findings revealed that 5-InsP7 is a physiological regulator of adiponectin biosynthesis that is amenable to pharmacological intervention for cardioprotection.
Collapse
Affiliation(s)
- Lin Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Jimin Du
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - David Furkert
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Megan L Shipton
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Xiaoqi Liu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Tim Aguirre
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Alfred C Chin
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Andrew M Riley
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Barry V L Potter
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Dorothea Fiedler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Chenglai Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Yangpu District, Shanghai 200092, China
| |
Collapse
|
5
|
Agbaedeng TA, Iroga PE, Rathnasekara VM, Zacharia AL. Adipokines and stroke: A systematic review and meta-analysis of disease risk and patient outcomes. Obes Rev 2024; 25:e13684. [PMID: 38291816 DOI: 10.1111/obr.13684] [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: 08/09/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 02/01/2024]
Abstract
Obesity is reported to increase stroke risk, with adipocyte-derived cytokines or adipokines implicated as mediators. However, the relationship between adipokines and stroke is not well clarified. Thus, we aimed to evaluate the association of adipokines with stroke using fully adjusted risk estimates that incorporated body mass index in a meta-analysis. Data from 52 studies (62,428 patients) were pooled in a random-effects meta-analysis. Adiponectin was independently associated with a lower risk of pre-existing stroke (adjusted odds ratio: 0.64 [95% confidence interval: 0.46-0.88], p < 0.01), whereas leptin (1.08 [1.00-1.17], p = 0.04), resistin (1.06 [1.04-1.08], p < 0.01) and visfatin (1.04 [1.01-1.07], p = 0.01) are associated with a higher risk of stroke, but none with incident stroke. Adipokines independently associated with an ischaemic stroke subtype were adiponectin (0.48 [0.30-0.77], p < 0.01), leptin (1.10 [1.01-1.20], p = 0.04), and resistin (1.06 [1.04-1.08], p < 0.01). Fatty acid-binding protein-4 (FABP-4) independently predicted 6-month poor functional outcomes in stroke patients (adjusted hazard ratio: 1.09 [1.06-1.12], p < 0.01); whereas both FABP-4 (1.17 [1.03-1.34], p = 0.01) and visfatin (1.24 [1.00-1.55], p = 0.05) were predictive of 6-month mortality. Adipokines are associated with a greater risk of pre-existing stroke, but not with the relationship with incident stroke. Adipokines, such as FABP-4 and visfatin, may serve as biomarkers of stroke severity and worsening of stroke outcomes.
Collapse
Affiliation(s)
- Thomas A Agbaedeng
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Peter E Iroga
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | | | | |
Collapse
|
6
|
Yin T, Wang N, Jia F, Wu Y, Gao L, Zhang J, Hou R. Exosome-based WTAP siRNA delivery ameliorates myocardial ischemia-reperfusion injury. Eur J Pharm Biopharm 2024; 197:114218. [PMID: 38367759 DOI: 10.1016/j.ejpb.2024.114218] [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: 10/15/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/19/2024]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury is the primary cause of postischemicheartfailure. The increased expression of Thioredoxin-interacting protein (TXNIP) has been implicated in MI/R injury, although the detailed mechanism remains incompletely understood. In the present study, we observed the up-regulation of the m6A mRNA methylation complex component Wilms' tumor 1-associating protein (WTAP) in MI/R mice, which led to the m6A modification of TXNIP mRNA and an increase in mRNA abundance. Knock-down of WTAP resulted in a significant reduction in the m6A level of TXNIP mRNA and down-regulated TXNIP expression. Moreover, exosomes engineered with ischemic myocardium-targeting peptide (IMTP) were able to deliver WTAP siRNA into ischemic myocardial tissues, resulting in a specific gene knockdown and myocardial protection. In summary, our findings demonstrate that the WTAP-TXNIP regulatory axis plays a significant role in postischemicheartfailure, and the use of engineered exosomes targeting the ischemic heart shows promise as a strategy for siRNA therapy to protect the heart from injury.
Collapse
Affiliation(s)
- Tao Yin
- Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ning Wang
- Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fang Jia
- Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuchao Wu
- Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Gao
- Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rongrong Hou
- Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
7
|
Yue H, Zhang Q, Chang S, Zhao X, Wang M, Li W. Adiponectin protects against myocardial ischemia-reperfusion injury: a systematic review and meta-analysis of preclinical animal studies. Lipids Health Dis 2024; 23:51. [PMID: 38368320 PMCID: PMC10874037 DOI: 10.1186/s12944-024-02028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/22/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MIRI) is widespread in the treatment of ischemic heart disease, and its treatment options are currently limited. Adiponectin (APN) is an adipocytokine with cardioprotective properties; however, the mechanisms of APN in MIRI are unclear. Therefore, based on preclinical (animal model) evidence, the cardioprotective effects of APN and the underlying mechanisms were explored. METHODS The literature was searched for the protective effect of APN on MIRI in six databases until 16 November 2023, and data were extracted according to selection criteria. The outcomes were the size of the myocardial necrosis area and hemodynamics. Markers of oxidation, apoptosis, and inflammation were secondary outcome indicators. The quality evaluation was performed using the animal study evaluation scale recommended by the Systematic Review Center for Laboratory animal Experimentation statement. Stata/MP 14.0 software was used for the summary analysis. RESULTS In total, 20 papers with 426 animals were included in this study. The pooled analysis revealed that APN significantly reduced myocardial infarct size [weighted mean difference (WMD) = 16.67 (95% confidence interval (CI) = 13.18 to 20.16, P < 0.001)] and improved hemodynamics compared to the MIRI group [Left ventricular end-diastolic pressure: WMD = 5.96 (95% CI = 4.23 to 7.70, P < 0.001); + dP/dtmax: WMD = 1393.59 (95% CI = 972.57 to 1814.60, P < 0.001); -dP/dtmax: WMD = 850.06 (95% CI = 541.22 to 1158.90, P < 0.001); Left ventricular ejection fraction: WMD = 9.96 (95% CI = 7.29 to 12.63, P < 0.001)]. Apoptosis indicators [caspase-3: standardized mean difference (SMD) = 3.86 (95% CI = 2.97 to 4.76, P < 0.001); TUNEL-positive cells: WMD = 13.10 (95% CI = 8.15 to 18.05, P < 0.001)], inflammatory factor levels [TNF-α: SMD = 4.23 (95% CI = 2.48 to 5.98, P < 0.001)], oxidative stress indicators [Superoxide production: SMD = 4.53 (95% CI = 2.39 to 6.67, P < 0.001)], and lactate dehydrogenase levels [SMD = 2.82 (95% CI = 1.60 to 4.04, P < 0.001)] were significantly reduced. However, the superoxide dismutase content was significantly increased [SMD = 1.91 (95% CI = 1.17 to 2.65, P < 0.001)]. CONCLUSION APN protects against MIRI via anti-inflammatory, antiapoptotic, and antioxidant effects, and this effect is achieved by activating different signaling pathways.
Collapse
Affiliation(s)
- Hongyi Yue
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China
| | - Qunhui Zhang
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hunan, 421001, China
| | - Senhao Chang
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China
| | - Xinjie Zhao
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China
| | - Mengjie Wang
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China
| | - Wenhua Li
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China.
| |
Collapse
|
8
|
Engin A. Adiponectin Resistance in Obesity: Adiponectin Leptin/Insulin Interaction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:431-462. [PMID: 39287861 DOI: 10.1007/978-3-031-63657-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adiponectin (APN) levels in obesity are negatively correlated with chronic subclinical inflammation markers. The hypertrophic adipocytes cause obesity-linked insulin resistance and metabolic syndrome. Furthermore, macrophage polarization is a key determinant regulating adiponectin receptor (AdipoR1/R2) expression and differential adiponectin-mediated macrophage inflammatory responses in obese individuals. In addition to decrease in adiponectin concentrations, the decline in AdipoR1/R2 messenger ribonucleic acid (mRNA) expression leads to a decrement in adiponectin binding to cell membrane, and this turns into attenuation in the adiponectin effects. This is defined as APN resistance, and it is linked with insulin resistance in high-fat diet-fed subjects. The insulin-resistant group has a significantly higher leptin-to-APN ratio. The leptin-to-APN ratio is more than twofold higher in obese individuals. An increase in expression of AdipoRs restores insulin sensitivity and β-oxidation of fatty acids via triggering intracellular signal cascades. The ratio of high molecular weight to total APN is defined as the APN sensitivity index (ASI). This index is correlated to insulin sensitivity. Homeostasis model of assessment (HOMA)-APN and HOMA-estimated insulin resistance (HOMA-IR) are the most suitable methods to estimate the metabolic risk in metabolic syndrome. While morbidly obese patients display a significantly higher plasma leptin and soluble (s)E-selectin concentrations, leptin-to-APN ratio, there is a significant negative correlation between leptin-to-APN ratio and sP-selectin in obese patients. When comparing the metabolic dysregulated obese group with the metabolically healthy obese group, postprandial triglyceride clearance, insulin resistance, and leptin resistance are significantly delayed following the oral fat tolerance test in the first group. A neuropeptide, Spexin (SPX), is positively correlated with the quantitative insulin sensitivity check index (QUICKI) and APN. APN resistance together with insulin resistance forms a vicious cycle. Despite normal or high APN levels, an impaired post-receptor signaling due to adaptor protein-containing pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif 1 (APPL1)/APPL2 may alter APN efficiency and activity. However, APPL2 blocks adiponectin signaling through AdipoR1 and AdipoR2 because of the competitive inhibition of APPL1. APPL1, the intracellular binding partner of AdipoRs, is also an important mediator of adiponectin-dependent insulin sensitization. The elevated adiponectin levels with adiponectin resistance are compensatory responses in the condition of an unusual discordance between insulin resistance and APN unresponsiveness. Hypothalamic recombinant adeno-associated virus (rAAV)-leptin (Lep) gene therapy reduces serum APN levels, and it is a more efficient strategy for long-term weight maintenance.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
9
|
Begum M, Choubey M, Tirumalasetty MB, Arbee S, Mohib MM, Wahiduzzaman M, Mamun MA, Uddin MB, Mohiuddin MS. Adiponectin: A Promising Target for the Treatment of Diabetes and Its Complications. Life (Basel) 2023; 13:2213. [PMID: 38004353 PMCID: PMC10672343 DOI: 10.3390/life13112213] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Diabetes mellitus, a chronic metabolic disorder characterized by hyperglycemia, presents a formidable global health challenge with its associated complications. Adiponectin, an adipocyte-derived hormone, has emerged as a significant player in glucose metabolism and insulin sensitivity. Beyond its metabolic effects, adiponectin exerts anti-inflammatory, anti-oxidative, and vasoprotective properties, making it an appealing therapeutic target for mitigating diabetic complications. The molecular mechanisms by which adiponectin impacts critical pathways implicated in diabetic nephropathy, retinopathy, neuropathy, and cardiovascular problems are thoroughly examined in this study. In addition, we explore possible treatment options for increasing adiponectin levels or improving its downstream signaling. The multifaceted protective roles of adiponectin in diabetic complications suggest its potential as a novel therapeutic avenue. However, further translational studies and clinical trials are warranted to fully harness the therapeutic potential of adiponectin in the management of diabetic complications. This review highlights adiponectin as a promising target for the treatment of diverse diabetic complications and encourages continued research in this pivotal area of diabetes therapeutics.
Collapse
Affiliation(s)
- Mahmuda Begum
- Department of Internal Medicine, HCA-St David’s Medical Center, 919 E 32nd St, Austin, TX 78705, USA;
| | - Mayank Choubey
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA; (M.C.); (M.B.T.); (M.W.)
| | - Munichandra Babu Tirumalasetty
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA; (M.C.); (M.B.T.); (M.W.)
| | - Shahida Arbee
- Institute for Molecular Medicine, Aichi Medical University, 1-Yazako, Karimata, Aichi, Nagakute 480-1103, Japan;
| | - Mohammad Mohabbulla Mohib
- Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Magdeburger Straße 6, 06112 Halle, Germany;
| | - Md Wahiduzzaman
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA; (M.C.); (M.B.T.); (M.W.)
| | - Mohammed A. Mamun
- CHINTA Research Bangladesh, Savar 1342, Bangladesh;
- Department of Public Health and Informatics, Jahangirnagar University, Savar 1342, Bangladesh
| | - Mohammad Borhan Uddin
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh;
| | - Mohammad Sarif Mohiuddin
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA; (M.C.); (M.B.T.); (M.W.)
| |
Collapse
|
10
|
He H, Liu P, Li P. Dexmedetomidine Ameliorates Cardiac Ischemia/Reperfusion Injury by Enhancing Autophagy Through Activation of the AMPK/SIRT3 Pathway. Drug Des Devel Ther 2023; 17:3205-3218. [PMID: 37908314 PMCID: PMC10613569 DOI: 10.2147/dddt.s428024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023] Open
Abstract
Objective Myocardial ischemia-reperfusion (I/R) injury is a detrimental disease, resulting in high morbidity and mortality globally. In this study, we aimed to investigate the role of Dex in mitigating cardiac I/R injury. Methods H9c2 cells were treated with Dex (1 μM) for 24 h followed by oxygen-glucose deprivation/re-oxygenation (OGD/R). ANP and BNP mRNA of H9c2 cells and the LDH release were measured. Apoptosis of H9c2 cells was analyzed by flow cytometry. Mitochondrial membrane potential and superoxide production were detected by JC-1 staining and MitoSOXTM Red, respectively. Cell aerobic respiration was measured using Seahorse analysis. In vivo, mice were injected with Dex (25 μg/kg, i.p., once daily) for 5 days and then subjected to heart I/R. Heart function was analyzed by echocardiography. CK-MB and LDH were measured by Elisa. Infarct size was measured using TTC-Evans blue staining. Mitochondrial ultrastructure was observed using transmission electron microscopy. DHE staining, SOD activity, the content of MDA, and the content of GSH/GSSG of heart were measured to evaluate the oxidative stress. In addition, inflammatory cytokines were measured in vivo and in vitro. Furthermore, AMPK, SIRT3, and autophagy-related protein expression in the heart were detected by Western blot. Results Dex reduced the H9c2 cells injury exposed to OGD/R, accompanied by improved mitochondrial function and membrane potential. In vivo, Dex improved heart function, myocardial injury, and the mitochondria ultrastructure following I/R injury. Meanwhile, Dex inhibited myocardial oxidative stress and inflammation in the myocardial I/R. Furthermore, Compound C (an AMPK inhibitor) could inhibit Dex-induced autophagy in the I/R heart and the 3-MA (an autophagy inhibitor) could partially interfere with the effects of Dex on the protection of I/R heart. Conclusion Dex suppressed oxidative stress and inflammation by promoting autophagy through activating the AMPK/SIRT3 pathway, thus protecting the heart against the I/R injury.
Collapse
Affiliation(s)
- Hong He
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072People’s Republic of China
| | - Peng Liu
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610044People’s Republic of China
| | - Peng Li
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072People’s Republic of China
| |
Collapse
|
11
|
Zhang J, Liu L, Dong Z, Lu X, Hong W, Liu J, Zou X, Gao J, Jiang H, Sun X, Hu K, Yang Y, Ge J, Luo X, Sun A. An ischemic area-targeting, peroxynitrite-responsive, biomimetic carbon monoxide nanogenerator for preventing myocardial ischemia-reperfusion injury. Bioact Mater 2023; 28:480-494. [PMID: 37408796 PMCID: PMC10318466 DOI: 10.1016/j.bioactmat.2023.05.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
Myocardial ischemia-reperfusion (MI/R) injury is common in patients who undergo revascularization therapy for myocardial infarction, often leading to cardiac dysfunction. Carbon monoxide (CO) has emerged as a therapeutic molecule due to its beneficial properties such as anti-inflammatory, anti-apoptotic, and mitochondrial biogenesis-promoting properties. However, its clinical application is limited due to uncontrolled release, potential toxicity, and poor targeting efficiency. To address these limitations, a peroxynitrite (ONOO-)-triggered CO donor (PCOD585) is utilized to generate a poly (lactic-co-glycolic acid) (PLGA)-based, biomimetic CO nanogenerator (M/PCOD@PLGA) that is coated with the macrophage membrane, which could target to the ischemic area and neutralize proinflammatory cytokines. In the ischemic area, local produced ONOO- triggers the continuous release of CO from M/PCOD@PLGA, which efficiently ameliorates MI/R injury by clearing harmful ONOO-, attenuating the inflammatory response, inhibiting cardiomyocyte apoptosis, and promoting mitochondrial biogenesis. This study provides a novel insight into the safe therapeutic use of CO for MI/R injury by utilizing a novel CO donor combined with biomimetic technology. The M/PCOD@PLGA nanogenerator offers targeted delivery of CO to the ischemic area, minimizing potential toxicity and enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Jinyan Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Liwei Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Xicun Lu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenxuan Hong
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Jin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Xiaoyi Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Jinfeng Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Hao Jiang
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Youjun Yang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Xiao Luo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Viral Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| |
Collapse
|
12
|
Du MF, Zhang X, Hu GL, Mu JJ, Chu C, Liao YY, Chen C, Wang D, Ma Q, Yan Y, Jia H, Wang KK, Sun Y, Niu ZJ, Man ZY, Wang L, Zhang XY, Luo WJ, Gao WH, Li H, Wu GJ, Gao K, Zhang J, Wang Y. Associations of lipid accumulation product, visceral adiposity index, and triglyceride-glucose index with subclinical organ damage in healthy Chinese adults. Front Endocrinol (Lausanne) 2023; 14:1164592. [PMID: 37795361 PMCID: PMC10546403 DOI: 10.3389/fendo.2023.1164592] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023] Open
Abstract
Background and aims Obesity is an independent risk factor for cardiovascular disease development. Here, we aimed to examine and compare the predictive values of three novel obesity indices, lipid accumulation product (LAP), visceral adiposity index (VAI), and triglyceride-glucose (TyG) index, for cardiovascular subclinical organ damage. Methods A total of 1,773 healthy individuals from the Hanzhong Adolescent Hypertension Study cohort were enrolled. Anthropometric, biochemical, urinary albumin-to-creatinine ratio (uACR), brachial-ankle pulse wave velocity (baPWV), and Cornell voltage-duration product data were collected. Furthermore, the potential risk factors for subclinical organ damage were investigated, with particular emphasis on examining the predictive value of the LAP, VAI, and TyG index for detecting subclinical organ damage. Results LAP, VAI, and TyG index exhibited a significant positive association with baPWV and uACR. However, only LAP and VAI were found to have a positive correlation with Cornell product. While the three indices did not show an association with electrocardiographic left ventricular hypertrophy, higher values of LAP and TyG index were significantly associated with an increased risk of arterial stiffness and albuminuria. Furthermore, after dividing the population into quartiles, the fourth quartiles of LAP and TyG index showed a significant association with arterial stiffness and albuminuria when compared with the first quartiles, in both unadjusted and fully adjusted models. Additionally, the concordance index (C-index) values for LAP, VAI, and TyG index were reasonably high for arterial stiffness (0.856, 0.856, and 0.857, respectively) and albuminuria (0.739, 0.737, and 0.746, respectively). Lastly, the analyses of continuous net reclassification improvement (NRI) and integrated discrimination improvement (IDI) demonstrated that the TyG index exhibited significantly higher predictive values for arterial stiffness and albuminuria compared with LAP and VAI. Conclusion LAP, VAI, and, especially, TyG index demonstrated utility in screening cardiovascular subclinical organ damage among Chinese adults in this community-based sample. These indices have the potential to function as markers for early detection of cardiovascular disease in otherwise healthy individuals.
Collapse
Affiliation(s)
- Ming-Fei Du
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xi Zhang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Gui-Lin Hu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jian-Jun Mu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chao Chu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yue-Yuan Liao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chen Chen
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dan Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Qiong Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yu Yan
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hao Jia
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ke-Ke Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yue Sun
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ze-Jiaxin Niu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zi-Yue Man
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lan Wang
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, China
| | - Xiao-Yu Zhang
- Department of Cardiology, Northwest Women’s and Children’s Hospital of Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wen-Jing Luo
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wei-Hua Gao
- Department of Cardiology, Xi’an No.1 Hospital, Xi’an, China
| | - Hao Li
- Department of Critical Care Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Guan-Ji Wu
- Department of Cardiology, Xi’an Central Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ke Gao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jie Zhang
- Department of Cardiology, Xi’an People’s Hospital, Xi’an, China
| | - Yang Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
13
|
Meng Z, Liang B, Wu Y, Liu C, Wang H, Du Y, Gan L, Gao E, Lau WB, Christopher TA, Lopez BL, Koch WJ, Ma X, Zhao F, Wang Y, Zhao J. Hypoadiponectinemia-induced upregulation of microRNA449b downregulating Nrf-1 aggravates cardiac ischemia-reperfusion injury in diabetic mice. J Mol Cell Cardiol 2023; 182:1-14. [PMID: 37437402 PMCID: PMC10566306 DOI: 10.1016/j.yjmcc.2023.06.004] [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: 10/01/2022] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023]
Abstract
Diabetes enhances myocardial ischemic/reperfusion (MI/R) injury via an incompletely understood mechanism. Adiponectin (APN) is a cardioprotective adipokine suppressed by diabetes. However, how hypoadiponectinemia exacerbates cardiac injury remains incompletely understood. Dysregulation of miRNAs plays a significant role in disease development. However, whether hypoadiponectinemia alters cardiac miRNA profile, contributing to diabetic heart injury, remains unclear. Methods and Results: Wild-type (WT) and APN knockout (APN-KO) mice were subjected to MI/R. A cardiac microRNA profile was determined. Among 23 miRNAs increased in APN-KO mice following MI/R, miR-449b was most significantly upregulated (3.98-fold over WT mice). Administrating miR-449b mimic increased apoptosis, enlarged infarct size, and impaired cardiac function in WT mice. In contrast, anti-miR-449b decreased apoptosis, reduced infarct size, and improved cardiac function in APN-KO mice. Bioinformatic analysis predicted 73 miR-449b targeting genes, and GO analysis revealed oxidative stress as the top pathway regulated by these genes. Venn analysis followed by luciferase assay identified Nrf-1 and Ucp3 as the two most important miR-449b targets. In vivo administration of anti-miR-449b in APN-KO mice attenuated MI/R-stimulated superoxide overproduction. In vitro experiments demonstrated that high glucose/high lipid and simulated ischemia/reperfusion upregulated miR-449b and inhibited Nrf-1 and Ucp3 expression. These pathological effects were attenuated by anti-miR-449b or Nrf-1 overexpression. In a final attempt to validate our finding in a clinically relevant model, high-fat diet (HFD)-induced diabetic mice were subjected to MI/R and treated with anti-miR-449b or APN. Diabetes significantly increased miR-449b expression and downregulated Nrf-1 and Ucp3 expression. Administration of anti-miR-449b or APN preserved cardiac Nrf-1 expression, reduced cardiac oxidative stress, decreased apoptosis and infarct size, and improved cardiac function. Conclusion: We demonstrated for the first time that hypoadiponectinemia upregulates miR-449b and suppresses Nrf-1/Ucp3 expression, promoting oxidative stress and exacerbating MI/R injury in this population. Dysregulated APN/miR-449b/oxidative stress pathway is a potential therapeutic target against diabetic MI/R injury.
Collapse
Affiliation(s)
- Zhijun Meng
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Bin Liang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Yalin Wu
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, United States of America
| | - Caihong Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Han Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Yunhui Du
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Erhe Gao
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, United States of America
| | - Wayne B Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Theodore A Christopher
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Bernard L Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Walter J Koch
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, United States of America
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Fujie Zhao
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, United States of America
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America; Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, United States of America.
| | - Jianli Zhao
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, United States of America.
| |
Collapse
|
14
|
Vasamsetti SB, Natarajan N, Sadaf S, Florentin J, Dutta P. Regulation of cardiovascular health and disease by visceral adipose tissue-derived metabolic hormones. J Physiol 2023; 601:2099-2120. [PMID: 35661362 PMCID: PMC9722993 DOI: 10.1113/jp282728] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/04/2022] [Indexed: 11/08/2022] Open
Abstract
Visceral adipose tissue (VAT) is a metabolic organ known to regulate fat mass, and glucose and nutrient homeostasis. VAT is an active endocrine gland that synthesizes and secretes numerous bioactive mediators called 'adipocytokines/adipokines' into systemic circulation. These adipocytokines act on organs of metabolic importance like the liver and skeletal muscle. Multiple preclinical and in vitro studies showed strong evidence of the roles of adipocytokines in the regulation of metabolic disorders like diabetes, obesity and insulin resistance. Adipocytokines, such as adiponectin and omentin, are anti-inflammatory and have been shown to prevent atherogenesis by increasing nitric oxide (NO) production by the endothelium, suppressing endothelium-derived inflammation and decreasing foam cell formation. By inhibiting differentiation of vascular smooth muscle cells (VSMC) into osteoblasts, adiponectin and omentin prevent vascular calcification. On the other hand, adipocytokines like leptin and resistin induce inflammation and endothelial dysfunction that leads to vasoconstriction. By promoting VSMC migration and proliferation, extracellular matrix degradation and inflammatory polarization of macrophages, leptin and resistin increase the risk of atherosclerotic plaque vulnerability and rupture. Additionally, the plasma concentrations of these adipocytokines alter in ageing, rendering older humans vulnerable to cardiovascular disease. The disturbances in the normal physiological concentrations of these adipocytokines secreted by VAT under pathological conditions impede the normal functions of various organs and affect cardiovascular health. These adipokines could be used for both diagnostic and therapeutic purposes in cardiovascular disease.
Collapse
Affiliation(s)
- Sathish Babu Vasamsetti
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
| | - Niranjana Natarajan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
| | - Samreen Sadaf
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
| | - Jonathan Florentin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA, 15213
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA, 15213
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
15
|
Peng J, Chen Q, Wu C. The role of adiponectin in cardiovascular disease. Cardiovasc Pathol 2023; 64:107514. [PMID: 36634790 DOI: 10.1016/j.carpath.2022.107514] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease (CVD) is a common disease that seriously threatens the health of human beings, especially middle-aged and elderly people over 50 years old. It has the characteristics of high prevalence, high disability rate and high mortality rate. Previous studies have shown that adiponectin has therapeutic effects on a variety of CVDs. As a key adipokine, adiponectin, is an abundant peptide-regulated hormone that is mainly released by adipocytes and cardiomyocytes, as well as endothelial and skeletal cells. Adiponectin can protect against CVD by improving lipid metabolism, protecting vascular endothelial cells and inhibiting foam cell formation and vascular smooth muscle cell proliferation. Further investigation of the molecular and cellular mechanisms underlying the adiponectin system may provide new ideas for the treatment of CVD. Herein, this review aims to describe the structure and function of adiponectin and adiponectin receptors, introduce the function of adiponectin in the protection of cardiovascular disease and analyze the potential use and clinical significance of this hormone in the protection and treatment of cardiovascular disease, which shows that adiponectin can be expected to become a new therapeutic target and biomarker for the diagnosis and treatment of CVD.
Collapse
Affiliation(s)
- Jin Peng
- Clinical Medical Research Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qian Chen
- Clinical Medical Research Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Chuncao Wu
- Insititution of Chinese Materia Medica Preparation, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China.
| |
Collapse
|
16
|
Gurevitz C, Assali A, Mohsan J, Gmach SF, Beigel R, Ovdat T, Zwas DR, Kornowski R, Orvin K, Eisen A. The obesity paradox in patients with acute coronary syndromes over 2 decades - the ACSIS registry 2000-2018. Int J Cardiol 2023; 380:48-55. [PMID: 36940822 DOI: 10.1016/j.ijcard.2023.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/05/2023] [Accepted: 03/17/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Obesity is a worldwide epidemic which is associated with major cardiovascular (CV) risk factors. Nevertheless, substantial distant data, mostly published more than a decade ago, have demonstrated an obesity paradox, where obese patients generally have a better short- and long-term prognosis than do their leaner counterparts with the same CV profile. Nonetheless, it is not fully elucidated whether the obesity paradox is still relevant in the contemporary cardiology era among patients with acute coronary syndrome (ACS). We aimed to examine temporal trends in the clinical outcomes of ACS patients by their BMI status. METHODS Data from the ACSIS registry including all patients with calculated BMI data between the years 2002-2018. Patients were stratified by BMI groups to underweight, normal, overweight and obese. Clinical endpoints included 30d major cardiovascular events (MACE), and 1-year mortality. Temporal trends were examined in the late (2010-2018) vs. the early period (2002-2008). Multivariable models examined factors associated with clinical outcomes by BMI status. RESULTS Among the 13,816 patients from the ACSIS registry with available BMI data, 104 were underweight, 3921 were normal weight, 6224 were overweight and 3567 were obese. 1-year mortality was highest among underweight patients (24.8%), as compared to normal weight patients (10.7%) and lowest among overweight and obese patients (7.1% and 7.5% respectively; p for trend <0.001). 30-day MACE rates followed a similar pattern (24.3% for underweight, 13.6% for normal weight, 11.6% for overweight, and 11.7% for obese; p for trend<0.001). Comparing the 2 time-periods, 30-day MACE was significantly lower in the late period in all BMI groups, but unchanged in patients who were underweight. Similarly, 1-year mortality has decreased in normal weight and obese patients but remained similarly high in underweight patients. CONCLUSIONS In ACS patients, during 2-decades, 30-day MACE and 1-year mortality were lower among overweight and obese patients compared to underweight and even normal weight patients. Temporal trends revealed that 30-day MACE and 1-year mortality have decreased among all BMI groups other than the underweight ACS patients, among whom the adverse CV rates were consistently high. Our findings suggest that the obesity paradox is still relevant in ACS patients in the current cardiology era.
Collapse
Affiliation(s)
- Chen Gurevitz
- Cardiology division, Rabin Medical Center, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Aseel Assali
- Internal medicine division, Sourasky Medical Center, Tel-Aviv, Israel
| | - Jamil Mohsan
- Cardiology department, Hillel Yaffe Medical Center, Hadera, Israel
| | | | - Roy Beigel
- Cardiology department, Sheba Medical Center, Ramat-Gan, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Ovdat
- Cardiology department, Sheba Medical Center, Ramat-Gan, Israel
| | - Donna R Zwas
- Cardiology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Ran Kornowski
- Cardiology division, Rabin Medical Center, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Katia Orvin
- Cardiology division, Rabin Medical Center, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alon Eisen
- Cardiology division, Rabin Medical Center, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Saeedi-Boroujeni A, Purrahman D, Shojaeian A, Poniatowski ŁA, Rafiee F, Mahmoudian-Sani MR. Progranulin (PGRN) as a regulator of inflammation and a critical factor in the immunopathogenesis of cardiovascular diseases. J Inflamm (Lond) 2023; 20:1. [PMID: 36658641 PMCID: PMC9851114 DOI: 10.1186/s12950-023-00327-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/15/2023] [Indexed: 01/20/2023] Open
Abstract
Immune dysregulation has been identified as a critical cause of the most common types of cardiovascular diseases (CVDs). Notably, the innate and adaptive immune responses under physiological conditions are typically regulated with high sensitivity to avoid the exacerbation of inflammation, but any dysregulation can probably be associated with CVDs. In this respect, progranulin (PGRN) serves as one of the main components of the regulation of inflammatory processes, which significantly contributes to the immunopathogenesis of such disorders. PGRN has been introduced among the secreted growth factors as one related to wound healing, inflammation, and human embryonic development, as well as a wide variety of autoimmune diseases. The relationship between the serum PGRN and TNF-α ratio with the spontaneous bacterial peritonitis constitute one of the independent predictors of these conditions. The full-length PGRN can thus effectively reduce the calcification of valve interstitial cells, and the granulin precursor (GRN), among the degradation products of PGRN, can be beneficial. Moreover, it was observed that, PGRN protects the heart against ischemia-reperfusion injury. Above all, PGRN also provides protection in the initial phase following myocardial ischemia-reperfusion injury. The protective impact of PGRN on this may be associated with the early activation of the PI3K/Akt signaling pathway. PGRN also acts as a protective factor in hyperhomocysteinemia, probably by down-regulating the wingless-related integration site Wnt/β-catenin signaling pathway. Many studies have further demonstrated that SARS-CoV-2 (COVID-19) has dramatically increased the risks of CVDs due to inflammation, so PGRN has drawn much more attention among scholars. Lysosomes play a pivotal role in the inflammation process, and PGRN is one of the key regulators in their functioning, which contributes to the immunomodulatory mechanism in the pathogenesis of CVDs. Therefore, investigation of PGRN actions can help find new prospects in the treatment of CVDs. This review aims to summarize the role of PGRN in the immunopathogenesis of CVD, with an emphasis on its treatment.
Collapse
Affiliation(s)
- Ali Saeedi-Boroujeni
- Department of Microbiology, School of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Daryush Purrahman
- grid.411230.50000 0000 9296 6873Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Shojaeian
- grid.411950.80000 0004 0611 9280Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Łukasz A. Poniatowski
- grid.491786.50000 0001 0211 9062Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Neubrandenburg, Germany
| | - Fatemeh Rafiee
- grid.469309.10000 0004 0612 8427Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Science, Zanjan, Iran
| | - Mohammad-Reza Mahmoudian-Sani
- grid.411230.50000 0000 9296 6873Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran ,grid.411230.50000 0000 9296 6873Clinical Research Development Unit, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
18
|
Zheng S, Shen M, Qian Y, Li S, Chen Y, Jiang H, Lv H, Chen D, Zhao R, Zheng X, Sun M, Yang T, Shi Y, Fu Q. Growth differentiation factor-15/adiponectin ratio as a potential biomarker for metabolic syndrome in Han Chinese. Front Endocrinol (Lausanne) 2023; 14:1146376. [PMID: 37152921 PMCID: PMC10154592 DOI: 10.3389/fendo.2023.1146376] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Aims Growth differentiation factor-15 (GDF-15) and adiponectin are adipokines that regulate metabolism. This study aimed to evaluate the roles of GDF-15, adiponectin, and GDF-15/adiponectin ratio (G/A ratio) as biomarkers for detecting metabolic syndrome (MS). Materials and methods This cross-sectional study included 676 participants aged 20-70 years in Jurong, China. The participants were divided into four groups based on sex and age (<40 and ≥40 years). MS was defined according to the modified National Cholesterol Education Program Adult Treatment Panel III criteria. Receiver operating characteristic curves were used to evaluate the performance of GDF-15, adiponectin, and the G/A ratio in predicting MS. Results The prevalence of MS was 22.0% (149/676). Logistic regression analysis indicated that the G/A ratio and adiponectin levels, but not GDF-15 levels, were correlated with MS [odds ratio; 95% CI 1.010 (1.006-1.013) and 0.798 (0.735-0.865), respectively] after adjusting for confounding factors. The G/A ratio displayed a significant relationship with MS in each subgroup and with each MS component in both men and women; however, adiponectin concentrations were significantly associated with MS and all its components only in men (all P <0.05). The area under the curve (AUC) of the G/A ratio and the adiponectin level for MS was 0.758 and 0.748, respectively. The highest AUC was 0.757 for the adiponectin level in men and 0.724 for the G/A ratio in women. Conclusions This study suggests that the G/A ratio and adiponectin are potential biomarkers for detecting MS in women and men, respectively.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yun Shi
- *Correspondence: Qi Fu, ; Yun Shi,
| | - Qi Fu
- *Correspondence: Qi Fu, ; Yun Shi,
| |
Collapse
|
19
|
Han W, Yang S, Xiao H, Wang M, Ye J, Cao L, Sun G. Role of Adiponectin in Cardiovascular Diseases Related to Glucose and Lipid Metabolism Disorders. Int J Mol Sci 2022; 23:15627. [PMID: 36555264 PMCID: PMC9779180 DOI: 10.3390/ijms232415627] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Lifestyle changes have led to increased incidence of cardiovascular disease (CVD); therefore, potential targets against CVD should be explored to mitigate its risks. Adiponectin (APN), an adipokine secreted by adipose tissue, has numerous beneficial effects against CVD related to glucose and lipid metabolism disorders, including regulation of glucose and lipid metabolism, increasing insulin sensitivity, reduction of oxidative stress and inflammation, protection of myocardial cells, and improvement in endothelial cell function. These effects demonstrate the anti-atherosclerotic and antihypertensive properties of APN, which could aid in improving myocardial hypertrophy, and reducing myocardial ischemia/reperfusion (MI/R) injury and myocardial infarction. APN can also be used for diagnosing and predicting heart failure. This review summarizes and discusses the role of APN in the treatment of CVD related to glucose and lipid metabolism disorders, and explores future APN research directions and clinical application prospects. Future studies should elucidate the signaling pathway network of APN cardiovascular protective effects, which will facilitate clinical trials targeting APN for CVD treatment in a clinical setting.
Collapse
Affiliation(s)
- Wen Han
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Shuxian Yang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Haiyan Xiao
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Jingxue Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Li Cao
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| |
Collapse
|
20
|
Laukkanen JA, Kunutsor SK, Hernesniemi J, Immonen J, Eskola M, Zaccardi F, Niemelä M, Mäkikallio T, Hagnäs M, Piuhola J, Juvonen J, Sia J, Rummukainen J, Kervinen K, Karvanen J, Nikus K. Underweight and obesity are related to higher mortality in patients undergoing coronary angiography: The KARDIO invasive cardiology register study. Catheter Cardiovasc Interv 2022; 100:1242-1251. [PMID: 36378689 PMCID: PMC10098486 DOI: 10.1002/ccd.30463] [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: 07/22/2022] [Revised: 09/10/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND In patients with some cardiovascular disease conditions, slightly elevated body mass index (BMI) is associated with a lower mortality risk (termed "obesity paradox"). It is uncertain, however, if this obesity paradox exists in patients who have had invasive cardiology procedures. We evaluated the association between BMI and mortality in patients who underwent coronary angiography. METHODS We utilised the KARDIO registry, which comprised data on demographics, prevalent diseases, risk factors, coronary angiographies, and interventions on 42,636 patients. BMI was categorised based on WHO cut-offs or transformed using P-splines. Hazard ratios (HRs) with 95% confidence intervals (CIs) were estimated for all-cause mortality. RESULTS During a median follow-up of 4.9 years, 4688 all-cause deaths occurred. BMI was nonlinearly associated with mortality risk: compared to normal weight category (18.5-25 kg/m2 ), the age-adjusted HRs (95% CIs) for all-cause mortality were 1.90 (1.49, 2.43), 0.96 (0.92, 1.01), 1.04 (0.99, 1.09), 1.08 (0.96, 1.20), and 1.45 (1.22, 1.72) for underweight (<18.5 kg/m2 ), preobesity (25 to <30 kg/m2 ), obesity class I (30 to <35 kg/m2 ), obesity class II (35 to <40 kg/m2 ), and obesity class III (>40 kg/m2 ), respectively. The corresponding multivariable adjusted HRs (95% CIs) were 2.00 (1.55, 2.58), 0.92 (0.88, 0.97) 1.01 (0.95, 1.06), 1.10 (0.98, 1.23), and 1.49 (1.26, 1,78), respectively. CONCLUSIONS In patients undergoing coronary angiography, underweight and obesity class III are associated with increased mortality risk, and the lowest mortality was observed in the preobesity class. It appears the obesity paradox may be present in patients who undergo invasive coronary procedures.
Collapse
Affiliation(s)
- Jari A. Laukkanen
- School of MedicineInstitute of Clinical Medicine, University of Eastern FinlandKuopioFinland
- Central Finland Health Care District, Department of MedicineJyväskyläFinland
| | - Setor K. Kunutsor
- Central Finland Health Care District, Department of MedicineJyväskyläFinland
- Leicester Real World Evidence Unit, Diabetes Research CentreUniversity of Leicester, Leicester General HospitalLeicesterUK
- Translational Health Sciences, Bristol Medical SchoolUniversity of Bristol, Learning & Research Building (Level 1), Southmead HospitalBristolUK
| | - Jussi Hernesniemi
- Heart Center, Department of CardiologyTampere University HospitalTampereFinland
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Jaakko Immonen
- Central Finland Health Care District, Department of MedicineJyväskyläFinland
| | - Markku Eskola
- Heart Center, Department of CardiologyTampere University HospitalTampereFinland
| | - Francesco Zaccardi
- Central Finland Health Care District, Department of MedicineJyväskyläFinland
- Leicester Real World Evidence Unit, Diabetes Research CentreUniversity of Leicester, Leicester General HospitalLeicesterUK
| | - Matti Niemelä
- Division of Cardiology, Department of Internal MedicineOulu University HospitalOuluFinland
| | - Timo Mäkikallio
- Department of MedicineSouth‐Karelia Central HospitalLappeenrantaFinland
| | - Magnus Hagnäs
- Lapland Health Care District, Department of Internal MedicineRovaniemiFinland
| | - Jarkko Piuhola
- Division of Cardiology, Department of Internal MedicineOulu University HospitalOuluFinland
| | - Jukka Juvonen
- Department of Internal MedicineKainuu Central HospitalKajaaniFinland
| | - Jussi Sia
- Department of CardiologyKokkola Central HospitalKokkolaFinland
| | - Juha Rummukainen
- Department of Internal MedicineSatakunta Central HospitalPoriFinland
| | - Kari Kervinen
- Division of Cardiology, Department of Internal MedicineOulu University HospitalOuluFinland
| | - Juha Karvanen
- Department of Mathematics and StatisticsUniversity of JyvaskylaJyväskyläFinland
| | - Kjell Nikus
- Heart Center, Department of CardiologyTampere University HospitalTampereFinland
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | | |
Collapse
|
21
|
Zhou M, Zhang H, Chen H, Qi B. Adiponectin protects skeletal muscle from ischaemia–reperfusion injury in mice through
miR
‐21/
PI3K
/Akt signalling pathway. Int Wound J 2022; 20:1647-1661. [PMID: 36426910 PMCID: PMC10088838 DOI: 10.1111/iwj.14022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
Previous studies have confirmed that adiponectin (APN) plays a protective role in myocardial ischaemia-reperfusion (IR) injury, and the aim of this study was to investigate its effect on skeletal muscle. ELISA was used to detect the levels of Creatinine Kinase (CK), LDH, SOD and MDA in the plasma of the lower limbs of mice, and the levels of IL-6, IL-1β and TNF-α in the gastrocnemius. Quantitative PCR was used to detect the expression level of miR-21. TUNEL staining was used to detect the apoptosis of the gastrocnemius. The expression levels of apoptosis proteins, autophagy marker proteins and downstream target genes of miR-21 in gastrocnemius were detected by Western Blot. The results of this study revealed that APN levels were significantly reduced in gastrocnemius of IR mice. The oxidative stress, inflammatory response, apoptosis and autophagy induced by IR were significantly ameliorated by APN injection. The above effects of APN may be achieved through miR-21/PI3K signalling pathway, as found by interfering gene expression levels with miRNA antagomir and lentiviral injection. Taken together, our study revealed that APN protects skeletal muscle from IR injury through miR-21 /PI3K/Akt signalling pathway through inhibiting inflammatory response, apoptosis and autophagy.
Collapse
Affiliation(s)
- Min Zhou
- Department of Orthopedics Trauma and Microsurgery Zhongnan Hospital of Wuhan University Wuhan China
| | - Hao Zhang
- Department of Orthopedics Trauma and Microsurgery Zhongnan Hospital of Wuhan University Wuhan China
| | - Hairen Chen
- Department of Orthopedics Trauma and Microsurgery Zhongnan Hospital of Wuhan University Wuhan China
| | - Baiwen Qi
- Department of Orthopedics Trauma and Microsurgery Zhongnan Hospital of Wuhan University Wuhan China
| |
Collapse
|
22
|
Xia W, Li X, Wu Q, Xu A, Zhang L, Xia Z. The importance of caveolin as a target in the prevention and treatment of diabetic cardiomyopathy. Front Immunol 2022; 13:951381. [PMID: 36405687 PMCID: PMC9666770 DOI: 10.3389/fimmu.2022.951381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
The diabetic population has been increasing in the past decades and diabetic cardiomyopathy (DCM), a pathology that is defined by the presence of cardiac remodeling and dysfunction without conventional cardiac risk factors such as hypertension and coronary heart diseases, would eventually lead to fatal heart failure in the absence of effective treatment. Impaired insulin signaling, commonly known as insulin resistance, plays an important role in the development of DCM. A family of integral membrane proteins named caveolins (mainly caveolin-1 and caveolin-3 in the myocardium) and a protein hormone adiponectin (APN) have all been shown to be important for maintaining normal insulin signaling. Abnormalities in caveolins and APN have respectively been demonstrated to cause DCM. This review aims to summarize recent research findings of the roles and mechanisms of caveolins and APN in the development of DCM, and also explore the possible interplay between caveolins and APN.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xia Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Liangqing Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
23
|
Wang C, Du X, Fu F, Li X, Wang Z, Zhou Y, Gou L, Li W, Li J, Zhang J, Liao G, Li L, Han YP, Tong N, Liu J, Chen Y, Cheng J, Cao Q, Ilegems E, Lu Y, Zheng X, Berggren PO. Adiponectin gene therapy prevents islet loss after transplantation. J Cell Mol Med 2022; 26:4847-4858. [PMID: 35975481 PMCID: PMC9465193 DOI: 10.1111/jcmm.17515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 07/06/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Significant pancreatic islet dysfunction and loss shortly after transplantation to the liver limit the widespread implementation of this procedure in the clinic. Nonimmune factors such as reactive oxygen species and inflammation have been considered as the primary driving force for graft failure. The adipokine adiponectin plays potent roles against inflammation and oxidative stress. Previous studies have demonstrated that systemic administration of adiponectin significantly prevented islet loss and enhanced islet function at post‐transplantation period. In vitro studies indicate that adiponectin protects islets from hypoxia/reoxygenation injury, oxidative stress as well as TNF‐α‐induced injury. By applying adenovirus mediated transfection, we now engineered islet cells to express exogenous adiponectin gene prior to islet transplantation. Adenovirus‐mediated adiponectin transfer to a syngeneic suboptimal islet graft transplanted under kidney capsule markedly prevented inflammation, preserved islet graft mass and improved islet transplant outcomes. These results suggest that adenovirus‐mediated adiponectin gene therapy would be a beneficial clinical engineering approach for islet preservation in islet transplantation.
Collapse
Affiliation(s)
- Chengshi Wang
- Key Laboratory of Transplant Engineering and Immunology, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China.,Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojiong Du
- Department of Vascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fudong Fu
- West China Hospital, Institutes for Systems Genetics, Sichuan University, Chengdu, China
| | - Xiaoyu Li
- West China Hospital, Institutes for Systems Genetics, Sichuan University, Chengdu, China
| | - Zhenghao Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China.,The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Liping Gou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Juan Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayi Zhang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Guangneng Liao
- Key Laboratory of Transplant Engineering and Immunology, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan-Ping Han
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, The Center for Growth, Metabolism and Aging, The College of Life Sciences, Sichuan University, Chengdu, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Erwin Ilegems
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Per-Olof Berggren
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China.,The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
DDAH1/ADMA Regulates Adiponectin Resistance in Cerebral Ischemia via the ROS/FOXO1/APR1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2350857. [PMID: 35509834 PMCID: PMC9060971 DOI: 10.1155/2022/2350857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/14/2022] [Accepted: 03/30/2022] [Indexed: 11/27/2022]
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) protects against cerebral ischemia injury via regulating the level of asymmetric dimethylarginine (ADMA). This study is aimed at exploring the effect of adiponectin resistance on ADMA-induced neuronal loss in ischemic stroke (IS) and the underlying mechanism. DDAH1 knockout (DDAH1−/−) and wild-type (DDAH1+/+) rats underwent middle cerebral artery occlusion/reperfusion (MCAO/R). Plasma and brain adiponectin levels and the expressions of adiponectin receptor 1 (APR1), adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1), adenosine monophosphate-activated protein kinase (AMPK), and phosphorylated AMPK were determined after 24 h, 3 days, and 7 days. Neurological behavior, infarct volume, and adiponectin signaling were evaluated using adiponectin peptide or AdipoRon. The levels of reactive oxygen species (ROS) and Forkhead box O1 (FOXO1) (a transcription factor for APR1) were also assessed. An oxygen-glucose deprivation/reoxygenation (OGD/R) model was established in primary neurons. DDAH1 was overexpressed in neurons, after which FOXO1 expression, ROS production, adiponectin resistance, and cell viability were detected. DDAH1−/− rats showed no significant difference in adiponectin level in either plasma or brain after MCAO/R in DDAH1+/+ rats, but downregulated APR1 expression and suppressed adiponectin signaling were observed. AdipoRon, but not adiponectin peptide, attenuated the neurological deficits and adiponectin resistance in DDAH1−/− rats. ROS accumulation and phosphorylated FOXO1 expression also increased with DDAH1 depletion. Following DDAH1 overexpression, decreased cell viability and inhibited adiponectin signaling induced by OGD/R were alleviated in primary neurons, accompanied by reduced ROS production and phosphorylated FOXO1 expression. Our study elucidated that in IS, DDAH1 protected against adiponectin resistance in IS via the ROS/FOXO1/APR1 pathway.
Collapse
|
25
|
Zhao H, Chen X, Hu G, Li C, Guo L, Zhang L, Sun F, Xia Y, Yan W, Cui Z, Guo Y, Guo X, Huang C, Fan M, Wang S, Zhang F, Tao L. Small Extracellular Vesicles From Brown Adipose Tissue Mediate Exercise Cardioprotection. Circ Res 2022; 130:1490-1506. [PMID: 35387487 DOI: 10.1161/circresaha.121.320458] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Long-term exercise provides reliable cardioprotection via mechanisms still incompletely understood. Although traditionally considered a thermogenic tissue, brown adipose tissue (BAT) communicates with remote organs (eg, the heart) through its endocrine function. BAT expands in response to exercise, but its involvement in exercise cardioprotection remains undefined. OBJECTIVE This study investigated whether small extracellular vesicles (sEVs) secreted by BAT and their contained microRNAs (miRNAs) regulate cardiomyocyte survival and participate in exercise cardioprotection in the context of myocardial ischemia/reperfusion (MI/R) injury. METHODS AND RESULTS Four weeks of exercise resulted in a significant BAT expansion in mice. Surgical BAT ablation before MI/R weakened the salutary effects of exercise. Adeno-associated virus 9 vectors carrying short hairpin RNA targeting Rab27a (a GTPase required for sEV secretion) or control viruses were injected in situ into the interscapular BAT. Exercise-mediated protection against MI/R injury was greatly attenuated in mice whose BAT sEV secretion was suppressed by Rab27a silencing. Intramyocardial injection of the BAT sEVs ameliorated MI/R injury, revealing the cardioprotective potential of BAT sEVs. Discovery-driven experiments identified miR-125b-5p, miR-128-3p, and miR-30d-5p (referred to as the BAT miRNAs) as essential BAT sEV components for mediating cardioprotection. BAT-specific inhibition of the BAT miRNAs prevented their upregulation in plasma sEVs and hearts of exercised mice and attenuated exercise cardioprotection. Mechanistically, the BAT miRNAs cooperatively suppressed the proapoptotic MAPK (mitogen-associated protein kinase) pathway by targeting a series of molecules (eg, Map3k5, Map2k7, and Map2k4) in the signaling cascade. Delivery of BAT sEVs into hearts or cardiomyocytes suppressed MI/R-related MAPK pathway activation, an effect that disappeared with the combined use of the BAT miRNA inhibitors. CONCLUSIONS The sEVs secreted by BAT participate in exercise cardioprotection via delivering the cardioprotective miRNAs into the heart. These results provide novel insights into the mechanisms underlying the BAT-cardiomyocyte interaction and highlight BAT sEVs and their contained miRNAs as alternative candidates for exercise cardioprotection.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Department of Pharmacy, the 960th Hospital of the Logistics Support Force, Jinan, China (H.Z.)
| | - Xiyao Chen
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Department of Geriatrics, The Fourth Military Medical University, Xi'an, China. (X.C.)
| | - Guangyu Hu
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Congye Li
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Lanyan Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ling Zhang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Fangfang Sun
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Yunlong Xia
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Wenjun Yan
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ze Cui
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Yongzhen Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Xijing Hospital and Department of Toxicology, School of Public Health, The Fourth Military Medical University, Xi'an, China. (Y.G.)
| | - Xiong Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Chong Huang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Miaomiao Fan
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Shan Wang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Fuyang Zhang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ling Tao
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| |
Collapse
|
26
|
Agbaedeng TA, Zacharia AL, Iroga PE, Rathnasekara VM, Munawar DA, Bursill C, Noubiap JJ. Associations between adipokines and atrial fibrillation: A systematic review and meta-analysis. Nutr Metab Cardiovasc Dis 2022; 32:853-862. [PMID: 35227548 DOI: 10.1016/j.numecd.2022.01.019] [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: 09/24/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/24/2022]
Abstract
AIMS Although overweight and obesity are associated with increased risk of atrial fibrillation (AF), the underlying mechanisms are not well characterised. Recent data suggest that this link may be partly due to abnormal adipose tissue-derived cytokines or adipokines. However, this relationship is not well clarified. To evaluate the association between adipokines and AF in a systematic review and meta-analysis. DATA SYNTHESIS PubMed, Embase, and Web of Science Core Collection were searched from inception through 1st March 2021. Studies were included if they reported any adipokine and AF, with their quality assessed using the Newcastle-Ottawa scale. Data were independently abstracted, with unadjusted and multivariable adjusted estimates pooled in a random-effects meta-analysis. Data are presented for overall prevalent or incident AF and AF subtypes (paroxysmal, persistent, or non-paroxysmal AF). A total of 34 studies, with 31,479 patients, were included. The following adipokines were significantly associated with AF in the pooled univariate data - apelin (risk ratio for prevalent AF: 0.05 [0.00-0.50], p = 0.01; recurrent AF: 0.21 [0.11-0.42], p < 0.01) and resistin (incident AF: 2.05 [1.02-4.1], p = 0.04; prevalent AF: 2.62 [1.78-3.85], p < 0.01). Pooled analysis of multivariable adjusted effect size estimates showed adiponectin as the sole independent predictor of AF incidence (1.14 [1.02-1.27], p = 0.02). Moreover, adiponectin was associated with non-paroxysmal AF (persistent AF: 1.45 [1.08-1.94, p = 0.01; non-paroxysmal versus paroxysmal AF: 3.14 [1.87-5.27, p < 0.01). CONCLUSIONS Adipokines, principally adiponectin, apelin, and resistin, are associated with the risk of atrial fibrillation. However, the association is not seen after multivariate adjustment, likely reflecting the lack of statistical power. Future research should investigate these relationships in larger prospective cohorts and how they can refine AF monitoring strategies. PROSPERO ID CRD42020208879.
Collapse
Affiliation(s)
- Thomas A Agbaedeng
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia.
| | | | - Peter E Iroga
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | | | - Dian A Munawar
- Lyell McEwin Hospital, The University of Adelaide, Adelaide, Australia; Department of Cardiology and Vascular Medicine, University of Indonesia, Jakarta, Indonesia
| | - Christina Bursill
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia; Vascular Research Centre, Lifelong Health Theme, SAHMRI, Adelaide, Australia
| | - Jean Jacques Noubiap
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia; Centre for Heart Rhythm Disorders, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
27
|
Gruzdeva OV, Dyleva YA, Belik EV, Sinitsky MY, Stasev AN, Kokov AN, Brel NK, Krivkina EO, Bychkova EE, Tarasov RS, Barbarash OL. Relationship between Epicardial and Coronary Adipose Tissue and the Expression of Adiponectin, Leptin, and Interleukin 6 in Patients with Coronary Artery Disease. J Pers Med 2022; 12:jpm12020129. [PMID: 35207618 PMCID: PMC8877574 DOI: 10.3390/jpm12020129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Adipose tissue (AT) is an endocrine and paracrine organ that synthesizes biologically active adipocytokines, which affect inflammation, fibrosis, and atherogenesis. Epicardial and perivascular fat depots are of great interest to researchers, owing to their potential effects on the myocardium and blood vessels. The aim of the study was to assess the expression and secretion of adipocytokine genes in the AT of patients with coronary artery disease (CAD) and patients with aortic or mitral valve replacement. This study included 84 patients with CAD and 50 patients with aortic or mitral valve replacement. Adipocytes were isolated from subcutaneous, epicardial (EAT), and perivascular AT (PVAT), and were cultured for 24 h. EAT exhibited the lowest level of adiponectin gene expression and secretion, regardless of nosology, and high expression levels of the leptin gene and interleukin-6 (IL-6). However, EAT adipocytes in patients with CAD were characterized by more pronounced changes in comparison with the group with heart defects. High leptin and IL-6 levels resulted in increased pro-inflammatory activity, as observed in both EAT and PVAT adipocytes, especially in individuals with CAD. Therefore, our results revealed the pathogenetic significance of alterations in the adipokine and cytokine status of adipocytes of EAT and PVAT in patients with CAD.
Collapse
|
28
|
The Roles and Associated Mechanisms of Adipokines in Development of Metabolic Syndrome. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020334. [PMID: 35056647 PMCID: PMC8781412 DOI: 10.3390/molecules27020334] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome is a cluster of metabolic indicators that increase the risk of diabetes and cardiovascular diseases. Visceral obesity and factors derived from altered adipose tissue, adipokines, play critical roles in the development of metabolic syndrome. Although the adipokines leptin and adiponectin improve insulin sensitivity, others contribute to the development of glucose intolerance, including visfatin, fetuin-A, resistin, and plasminogen activator inhibitor-1 (PAI-1). Leptin and adiponectin increase fatty acid oxidation, prevent foam cell formation, and improve lipid metabolism, while visfatin, fetuin-A, PAI-1, and resistin have pro-atherogenic properties. In this review, we briefly summarize the role of various adipokines in the development of metabolic syndrome, focusing on glucose homeostasis and lipid metabolism.
Collapse
|
29
|
Tarkhnishvili A, Koentges C, Pfeil K, Gollmer J, Byrne NJ, Vosko I, Lueg J, Vogelbacher L, Birkle S, Tang S, Bon-Nawul Mwinyella T, Hoffmann MM, Odening KE, Michel NA, Wolf D, Stachon P, Hilgendorf I, Wallner M, Ljubojevic-Holzer S, von Lewinski D, Rainer P, Sedej S, Sourij H, Bode C, Zirlik A, Bugger H. Effects of Short Term Adiponectin Receptor Agonism on Cardiac Function and Energetics in Diabetic db/db Mice. J Lipid Atheroscler 2022; 11:161-177. [PMID: 35656151 PMCID: PMC9133777 DOI: 10.12997/jla.2022.11.2.161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/01/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Objective Impaired cardiac efficiency is a hallmark of diabetic cardiomyopathy in models of type 2 diabetes. Adiponectin receptor 1 (AdipoR1) deficiency impairs cardiac efficiency in non-diabetic mice, suggesting that hypoadiponectinemia in type 2 diabetes may contribute to impaired cardiac efficiency due to compromised AdipoR1 signaling. Thus, we investigated whether targeting cardiac adiponectin receptors may improve cardiac function and energetics, and attenuate diabetic cardiomyopathy in type 2 diabetic mice. Methods A non-selective adiponectin receptor agonist, AdipoRon, and vehicle were injected intraperitoneally into Eight-week-old db/db or C57BLKS/J mice for 10 days. Cardiac morphology and function were evaluated by echocardiography and working heart perfusions. Results Based on echocardiography, AdipoRon treatment did not alter ejection fraction, left ventricular diameters or left ventricular wall thickness in db/db mice compared to vehicle-treated mice. In isolated working hearts, an impairment in cardiac output and efficiency in db/db mice was not improved by AdipoRon. Mitochondrial respiratory capacity, respiration in the presence of oligomycin, and 4-hydroxynonenal levels were similar among all groups. However, AdipoRon induced a marked shift in the substrate oxidation pattern in db/db mice towards increased reliance on glucose utilization. In parallel, the diabetes-associated increase in serum triglyceride levels in vehicle-treated db/db mice was blunted by AdipoRon treatment, while an increase in myocardial triglycerides in vehicle-treated db/db mice was not altered by AdipoRon treatment. Conclusion AdipoRon treatment shifts myocardial substrate preference towards increased glucose utilization, likely by decreasing fatty acid delivery to the heart, but was not sufficient to improve cardiac output and efficiency in db/db mice.
Collapse
Affiliation(s)
| | - Christoph Koentges
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Katharina Pfeil
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Johannes Gollmer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Nikole J Byrne
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Ivan Vosko
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Julia Lueg
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Laura Vogelbacher
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Stephan Birkle
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Sibai Tang
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
| | | | - Michael M Hoffmann
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Center – University of Freiburg, Germany
| | - Katja E Odening
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Translational Cardiology, Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Nathaly Anto Michel
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dennis Wolf
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus Wallner
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Senka Ljubojevic-Holzer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Dirk von Lewinski
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter Rainer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Simon Sedej
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Cardiovascular Diabetology Research Group, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Christoph Bode
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Zirlik
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heiko Bugger
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Cosentino RG, Churilla JR, Josephson S, Molle-Rios Z, Hossain MJ, Prado WL, Balagopal PB. Branched-chain Amino Acids and Relationship With Inflammation in Youth With Obesity: A Randomized Controlled Intervention Study. J Clin Endocrinol Metab 2021; 106:3129-3139. [PMID: 34286837 DOI: 10.1210/clinem/dgab538] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Indexed: 12/31/2022]
Abstract
CONTEXT Elevated concentrations of branched-chain amino acids (BCAA) are strong predictors of type 2 diabetes mellitus (T2DM). Their association with cardiovascular disease (CVD) remains uncertain, particularly in youth. OBJECTIVE We investigated the role of BCAA and aromatic amino acids (AAA) in obesity, their relationships with novel biomarkers of CVD, and response to a physical activity-based lifestyle intervention (PAL-I) in a randomized controlled study in youth with normal weight (NW) and obesity (OB). METHODS Age (14-18 years) and Tanner stage (≥IV) matched youth (OB, n = 15 and NW, n = 6) were studied; the 15 participants with OB underwent a 3-month randomized controlled PAL-I. Circulating amino acid profile, glucose, insulin, lipids, adiponectin, retinol binding protein-4, fibrinogen, high-sensitivity C-reactive protein, interleukin-6, and 25-hydroxy vitamin-D, along with body composition, were measured at baseline and after PAL-I. Independent t tests, analysis of covariance, and mixed-effect models were used for analysis of the data. RESULTS Compared with NW, the concentration of various amino acids, including BCAA and AAA, were altered in OB (P < 0.05). BCAA and AAA showed baseline correlations with body composition and novel biomarkers of CVD, particularly inflammatory factors (all P < 0.05). The PAL-I produced only negligible effects (P > 0.05) on BCAA and AAA. Glutamine, glycine, and aspartic acid decreased with PAL-I (all P < 0.05). CONCLUSION The novel finding of the BCAA-inflammation relationship, along with strong correlations with nontraditional biomarkers of CVD, may raise the prospect of BCAA as a biomarker of CVD and evoke a potential link between obesity, T2DM, and CVD.
Collapse
Affiliation(s)
- Ralph G Cosentino
- Department of Clinical and Applied Movement Sciences, University of North Florida, Jacksonville, FL 32224, USA
| | - James R Churilla
- Department of Clinical and Applied Movement Sciences, University of North Florida, Jacksonville, FL 32224, USA
| | - Samantha Josephson
- Biomedical Research, Nemours Children's Health System, Jacksonville, FL 32207, USA
| | - Zarela Molle-Rios
- Division of Gastroenterology, Nemours Children's Health System, Wilmington, DE 19802, USA
| | - Md Jobayer Hossain
- Biomedical Research, Nemours Children's Health System, Wilmington, DE 19802, USA
| | - Wagner L Prado
- Department of Kinesiology, California State University, San Bernardino, CA 92407, USA
| | - P Babu Balagopal
- Biomedical Research, Nemours Children's Health System, Jacksonville, FL 32207, USA
- Department of Pediatrics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
31
|
Sharma A, Mah M, Ritchie RH, De Blasio MJ. The adiponectin signalling pathway - A therapeutic target for the cardiac complications of type 2 diabetes? Pharmacol Ther 2021; 232:108008. [PMID: 34610378 DOI: 10.1016/j.pharmthera.2021.108008] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/17/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022]
Abstract
Diabetes is associated with an increased risk of heart failure (HF). This is commonly termed diabetic cardiomyopathy and is often characterised by increased cardiac fibrosis, pathological hypertrophy, increased oxidative and endoplasmic reticulum stress as well as diastolic dysfunction. Adiponectin is a cardioprotective adipokine that is downregulated in settings of type 2 diabetes (T2D) and obesity. Furthermore, both adiponectin receptors (AdipoR1 and R2) are also downregulated in these settings which further results in impaired cardiac adiponectin signalling and reduced cardioprotection. In many cardiac pathologies, adiponectin signalling has been shown to protect against cardiac remodelling and lipotoxicity, however its cardioprotective actions in T2D-induced cardiomyopathy remain unresolved. Diabetic cardiomyopathy has historically lacked effective treatment options. In this review, we summarise the current evidence for links between the suppressed adiponectin signalling pathway and cardiac dysfunction, in diabetes. We describe adiponectin receptor-mediated signalling pathways that are normally associated with cardioprotection, as well as current and potential future therapeutic approaches that could target this pathway as possible interventions for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Abhipree Sharma
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Michael Mah
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Department of Pharmacology, Monash University, Clayton, VIC 3800, Australia; Department of Medicine, Monash University, Clayton, VIC 3800, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Department of Pharmacology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
32
|
Wang Y, Lau WB, Ma X. "Know Diabetes by Heart": role of adipocyte-cardiomyocyte communications. MEDICAL REVIEW (2021) 2021; 1:14-17. [PMID: 37724081 PMCID: PMC10471098 DOI: 10.1515/mr-2021-0008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/06/2021] [Indexed: 09/20/2023]
Abstract
Cardiovascular disease is the leading cause of morbidity in patients with diabetes mellitus. In 2019, the American Heart Association and the American Diabetes Association (along with industry leaders) launched the groundbreaking collaborative initiative "Know Diabetes by Heart™" to reduce cardiovascular deaths in type 2 diabetic patients. The molecular basis linking diabetes with cardiovascular complications has not yet been fully defined. Recent clinical and experimental studies strongly suggest that adipocyte dysfunction and subsequent pathological communications between adipocyte and cardiomyocytes play important roles in diabetic cardiac injury. This perspective article will review recent development concerning adipocyte-cardiomyocyte communications, and identify the most critical questions remain to be answered in this filed.
Collapse
Affiliation(s)
- Yajing Wang
- Department of Emergency Medicine,
Thomas Jefferson
University, Philadelphia 19107,
PA, USA
| | - Wayne Bond Lau
- Department of Emergency Medicine,
Thomas Jefferson
University, Philadelphia 19107,
PA, USA
| | - Xinliang Ma
- Department of Emergency Medicine,
Thomas Jefferson
University, Philadelphia 19107,
PA, USA
| |
Collapse
|
33
|
Bermúdez V, Durán P, Rojas E, Díaz MP, Rivas J, Nava M, Chacín M, Cabrera de Bravo M, Carrasquero R, Ponce CC, Górriz JL, D´Marco L. The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium. Front Endocrinol (Lausanne) 2021; 12:735070. [PMID: 34603210 PMCID: PMC8479191 DOI: 10.3389/fendo.2021.735070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue (AT) biology is linked to cardiovascular health since obesity is associated with cardiovascular disease (CVD) and positively correlated with excessive visceral fat accumulation. AT signaling to myocardial cells through soluble factors known as adipokines, cardiokines, branched-chain amino acids and small molecules like microRNAs, undoubtedly influence myocardial cells and AT function via the endocrine-paracrine mechanisms of action. Unfortunately, abnormal total and visceral adiposity can alter this harmonious signaling network, resulting in tissue hypoxia and monocyte/macrophage adipose infiltration occurring alongside expanded intra-abdominal and epicardial fat depots seen in the human obese phenotype. These processes promote an abnormal adipocyte proteomic reprogramming, whereby these cells become a source of abnormal signals, affecting vascular and myocardial tissues, leading to meta-inflammation, atrial fibrillation, coronary artery disease, heart hypertrophy, heart failure and myocardial infarction. This review first discusses the pathophysiology and consequences of adipose tissue expansion, particularly their association with meta-inflammation and microbiota dysbiosis. We also explore the precise mechanisms involved in metabolic reprogramming in AT that represent plausible causative factors for CVD. Finally, we clarify how lifestyle changes could promote improvement in myocardiocyte function in the context of changes in AT proteomics and a better gut microbiome profile to develop effective, non-pharmacologic approaches to CVD.
Collapse
Affiliation(s)
- Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Pablo Durán
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Edward Rojas
- Cardiovascular Division, University Hospital, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - María P. Díaz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - José Rivas
- Department of Medicine, Cardiology Division, University of Florida-College of Medicine, Jacksonville, FL, United States
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla, Colombia
| | | | - Rubén Carrasquero
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Clímaco Cano Ponce
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - José Luis Górriz
- Servicio de Nefrología, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Luis D´Marco
- Servicio de Nefrología, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
34
|
Polypeptide Globular Adiponectin Ameliorates Hypoxia/Reoxygenation-Induced Cardiomyocyte Injury by Inhibiting Both Apoptosis and Necroptosis. J Immunol Res 2021; 2021:1815098. [PMID: 34307691 PMCID: PMC8282401 DOI: 10.1155/2021/1815098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/16/2021] [Indexed: 01/11/2023] Open
Abstract
Adiponectin is a small peptide secreted and a key component of the endocrine system and immune system. Although globular adiponectin protects myocardial ischemia/reperfusion-induced cardiomyocyte injury, the protective mechanisms remain largely unresolved. Using a neonatal rat ventricular myocyte hypoxia/reoxygenation model, we investigated the role of its potential mechanisms of necroptosis in globular adiponectin-mediated protection in hypoxia/reoxygenation-induced cardiomyocyte injury as compared to apoptosis. We found that globular adiponectin treatment attenuated cardiomyocyte injury as indicated by increased cell viability and reduced lactate dehydrogenase release following hypoxia/reoxygenation. Immunofluorescence staining and Western blotting demonstrated that both necroptosis and apoptosis were triggered by hypoxia/reoxygenation and diminished by globular adiponectin. Necrostatin-1 (RIP1-specific inhibitor) and Z-VAD-FMK (pan-caspase inhibitor) only mimicked the inhibition of necroptosis and apoptosis, respectively, by globular adiponectin in hypoxia/reoxygenation-treated cardiomyocytes. Globular adiponectin attenuated reactive oxygen species production, oxidative damage, and p38MAPK and NF-κB signaling, all important for necroptosis and apoptosis. Collectively, our study suggests that globular adiponectin inhibits hypoxia/reoxygenation-induced necroptosis and apoptosis in cardiomyocytes probably by reducing oxidative stress and interrupting p38MAPK signaling.
Collapse
|
35
|
Caldwell JT, Jones KMD, Park H, Pinto JR, Ghosh P, Reid-Foley EC, Ulrich B, Delp MD, Behnke BJ, Muller-Delp JM. Aerobic exercise training reduces cardiac function and coronary flow-induced vasodilation in mice lacking adiponectin. Am J Physiol Heart Circ Physiol 2021; 321:H1-H14. [PMID: 33989084 DOI: 10.1152/ajpheart.00885.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We tested the hypothesis that adiponectin deficiency attenuates cardiac and coronary microvascular function and prevents exercise training-induced adaptations of the myocardium and the coronary microvasculature in adult mice. Adult wild-type (WT) or adiponectin knockout (adiponectin KO) mice underwent treadmill exercise training or remained sedentary for 8-10 wk. Systolic and diastolic functions were assessed before and after exercise training or cage confinement. Vasoreactivity of coronary resistance arteries was assessed at the end of exercise training or cage confinement. Before exercise training, ejection fraction and fractional shortening were similar in adiponectin KO and WT mice, but isovolumic contraction time was significantly lengthened in adiponectin KO mice. Exercise training increased ejection fraction (12%) and fractional shortening (20%) with no change in isovolumic contraction time in WT mice. In adiponectin KO mice, both ejection fraction (-9%) and fractional shortening (-12%) were reduced after exercise training and these decreases were coupled to a further increase in isovolumic contraction time (20%). In sedentary mice, endothelium-dependent dilation to flow was higher in arterioles from adiponectin KO mice as compared with WT mice. Exercise training enhanced dilation to flow in WT mice but decreased flow-induced dilation in adiponectin KO mice. These data suggest that compensatory mechanisms contribute to the maintenance of cardiac and coronary microvascular function in sedentary mice lacking adiponectin; however, in the absence of adiponectin, cardiac and coronary microvascular adaptations to exercise training are compromised.NEW & NOTEWORTHY We report that compensatory mechanisms contribute to the maintenance of cardiac and coronary microvascular function in sedentary mice in which adiponectin has been deleted; however, when mice lacking adiponectin are subjected to the physiological stress of exercise training, beneficial coronary microvascular and cardiac adaptations are compromised or absent.
Collapse
Affiliation(s)
- Jacob T Caldwell
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | | | - Hyerim Park
- Department of Nutrition, Food and Exercise Science, Florida State University, Tallahassee, Florida
| | - Jose R Pinto
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - Payal Ghosh
- Department of Nutrition, Food and Exercise Science, Florida State University, Tallahassee, Florida
| | - Emily C Reid-Foley
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - Brody Ulrich
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - Michael D Delp
- Department of Nutrition, Food and Exercise Science, Florida State University, Tallahassee, Florida
| | - Brad J Behnke
- Department of Kinesiology, Johnson Cancer Research Center, Kansas State University, Manhattan, Kansas
| | - Judy M Muller-Delp
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| |
Collapse
|
36
|
Tejedor S, Dolz‐Pérez I, Decker CG, Hernándiz A, Diez JL, Álvarez R, Castellano D, García NA, Ontoria‐Oviedo I, Nebot VJ, González‐King H, Igual B, Sepúlveda P, Vicent MJ. Polymer Conjugation of Docosahexaenoic Acid Potentiates Cardioprotective Therapy in Preclinical Models of Myocardial Ischemia/Reperfusion Injury. Adv Healthc Mater 2021; 10:e2002121. [PMID: 33720548 DOI: 10.1002/adhm.202002121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/16/2021] [Indexed: 01/16/2023]
Abstract
While coronary angioplasty represents an effective treatment option following acute myocardial infarction, the reperfusion of the occluded coronary artery can prompt ischemia-reperfusion (I/R) injury that significantly impacts patient outcomes. As ω-3 polyunsaturated fatty acids (PUFAs) have proven, yet limited cardioprotective abilities, an optimized polymer-conjugation approach is reported that improves PUFAs bioavailability to enhance cardioprotection and recovery in animal models of I/R-induced injury. Poly-l-glutamic acid (PGA) conjugation improves the solubility and stability of di-docosahexaenoic acid (diDHA) under physiological conditions and protects rat neonatal ventricular myocytes from I/R injury by reducing apoptosis, attenuating autophagy, inhibiting reactive oxygen species generation, and restoring mitochondrial membrane potential. Enhanced protective abilities are associated with optimized diDHA loading and evidence is provided for the inherent cardioprotective potential of PGA itself. Pretreatment with PGA-diDHA before reperfusion in a small animal I/R model provides for cardioprotection and limits area at risk (AAR). Furthermore, the preliminary findings suggest that PGA-diDHA administration in a swine I/R model may provide cardioprotection, limit edema and decrease AAR. Overall, the evaluation of PGA-diDHA in relevant preclinical models provides evidence for the potential of polymer-conjugated PUFAs in the mitigation of I/R injury associated with coronary angioplasty.
Collapse
Affiliation(s)
- Sandra Tejedor
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Irene Dolz‐Pérez
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| | - Caitlin G. Decker
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| | - Amparo Hernándiz
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Jose L. Diez
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Raquel Álvarez
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Delia Castellano
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Nahuel A. García
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Imelda Ontoria‐Oviedo
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Vicent J. Nebot
- Polypeptide Therapeutic Solutions S.L. Av. Benjamin Franklin 19, Paterna Valencia 46980 Spain
| | - Hernán González‐King
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Begoña Igual
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| |
Collapse
|
37
|
Roy B, Palaniyandi SS. Tissue-specific role and associated downstream signaling pathways of adiponectin. Cell Biosci 2021; 11:77. [PMID: 33902691 PMCID: PMC8073961 DOI: 10.1186/s13578-021-00587-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
According to the World Health Organization, metabolic syndrome (MetS) can be defined as a pathological condition characterized by abdominal obesity, insulin resistance, hypertension, and hyperlipidemia. The incidence of MetS keeps rising, as at least 35% of the USA population suffers from MetS. One of the worst comorbidities of metabolic syndrome are cardiovascular diseases that significantly amplifies the mortality associated with this syndrome. There is an urgent need to understand the pathophysiology of MetS to find novel diagnosis, treatment and management to mitigate the MetS and associated complications. Altered circulatory adiponectin levels have been implicated in MetS. Adiponectin has numerous biologic functions including antioxidative, anti-nitrative, anti-inflammatory, and cardioprotective effects. Being a pleiotropic hormone of multiple tissues, tissue-specific key signaling pathways of adiponectin will help finding specific target/s to blunt the pathophysiology of metabolic syndrome and associated disorders. The purpose of this review is to elucidate tissue-specific signaling pathways of adiponectin and possibly identify potential therapeutic targets for MetS as well as to evaluate the potential of adiponectin as a biomarker/therapeutic option in MetS.
Collapse
Affiliation(s)
- Bipradas Roy
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Integrative Biosciences Center (IBio), Room #3402, 6135 Woodward, Detroit, MI 48202 USA
- Department of Physiology, Wayne State University, Detroit, MI 48202 USA
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Integrative Biosciences Center (IBio), Room #3402, 6135 Woodward, Detroit, MI 48202 USA
- Department of Physiology, Wayne State University, Detroit, MI 48202 USA
| |
Collapse
|
38
|
Rozenbaum Z, Klein E, Cohen T, Shlomo N, Pereg D, Shuvy M. Temporal trends in management and outcomes of patients with acute coronary syndrome according to body mass index. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2021; 10:170-175. [PMID: 30663317 DOI: 10.1177/2048872619825569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/22/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Obesity is a major public health concern. We sought to investigate temporal trends in characteristics and outcomes of acute coronary syndrome patients according to body mass index. METHODS The study population consisted of patients who were included in the Acute Coronary Syndromes Israeli Surveys during 2000-2016. Patients were categorised into three groups according to body mass index: below 25 kg/m2, 25-30 kg/m2 (overweight) and above 30 kg/m2 (obese). Among each body mass index group the outcomes of two time frames were compared - early (2000-2006) versus late (2008-2016). RESULTS Overall 12,167 patients were included. Between the years 2000 and 2016, the percentage of obese patients increased from 20% to 30%. Obese patients were more frequently selected for an invasive approach, and had the lowest all-cause mortality rates. A significant reduction in 1-year mortality in recent compared to early surveys among patients with body mass index less than 25 kg/m2 and in obese patients but not for overweight patients was shown. Multivariable analysis showed that body mass index greater than 25 kg/m2 was associated with 30% lower 1-year mortality (hazard ratio 0.70, 95% confidence interval 0.55-0.90, P=0.005). CONCLUSION The prevalence of obesity among acute coronary syndrome patients has increased over the past two decades. A reduction of all-cause mortality was mainly seen in lean and obese patients.
Collapse
Affiliation(s)
- Zach Rozenbaum
- Department of Cardiology, Tel Aviv Sourasky Medical Center, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Eyal Klein
- Heart Institute, Hadassah Hebrew University Medical Center, Israel
| | - Tal Cohen
- Department of Cardiology, Sheba Medical Center, Israel
| | - Nir Shlomo
- Department of Cardiology, Sheba Medical Center, Israel
| | - David Pereg
- Sackler Faculty of Medicine, Tel-Aviv University, Israel
- Department Cardiology, Meir Medical Center, Israel
| | - Mony Shuvy
- Heart Institute, Hadassah Hebrew University Medical Center, Israel
| |
Collapse
|
39
|
Kim YS, Cho HH, Cho DI, Jeong HY, Lim SY, Jun JH, Kim MR, Kang BG, Cho M, Kang HJ, Kang WS, Oh GT, Ahn Y. The adipokine Retnla deficiency increases responsiveness to cardiac repair through adiponectin-rich bone marrow cells. Cell Death Dis 2021; 12:307. [PMID: 33753732 PMCID: PMC7985519 DOI: 10.1038/s41419-021-03593-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Resistin-like alpha (Retnla) is a member of the resistin family and known to modulate fibrosis and inflammation. Here, we investigated the role of Retnla in the cardiac injury model. Myocardial infarction (MI) was induced in wild type (WT), Retnla knockout (KO), and Retnla transgenic (TG) mice. Cardiac function was assessed by echocardiography and was significantly preserved in the KO mice, while worsened in the TG group. Angiogenesis was substantially increased in the KO mice, and cardiomyocyte apoptosis was markedly suppressed in the KO mice. By Retnla treatment, the expression of p21 and the ratio of Bax to Bcl2 were increased in cardiomyocytes, while decreased in cardiac fibroblasts. Interestingly, the numbers of cardiac macrophages and unsorted bone marrow cells (UBCs) were higher in the KO mice than in the WT mice. Besides, phosphorylated histone H3(+) cells were more frequent in bone marrow of KO mice. Moreover, adiponectin in UBCs was notably higher in the KO mice compared with WT mice. In an adoptive transfer study, UBCs were isolated from KO mice to transplant to the WT infarcted heart. Cardiac function was better in the KO-UBCs transplanted group in the WT-UBCs transplanted group. Taken together, proliferative and adiponectin-rich bone marrow niche was associated with substantial cardiac recovery by suppression of cardiac apoptosis and proliferation of cardiac fibroblast.
Collapse
Affiliation(s)
- Yong Sook Kim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Biomedical Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyang Hee Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Department of Molecular Medicine, Graduate School, Chonnam National University, Gwangju, Republic of Korea
| | - Dong Im Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Biomedical Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hye-Yun Jeong
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Soo Yeon Lim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Ju Hee Jun
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Mi Ra Kim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Bo Gyeong Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Meeyoung Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hye-Jin Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Wan Seok Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Youngkeun Ahn
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea. .,Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea. .,Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea.
| |
Collapse
|
40
|
Karamian M, Moossavi M, Hemmati M. From diabetes to renal aging: the therapeutic potential of adiponectin. J Physiol Biochem 2021; 77:205-214. [PMID: 33555532 DOI: 10.1007/s13105-021-00790-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022]
Abstract
Nowadays, the complications related to diabetes, such as nephropathy, cardiovascular problems, and aging, are highly being considered. Renal cell aging is affected by various mechanisms of inflammation, oxidative stress, and basement membrane thickening, which are significant causes of renal dysfunction in diabetes. Due to recent studies, adiponectin plays a key role in diabetes-related kidney diseases as a fat-derived hormone. In diabetes, reduced adiponectin levels are associated to renal cell aging. Oxidative stress and related signaling pathways are the main routes in which adiponectin may be effective to decline diabetes-associated aging. Therefore, adiponectin signaling in target tissues becomes one of the research areas of interest in metabolism and clinical medicine. Studies on adiponectin signaling will increase our understanding of adiponectin role in diabetes-linked diseases as well as shortening life span conditions which may guide the design of antidiabetic and anti-aging drugs.
Collapse
Affiliation(s)
- Mehdi Karamian
- Department of Parasitology and Mycology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Moossavi
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Mina Hemmati
- Department of Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
41
|
Adiponectin enhances the bioenergetics of cardiac myocytes via an AMPK- and succinate dehydrogenase-dependent mechanism. Cell Signal 2021; 78:109866. [PMID: 33271223 PMCID: PMC9619024 DOI: 10.1016/j.cellsig.2020.109866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
Adiponectin is one of the most abundant circulating hormones, which through adenosine monophosphate-activated protein kinase (AMPK), enhances fatty acid and glucose oxidation, and exerts a cardioprotective effect. However, its effects on cellular bioenergetics have not been explored. We have previously reported that 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside (AICAR, an AMPK activator) enhances mitochondrial respiration through a succinate dehydrogenase (SDH or complex II)-dependent mechanism in cardiac myocytes, leading us to predict that Adiponectin would exert a similar effect via activating AMPK. Our results show that Adiponectin enhances basal mitochondrial oxygen consumption rate (OCR), ATP production, and spare respiratory capacity (SRC), which were all abolished by the knockdown of AMPKγ1, inhibition of SDH complex assembly, via the knockdown of the SDH assembly factor 1 (Sdhaf1), or inhibition of SDH activity. Additionally, Adiponectin alleviated hypoxia-induced reductions in OCR and ATP production, in a Sdhaf1-dependent manner, whereas overexpression of Sdhaf1 confirmed its sufficiency for mediating these effects. Importantly, the levels of holoenzyme SDH under the various conditions correlated with OCR. We also show that the effects of Adiponectin, AMPK, Sdhaf1, as well as, SDH complex assembly all required sirtuin 3 (Sirt3). In conclusion, Adiponectin potentiates mitochondrial bioenergetics via promoting SDH complex assembly in an AMPK-, Sdhaf1-, and Sirt3-dependent fashion in cardiac myocytes.
Collapse
|
42
|
Elia E, Ministrini S, Carbone F, Montecucco F. Diabetic cardiomyopathy and inflammation: development of hostile microenvironment resulting in cardiac damage. Minerva Cardiol Angiol 2021; 70:357-369. [PMID: 33427423 DOI: 10.23736/s2724-5683.20.05454-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Diabetes mellitus is emerging as a major risk factor for heart failure. Diabetic cardiomyopathy is defined as a myocardial dysfunction that is not caused by underlying hypertension or coronary artery disease. Studies about clinical features, natural history and outcomes of the disease are few and often conflicting, because a universally accepted operative definition of diabetic cardiomyopathy is still lacking. Hyperglycemia and related metabolic and endocrine disorders are the triggering factors of myocardial damage in diabetic cardiomyopathy through multiple mechanisms. Among these mechanisms, inflammation has a relevant role, similar to other chronic myocardial disease, such as hypertensive or ischemic heart disease. A balance between inflammatory damage and healing processes is fundamental for homeostasis of myocardial tissue, whereas diabetes mellitus produces an imbalance, promoting inflammation and delaying healing. Therefore, diabetes-related chronic inflammatory state can produce a progressive qualitative deterioration of myocardial tissue, which reflects on progressive left ventricular functional impairment, which can be either diastolic, with prevalent myocardial hypertrophy, or systolic, with prevalent myocardial fibrosis. The aim of this narrative review is to summarize the existing evidence about the role of inflammation in diabetic cardiomyopathy onset and development. Ultimately, potential pharmacological strategies targeting inflammatory response will be reviewed and discussed.
Collapse
Affiliation(s)
- Edoardo Elia
- Division of Cardiology, Department of Internal Medicine, Città della Salute e della Scienza, Turin, Italy
| | - Stefano Ministrini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy - .,IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
43
|
Ratwatte S, Hyun K, D'Souza M, Barraclough J, Chew DP, Shetty P, Patel S, Amos D, Brieger D. Relation of Body Mass Index to Outcomes in Acute Coronary Syndrome. Am J Cardiol 2021; 138:11-19. [PMID: 33058799 DOI: 10.1016/j.amjcard.2020.09.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/24/2020] [Accepted: 09/30/2020] [Indexed: 12/20/2022]
Abstract
We assessed the association of BMI with all-cause and cardiovascular (CV) mortality in a contemporary acute coronary syndrome cohort. Patients from the Australian Cooperative National Registry of Acute Coronary Care, Guideline Adherence and Clinical Events and Global Registry of Acute Coronary Events between 2009 and 2019, were divided into BMI subgroups (underweight: <18.5, healthy: 18.5 to 24.9, overweight: 25 to 29.9, obese: 30 to 39.9, extremely obese: >40). Logistic regression was used to determine the association between BMI group and outcomes of all cause and CV death in hospital, and at 6 months. 8,503 patients were identified, mean age 64 ± 13, 72% male. The BMI breakdown was: underweight- 95, healthy- 2,140, overweight- 3,258, obese- 2,653, extremely obese- 357. Obese patients were younger (66 ± 12 vs 67 ± 13), with more hypertension, diabetes, and dyslipidemia vs healthy (all p < 0.05). Obese had lower hospital mortality than healthy: all-cause: 1% versus 4%, aOR (95% CI): 0.49(0.27, 0.87); CV: 1% versus 3%, 0.51(0.27, 0.96). At 6-month underweight had higher mortality than healthy: all-cause: 11% versus 4%, 2.69(1.26, 5.76); CV: 7% versus 1%, 3.54(1.19, 10.54); whereas obese had lower mortality: all-cause: 1% versus 4%, 0.48(0.29, 0.77); CV: 0.4% versus 1%, 0.42(0.19, 0.93). When BMI was plotted as a continuous variable against outcome a U-shaped relationship was demonstrated, with highest event rates in the most obese (>60). In conclusion, BMI is associated with mortality following an acute coronary syndrome. Obese patients had the best outcomes, suggesting persistence of the obesity paradox. However, there was a threshold effect, and favorable outcomes did not extend to the most obese.
Collapse
Affiliation(s)
- Seshika Ratwatte
- Department of Cardiology, Concord Repatriation General Hospital, Concord, Australia
| | - Karice Hyun
- ANZAC Research Institute, NSW, Australia; Westmead Applied Research Centre, University of Sydney, NSW, Australia
| | - Mario D'Souza
- University of Sydney, NSW, Australia; Clinical Research Centre, Sydney Local Health District, NSW, Australia
| | | | - Derek P Chew
- Department of Cardiology, Flinders University, Australia
| | - Pratap Shetty
- Department of Cardiology Wollongong Hospital, NSW, Australia
| | - Sanjay Patel
- Department of Cardiology Royal Prince Alfred Hospital, NSW, Australia
| | - David Amos
- Department of Cardiology, Orange Base Hospital, NSW, Australia
| | - David Brieger
- Department of Cardiology, Concord Repatriation General Hospital, Concord, Australia.
| |
Collapse
|
44
|
Mayer O, Seidlerová J, Bruthans J, Gelžinský J, Rychecká M, Mateřánková M, Karnosová P, Wohlfahrt P, Cífková R, Filipovský J. Is There Really an Association of High Circulating Adiponectin Concentration and Mortality or Morbidity Risk in Stable Coronary Artery Disease? Horm Metab Res 2020; 52:861-868. [PMID: 32746485 DOI: 10.1055/a-1212-8759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Adiponectin has several beneficial properties, namely, on the level of glucose metabolism, but paradoxically, its high concentrations were associated with increased mortality. We aimed to clarify the impact of high serum adiponectin on mortality and morbidity in patients with stable coronary artery heart disease (CAD). A total of 973 patients after myocardial infarction and/or coronary revascularization were followed in a prospective cohort study. All-cause and cardiovascular (CV) death, non-fatal cardiovascular events, and hospitalizations for heart failure (HF) were registered as outcomes. High serum adiponectin levels (≥8.58 ng/ml, i. e., above median) were independently associated with increased risk of 5-year all-cause, CV mortality or HF [with HRR 1.57 (95% CI: 1.07-2.30), 1.74 (95% CI: 1.08-2.81) or 1.94 (95% CI: 1.20-3.12), respectively] when adjusted just for conventional risk factors. However, its significance disappeared if brain natriuretic peptide (BNP) was included in a regression model. In line with this, we observed strong collinearity of adiponectin and BNP. Additionally, major adverse cardiovascular event (i. e., CV death, non-fatal myocardial infarction or stroke, coronary revascularization) incidence risk was not associated with high adiponectin. In conclusion, the observed inverse association between adiponectin concentrations and mortality risk seems to be attributable to concomitantly increased BNP, rather than high adiponectin being a causal factor.
Collapse
Affiliation(s)
- Otto Mayer
- 2nd Department of Internal Medicine, Faculty of Medicine in Pilsen, Charles University and University Hospital, Pilsen, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Jitka Seidlerová
- 2nd Department of Internal Medicine, Faculty of Medicine in Pilsen, Charles University and University Hospital, Pilsen, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Jan Bruthans
- 2nd Department of Internal Medicine, Faculty of Medicine in Pilsen, Charles University and University Hospital, Pilsen, Czech Republic
- Centre for Cardiovascular Prevention of the First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Julius Gelžinský
- 2nd Department of Internal Medicine, Faculty of Medicine in Pilsen, Charles University and University Hospital, Pilsen, Czech Republic
| | - Martina Rychecká
- 2nd Department of Internal Medicine, Faculty of Medicine in Pilsen, Charles University and University Hospital, Pilsen, Czech Republic
| | - Markéta Mateřánková
- 2nd Department of Internal Medicine, Faculty of Medicine in Pilsen, Charles University and University Hospital, Pilsen, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Petra Karnosová
- 2nd Department of Internal Medicine, Faculty of Medicine in Pilsen, Charles University and University Hospital, Pilsen, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Peter Wohlfahrt
- Centre for Cardiovascular Prevention of the First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Renata Cífková
- Centre for Cardiovascular Prevention of the First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Jan Filipovský
- 2nd Department of Internal Medicine, Faculty of Medicine in Pilsen, Charles University and University Hospital, Pilsen, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| |
Collapse
|
45
|
Zheng X, Liu D. Adiponectin alleviates the symptoms of ischemic renal disease by inhibiting renal cell apoptosis. Life Sci 2020; 265:118825. [PMID: 33275989 DOI: 10.1016/j.lfs.2020.118825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/06/2020] [Accepted: 11/21/2020] [Indexed: 11/15/2022]
Abstract
AIMS Ischemic renal disease (IRD) can cause kidney damage and eventually lead to end-stage renal disease. Adiponectin (APN), a recently discovered collagen-like protein secreted by adipose tissues, plays an important role in regulating energy metabolism and inflammation. This study aimed to explore the specific mechanism by which APN affects IRD. MAIN METHODS We cultured human renal tubular epithelial cells (HK-2) and created a mouse model of IRD to detect apoptosis-related indicators in vitro and in vivo. KEY FINDINGS Compared with those in the control group, the apoptosis rate and expression levels of Bax and Fas increased in the CoCl2-induced hypoxia model group. However, the expression of Bcl-2 decreased, and after the combined treatment with APN, the phenomenon mentioned above was reversed. Moreover, studies have found that stanniocalcin-1 (STC-1) and uncoupling protein3 (UCP3) are also involved in the protective effect of APN. Additionally, we found that the glomeruli of the mice were significantly enlarged after the APN gene was knocked out; furthermore, the number of collagen fibers in the renal tubules, as well as the expression of the corresponding fibrogenic factors, increased significantly. More importantly, after the knockout of the APN gene, the expression of the hypoxia-inducible factors HIF-1α and HIF-1β and the apoptotic rate of renal tissue cells also increased. SIGNIFICANCE These results indicate that APN can alleviate the symptoms of IRD by inhibiting renal cell apoptosis. Thus, in the future, APN may be a new target for the treatment of IRD. CHEMICAL COMPOUNDS Cobalt chloride (PubChem CID: 24643).
Collapse
Affiliation(s)
- Xiaotong Zheng
- Department of Nephrology, Shengjing Hospital of China Medical University, NO.39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning, PR China
| | - Dajun Liu
- Department of Nephrology, Shengjing Hospital of China Medical University, NO.39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning, PR China.
| |
Collapse
|
46
|
Wang WL, Ge TY, Chen X, Mao Y, Zhu YZ. Advances in the Protective Mechanism of NO, H 2S, and H 2 in Myocardial Ischemic Injury. Front Cardiovasc Med 2020; 7:588206. [PMID: 33195476 PMCID: PMC7661694 DOI: 10.3389/fcvm.2020.588206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022] Open
Abstract
Myocardial ischemic injury is among the top 10 leading causes of death from cardiovascular diseases worldwide. Myocardial ischemia is caused mainly by coronary artery occlusion or obstruction. It usually occurs when the heart is insufficiently perfused, oxygen supply to the myocardium is reduced, and energy metabolism in the myocardium is abnormal. Pathologically, myocardial ischemic injury generates a large number of inflammatory cells, thus inducing a state of oxidative stress. This sharp reduction in the number of normal cells as a result of apoptosis leads to organ and tissue damage, which can be life-threatening. Therefore, effective methods for the treatment of myocardial ischemic injury and clarification of the underlying mechanisms are urgently required. Gaseous signaling molecules, such as NO, H2S, H2, and combined gas donors, have gradually become a focus of research. Gaseous signaling molecules have shown anti-apoptotic, anti-oxidative and anti-inflammatory effects as potential therapeutic agents for myocardial ischemic injury in a large number of studies. In this review, we summarize and discuss the mechanism underlying the protective effect of gaseous signaling molecules on myocardial ischemic injury.
Collapse
Affiliation(s)
| | | | - Xu Chen
- Guilin Medical College, Guilin, China
| | - Yicheng Mao
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yi-Zhun Zhu
- Guilin Medical College, Guilin, China.,Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
47
|
Zhang Q, Zhang X, Ding N, Ge L, Dong Y, He C, Ding W. Globular adiponectin alleviates chronic intermittent hypoxia-induced H9C2 cardiomyocytes apoptosis via ER-phagy induction. Cell Cycle 2020; 19:3140-3153. [PMID: 33092434 DOI: 10.1080/15384101.2020.1836438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
This study evaluated the effects of endoplasmic reticulum autophagy (ER-phagy) and globular adiponectin (gAPN) on chronic intermittent hypoxia (CIH)-induced H9C2 cardiomyocytes injury while investigating potential mechanisms of action. The CIH model of H9C2 cardiomyocytes was established in this study. CCK-8 assay was used to determine cell viability post-exposure to various CIH times and gAPN concentrations. Flow cytometry was used to observe H9C2 cardiomyocytes apoptosis and immunofluorescence was used to measure ER-phagy and SEC62 activation. Western blot was used to observe ER stress and AMPK pathway. Results indicated that ER stress was activated in H9C2 cardiomyocytes exposed to CIH. Inhibition of ER stress reduced CIH-induced cell apoptosis. gAPN attenuated CIH-induced ER stress and H9C2 cardiomyocytes apoptosis. ER-phagy and SEC62 protein level were induced by CIH, while gAPN highly enhanced these changes. Inhibition of SEC62 expression reduced ER-phagy and increased ER stress and H9C2 cardiomyocytes apoptosis. Moreover, gAPN induced AMPK expression. Inhibition of AMPK expression reduced SEC62-mediated ER-phagy and increased the H9C2 cardiomyocytes apoptosis. Altogether, our study suggested that gAPN upregulated SEC62-mediated ER-phagy to extenuate ER stress, and mitigated H9C2 cardiomyocytes apoptosis induced by CIH through AMPK activation.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongda Hospital, Medical School, Southeast University , Nanjing, China
| | - Xilong Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University , Nanjing, China
| | - Ning Ding
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University , Nanjing, China
| | - Luyao Ge
- Department of Pulmonary and Critical Care Medicine, Zhongda Hospital, Medical School, Southeast University , Nanjing, China
| | - Yanbin Dong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University , Nanjing, China
| | - Can He
- Department of Pulmonary and Critical Care Medicine, Zhongda Hospital, Medical School, Southeast University , Nanjing, China
| | - Wenxiao Ding
- Department of Pulmonary and Critical Care Medicine, Zhongda Hospital, Medical School, Southeast University , Nanjing, China
| |
Collapse
|
48
|
Gao C, Wang R, Li B, Guo Y, Yin T, Xia Y, Zhang F, Lian K, Liu Y, Wang H, Zhang L, Gao E, Yan W, Tao L. TXNIP/Redd1 signalling and excessive autophagy: a novel mechanism of myocardial ischaemia/reperfusion injury in mice. Cardiovasc Res 2020; 116:645-657. [PMID: 31241142 DOI: 10.1093/cvr/cvz152] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/14/2019] [Accepted: 06/22/2019] [Indexed: 12/20/2022] Open
Abstract
AIMS Either insufficient or excessive autophagy causes cellular death and contributes to myocardial ischaemia/reperfusion (I/R) injury. However, mechanisms controlling the 'right-level' of autophagy in the heart remains unidentified. Thioredoxin-interacting protein (TXNIP) is a pro-oxidative molecule knowing to contribute to I/R injury. However, whether and how TXNIP may further inhibit suppressed autophagy or promote excessive cardiac autophagy in I/R heart has not been previously investigated. METHODS AND RESULTS Wild type or gene-manipulated adult male mice were subjected to myocardial I/R. TXNIP was increased in myocardium during I/R. Cardiac-specific TXNIP overexpression increased cardiomyocytes apoptosis and cardiac dysfunction, whereas cardiac-specific TXNIP knock-out significantly mitigated I/R-induced apoptosis and improved cardiac function. Importantly, TXNIP overexpression significantly promoted cardiac autophagy and TXNIP knock-out significantly inhibited cardiac autophagy. In vitro studies demonstrated that TXNIP increased autophagosome formation but inhibited autophagosome clearance during myocardial reperfusion. Atg5 siRNA significantly decreased hypoxia/reoxygenation induced apoptosis in cardiomyocytes with TXNIP overexpression. Mechanistically, TXNIP suppressed autophagosome clearance via increasing reactive oxygen species (ROS) level. However, TXNIP-increased autophagosome formation was not mediated by ROS as a ROS scavenger failed to block increased autophagosome formation in TXNIP overexpression heart. Finally, TXNIP directly interacted and stabilized Redd1 (an autophagy regulator), resulting in mTOR inhibition and autophagy activation. Redd1 knock-down significantly reduced autophagy formation and ameliorated I/R injury in TXNIP overexpression hearts. CONCLUSIONS Our results demonstrated that increased TXNIP-Redd1 expression is a novel signalling pathway that contributes to I/R injury by exaggerating excessive autophagy during reperfusion. These observations advance our understanding of the mechanisms of myocardial I/R injury.
Collapse
Affiliation(s)
- Chao Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Rutao Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Bing Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Yongzhen Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Tao Yin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Kun Lian
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Yi Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Han Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Rd, Xi'an 710032, China
| |
Collapse
|
49
|
Zhao D, Xue C, Li J, Feng K, Zeng P, Chen Y, Duan Y, Zhang S, Li X, Han J, Yang X. Adiponectin agonist ADP355 ameliorates doxorubicin-induced cardiotoxicity by decreasing cardiomyocyte apoptosis and oxidative stress. Biochem Biophys Res Commun 2020; 533:304-312. [PMID: 32958254 DOI: 10.1016/j.bbrc.2020.09.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Doxorubicin (DOX) is an anthracycline derivative and widely used as an anticancer drug. However, the severe cardiotoxicity of DOX limits its application. ADP355 is an adiponectin-based active peptide with anti-liver fibrosis and atherosclerosis properties. It remains unclear the effects and involved mechanisms of ADP355 in DOX-induced cardiotoxicity. C57BL/6J mice were intraperitoneally injected DOX once a week to induce heart failure while receiving ADP355 treatment daily for 4 weeks. At the end of experiment, blood and heart tissues were collected. We found that ADP355 markedly improved DOX-induced cardiac dysfunction and histopathological damage, and decreased serum creatine kinase, lactate dehydrogenase and hydroxybutyrate dehydrogenase levels. The anti-apoptotic activity of ADP355 was indicated by reduction in TUNEL-positive cells and cleaved caspase-3 expression, along with decreased BCL2-associated X protein/B cell lymphoma 2 (BAX/BCL2) levels in heart tissues. Additionally, ADP355 markedly increased DOX-decreased cell viability by reducing BAX/BCL2, but inhibited reactive oxygen species production in H9c2 cells. Mechanistically, ADP355 attenuated expression of DOX-reduced nuclear factor-erythroid 2-related factor 2 (Nrf2) and superoxide dismutase 2, as well as mRNA levels of Nrf2 downstream targets. Furthermore, ADP355 activated sirtuin 2 and its target genes. In conclusion, we demonstrate that ADP355 alleviates DOX-induced cardiotoxicity by inhibiting myocardial apoptosis and oxidative stress through Nrf2 and sirtuin 2 signaling pathways. These findings suggest that ADP355 can be a promising candidate for the treatment of cardiac dysfunction.
Collapse
Affiliation(s)
- Dan Zhao
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Chao Xue
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Jiaqi Li
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Ke Feng
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Peng Zeng
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoju Li
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Jihong Han
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
50
|
Ye JJ, Bian X, Lim J, Medzhitov R. Adiponectin and related C1q/TNF-related proteins bind selectively to anionic phospholipids and sphingolipids. Proc Natl Acad Sci U S A 2020; 117:17381-17388. [PMID: 32632018 PMCID: PMC7382265 DOI: 10.1073/pnas.1922270117] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Adiponectin (Acrp30) is an adipokine associated with protection from cardiovascular disease, insulin resistance, and inflammation. Although its effects are conventionally attributed to binding Adipor1/2 and T-cadherin, its abundance in circulation, role in ceramide metabolism, and homology to C1q suggest an overlooked role as a lipid-binding protein, possibly generalizable to other C1q/TNF-related proteins (CTRPs) and C1q family members. To investigate this, adiponectin, representative family members, and variants were expressed in Expi293 cells and tested for binding to lipids in liposomes using density centrifugation. Binding to physiological lipids were also analyzed using gradient ultracentrifugation, liquid chromatography-mass spectrometry, and shotgun lipidomics. Interestingly, adiponectin selectively bound several anionic phospholipids and sphingolipids, including phosphatidylserine, ceramide-1-phosphate, glucosylceramide, and sulfatide, via the C1q domain in an oligomerization-dependent fashion. Binding to lipids was observed in liposomes, low-density lipoproteins, cell membranes, and plasma. Other CTRPs and C1q family members (Cbln1, CTRP1, CTRP5, and CTRP13) also bound similar lipids. These findings suggest that adiponectin and CTRPs function not only as hormones, but also as lipid opsonins, as may other C1q family proteins.
Collapse
Affiliation(s)
- Jessica J Ye
- HHMI, Yale University, New Haven, CT 06520
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Xin Bian
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520
| | - Jaechul Lim
- HHMI, Yale University, New Haven, CT 06520
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Ruslan Medzhitov
- HHMI, Yale University, New Haven, CT 06520;
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|