1
|
Singal AM, Howard VJ, Judd SE, Carson AP, Zakai NA, Olson NC, Cushman M, Plante TB. Association between serum adiponectin and risk of incident hypertension: the REGARDS study. J Hypertens 2025:00004872-990000000-00684. [PMID: 40366102 DOI: 10.1097/hjh.0000000000004057] [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: 12/02/2024] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
INTRODUCTION Hypertension is a cardiovascular disease risk factor disproportionately affecting Black adults. Adiponectin is a cytokine secreted by adipocytes that improves insulin sensitivity, maintains vascular homeostasis, and is inversely associated with adiposity. We sought to determine the risk of incident hypertension by level of adiponectin. METHODS The REasons for Geographic And Racial Differences in Stroke (REGARDS) study recruited 30 239 adults from 2003 to 2007. We included REGARDS participants in the Biomarkers as Mediators of Racial Disparities in Risk Factors (BioMedioR) substudy. We estimated the risk ratio for incident hypertension in unadjusted and adjusted models for demographics factors, dietary patterns, measures of adiposity, and SBP. RESULTS Inverse odds ratio weighting estimated the excess hypertension incidence among Black participants that was explained by adiponectin. Of the 1498 BioMedioR participants, 35% developed incident hypertension in follow-up. White adults had higher baseline adiponectin levels than Black adults. For each 1-SD higher log adiponectin, the risk ratio of hypertension was 0.90 [95% confidence interval (95% CI) 0.84-0.96] in an unadjusted model, 0.92 (0.86-1.00) in a demographic adjusted model, and 0.99 (0.91-1.07) in a fully adjusted model. Lower adiponectin mediated 21-46% of the excess risk of incident hypertension among Black relative to White participants in models adjusting for just demographics and dietary patterns. CONCLUSION Among Black and White adults, lower adiponectin was associated with a greater risk of incident hypertension in unadjusted and minimally adjusted models. Future studies into how adiponectin changes in obesity could help to further explain its impact on hypertension risk.
Collapse
Affiliation(s)
- Aneesh M Singal
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts
| | | | - Suzanne E Judd
- University of Alabama at Birmingham, Birmingham, Alabama
| | - April P Carson
- University of Mississippi Medical Center, Jackson, Mississippi
| | - Neil A Zakai
- University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - Nels C Olson
- University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - Mary Cushman
- University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - Timothy B Plante
- University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| |
Collapse
|
2
|
Milena E, Maurizio M. Exploring the Cardiovascular Benefits of Extra Virgin Olive Oil: Insights into Mechanisms and Therapeutic Potential. Biomolecules 2025; 15:284. [PMID: 40001586 PMCID: PMC11852600 DOI: 10.3390/biom15020284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/01/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality worldwide, driven by complex interactions among genetic, environmental, and lifestyle factors, with diet playing a pivotal role. Extra Virgin Olive Oil (EVOO), a cornerstone of the Mediterranean diet (MedDiet), is a plant-based fat that has garnered attention for its robust cardiovascular benefits, which are attributed to its unique composition of monounsaturated fatty acids (MUFAs), particularly oleic acid (OA); and bioactive polyphenols, such as Hydroxytyrosol (HT) and oleocanthal. These compounds collectively exert antioxidant, anti-inflammatory, vasodilatory, and lipid-modulating effects. Numerous clinical and preclinical studies have demonstrated that EVOO's properties reduce major modifiable cardiovascular risk factors, including hypertension, dyslipidemia, obesity, and type 2 diabetes. EVOO also promotes endothelial function by increasing nitric oxide (NO) bioavailability, thus favoring vasodilation, lowering blood pressure (BP), and supporting vascular integrity. Furthermore, it modulates biomarkers of cardiovascular health, such as C-reactive protein, low-density lipoprotein (LDL) cholesterol, and NT-proBNP, aligning with improved hemostatic balance and reduced arterial vulnerability. Emerging evidence highlights its interaction with gut microbiota, further augmenting its cardioprotective effects. This review synthesizes current evidence, elucidating EVOO's multifaceted mechanisms of action and therapeutic potential. Future directions emphasize the need for advanced extraction techniques, nutraceutical formulations, and personalized dietary recommendations to maximize its health benefits. EVOO represents a valuable addition to dietary strategies aimed at reducing the global burden of cardiovascular diseases.
Collapse
Affiliation(s)
- Esposito Milena
- Department of Biology, Ecology & Earth Sciences, University of Calabria, 87036 Rende, Italy;
| | - Mandalà Maurizio
- Department of Biology, Ecology & Earth Sciences, University of Calabria, 87036 Rende, Italy;
- Department of Obstetrics, Gynecology and Reproductive Sciences, Larner College of Medicine, University of Vermont, Burlington, VT 05401, USA
| |
Collapse
|
3
|
Duan Y, Liu D, Yu H, Zhang S, Xia Y, Du Z, Qin Y, Wang Y, Ma X, Liu H, Du Y. Transcription and post-translational mechanisms: dual regulation of adiponectin-mediated Occludin expression in diabetes. Cell Biosci 2024; 14:126. [PMID: 39354565 PMCID: PMC11443667 DOI: 10.1186/s13578-024-01306-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Occludin, a crucial component of tight junctions, has emerged as a promising biomarker for the diagnosis of acute ischemic disease, highlighting its significant potential in clinical applications. In the diabetes, Occludin serves as a downstream target gene intricately regulated by the adiponectin (APN) signaling pathway. However, the specific mechanism by which adiponectin regulates Occludin expression remains unclear. METHODS AND RESULTS Endothelial-specific Ocln knockdown reduced APN-mediated blood flow recovery after femoral artery ligation and nullified APN's protection against high-fat diet (HFD)-triggered apoptosis and angiogenesis inhibition in vivo. Mechanically, we have meticulously elucidated APN's regulatory role in Occludin expression through a comprehensive analysis spanning transcriptional and post-translational dimensions. Foxo1 has been elucidated as a crucial transcriptional regulator of Occludin that is modulated by the APN/APPL1 signaling axis, as evidenced by validation through ChIP-qPCR assays and Western blot analysis. APN hindered Occludin degradation via the ubiquitin-proteasome pathway. Mass spectrometry analysis has recently uncovered a novel phosphorylation site, Tyr467, on Occludin. This site responds to APN, playing a crucial role in inhibiting Occludin ubiquitination by APN. The anti-apoptotic and pro-angiogenic effects of APN were attenuated in vitro and in vivo following Foxo1 knockdown or expression of a non-phosphorylatable mutant, OccludinY467A. Clinically, elevated plasma concentrations of Occludin were observed in patients with diabetes. A significant negative correlation was found between Occludin levels and APN concentrations. CONCLUSION Our study proposes that APN modulates Occludin expression through mechanisms involving both transcriptional and post-translational interactions, thereby conferring a protective effect on endothelial integrity within diabetic vasculature.
Collapse
Affiliation(s)
- Yanru Duan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Demin Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Huahui Yu
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, People's Republic of China
| | - Shihan Zhang
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, People's Republic of China
| | - Yihua Xia
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zhiyong Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, People's Republic of China
| | - Yanwen Qin
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, People's Republic of China
| | - Yajing Wang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing, China.
| | - Yunhui Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, People's Republic of China.
| |
Collapse
|
4
|
Rehman IU, Park JS, Choe K, Park HY, Park TJ, Kim MO. Overview of a novel osmotin abolishes abnormal metabolic-associated adiponectin mechanism in Alzheimer's disease: Peripheral and CNS insights. Ageing Res Rev 2024; 100:102447. [PMID: 39111409 DOI: 10.1016/j.arr.2024.102447] [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: 06/11/2024] [Revised: 07/19/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is a degenerative brain disease that affects millions of people worldwide. It is caused by abnormalities in cholinergic neurons, oxidative stress, and inflammatory cascades. The illness is accompanied by personality changes, memory issues, and dementia. Metabolic signaling pathways help with fundamental processes like DNA replication and RNA transcription. Being adaptable is essential for both surviving and treating illness. The body's metabolic signaling depends on adipokines, including adiponectin (APN) and other adipokines secreted by adipose tissues. Energy homeostasis is balanced by adipokines, and nutrients. Overconsumption of nutrients messes with irregular signaling of adipokines, such as APN in both peripheral and brain which leads to neurodegeneration, such as AD. Despite the failure of traditional treatments like memantine and cholinesterase inhibitors, natural plant bioactive substances like Osmotin (OSM) have been given a focus as potential therapeutics due to their antioxidant properties, better blood brain barrier (BBB) permeability, excellent cell viability, and especially nanoparticle approaches. The review highlights the published preclinical literature regarding the role of OSM in AD pathology while there is a need for more research to investigate the hidden therapeutic potential of OSM which may open a new gateway and further strengthen its healing role in the pathogenesis of neurodegeneration, especially AD.
Collapse
Affiliation(s)
- Inayat Ur Rehman
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands.
| | - Hyun Young Park
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands; Department of Pediatrics, Maastricht University Medical Center (MUMC+), Maastricht 6202 AZ, the Netherlands.
| | - Tae Ju Park
- Haemato-oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, United Kingdom.
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Alz-Dementia Korea Co., Jinju 52828, Republic of Korea.
| |
Collapse
|
5
|
Gandhi S, Sweeney G, Perry CGR. Recent Advances in Pre-Clinical Development of Adiponectin Receptor Agonist Therapies for Duchenne Muscular Dystrophy. Biomedicines 2024; 12:1407. [PMID: 39061981 PMCID: PMC11274162 DOI: 10.3390/biomedicines12071407] [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: 04/30/2024] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by genetic mutations in the cytoskeletal-sarcolemmal anchor protein dystrophin. Repeated cycles of sarcolemmal tearing and repair lead to a variety of secondary cellular and physiological stressors that are thought to contribute to weakness, atrophy, and fibrosis. Collectively, these stressors can contribute to a pro-inflammatory milieu in locomotor, cardiac, and respiratory muscles. Given the many unwanted side effects that accompany current anti-inflammatory steroid-based approaches for treating DMD (e.g., glucocorticoids), there is a need to develop new therapies that address inflammation and other cellular dysfunctions. Adiponectin receptor (AdipoR) agonists, which stimulate AdipoR1 and R2 isoforms on various cell types, have emerged as therapeutic candidates for DMD due to their anti-inflammatory, anti-fibrotic, and pro-myogenic properties in pre-clinical human and rodent DMD models. Although these molecules represent a new direction for therapeutic intervention, the mechanisms through which they elicit their beneficial effects are not yet fully understood, and DMD-specific data is limited. The overarching goal of this review is to investigate how adiponectin signaling may ameliorate pathology associated with dystrophin deficiency through inflammatory-dependent and -independent mechanisms and to determine if current data supports their future progression to clinical trials.
Collapse
Affiliation(s)
- Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
| | - Gary Sweeney
- Department of Biology and Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
| | - Christopher G. R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
| |
Collapse
|
6
|
Xie L, Wang H, Hu J, Liu Z, Hu F. The role of novel adipokines and adipose-derived extracellular vesicles (ADEVs): Connections and interactions in liver diseases. Biochem Pharmacol 2024; 222:116104. [PMID: 38428826 DOI: 10.1016/j.bcp.2024.116104] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/01/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
Adipose tissues (AT) are an important endocrine organ that secretes various functional adipokines, peptides, non-coding RNAs, and acts on AT themselves or other distant tissues or organs through autocrine, paracrine, or endocrine manners. An accumulating body of evidence has suggested that many adipokines play an important role in liver metabolism. Besides the traditional adipokines such as adiponectin and leptin, many novel adipokines have recently been identified to have regulatory effects on the liver. Additionally, AT can produce extracellular vesicles (EVs) that act on peripheral tissues. However, under pathological conditions, such as obesity and diabetes, dysregulation of adipokines is associated with functional changes in AT, which may cause liver diseases. In this review, we focus on the newly discovered adipokines and EVs secreted by AT and highlight their actions on the liver under the context of obesity, nonalcoholic fatty liver diseases (NAFLD), and some other liver diseases. Clarifying the action of adipokines and adipose tissue-derived EVs on the liver would help to identify novel therapeutic targets or biomarkers for metabolic diseases.
Collapse
Affiliation(s)
- Lijun Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Huiying Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jinying Hu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Zhuoying Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Health Law Research Center, School of Law, Central South University, Changsha, China.
| | - Fang Hu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
7
|
Domingo E, Marques P, Francisco V, Piqueras L, Sanz MJ. Targeting systemic inflammation in metabolic disorders. A therapeutic candidate for the prevention of cardiovascular diseases? Pharmacol Res 2024; 200:107058. [PMID: 38218355 DOI: 10.1016/j.phrs.2024.107058] [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: 07/26/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death and disability worldwide. While many factors can contribute to CVD, atherosclerosis is the cardinal underlying pathology, and its development is associated with several metabolic risk factors including dyslipidemia and obesity. Recent studies have definitively demonstrated a link between low-grade systemic inflammation and two relevant metabolic abnormalities: hypercholesterolemia and obesity. Interestingly, both metabolic disorders are also associated with endothelial dysfunction/activation, a proinflammatory and prothrombotic phenotype of the endothelium that involves leukocyte infiltration into the arterial wall, one of the earliest stages of atherogenesis. This article reviews the current literature on the intricate relationship between hypercholesterolemia and obesity and the associated systemic inflammation and endothelial dysfunction, and discusses the effectiveness of present, emerging and in-development pharmacological therapies used to treat these metabolic disorders with a focus on their effects on the associated systemic inflammatory state and cardiovascular risk.
Collapse
Affiliation(s)
- Elena Domingo
- Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; Department of Pharmacology, Faculty of Medicine and Odontology, University of Valencia, Valencia, Spain
| | - Patrice Marques
- Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; Department of Pharmacology, Faculty of Medicine and Odontology, University of Valencia, Valencia, Spain
| | - Vera Francisco
- Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain
| | - Laura Piqueras
- Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; Department of Pharmacology, Faculty of Medicine and Odontology, University of Valencia, Valencia, Spain; CIBERDEM, Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute (ISCIII), Spain.
| | - Maria-Jesus Sanz
- Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; Department of Pharmacology, Faculty of Medicine and Odontology, University of Valencia, Valencia, Spain; CIBERDEM, Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute (ISCIII), Spain.
| |
Collapse
|
8
|
Rasoul D, Ajay A, Abdullah A, Mathew J, Lee Wei En B, Mashida K, Sankaranarayanan R. Alcohol and Heart Failure. Eur Cardiol 2023; 18:e65. [PMID: 38213665 PMCID: PMC10782426 DOI: 10.15420/ecr.2023.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/10/2023] [Indexed: 01/13/2024] Open
Abstract
Alcohol is the most frequently consumed toxic substance in the world and remains a major global public health issue, with one in three adults consuming it worldwide. Alcohol use is a leading risk factor for disease, contributing to over 60 acute and chronic health conditions, with a particularly complex association with cardiovascular disease. Chronic excessive alcohol consumption is associated with a range of cardiac complications, including decreased myocardial contractility, hypertension, arrhythmias, MI and heart failure. However, low-level alcohol consumption is believed to have a protective effect against ischaemic heart disease and diabetes. In most cohort studies, small to moderate amounts of alcohol consumption have not been linked to heart failure, indicating a threshold effect of alcohol with individual (possibly genetic) predisposition rather than a continuous effect of exposure. This review article explores the potential benefits of alcohol on the heart, the association between alcohol use and alcoholic cardiomyopathy and the epidemiology, clinical correlates and management of alcoholic cardiomyopathy.
Collapse
Affiliation(s)
- Debar Rasoul
- Cardiology Department, Liverpool University Hospitals NHS Foundation TrustLiverpool, UK
- Liverpool Centre for Cardiovascular Science, University of LiverpoolLiverpool, UK
| | - Ashwin Ajay
- Cardiology Department, Liverpool University Hospitals NHS Foundation TrustLiverpool, UK
| | - Alend Abdullah
- Cardiology Department, The Dudley Group NHS Foundation TrustDudley, UK
| | - Jean Mathew
- Cardiology Department, Liverpool University Hospitals NHS Foundation TrustLiverpool, UK
| | - Benjamin Lee Wei En
- Cardiology Department, Liverpool University Hospitals NHS Foundation TrustLiverpool, UK
| | | | - Rajiv Sankaranarayanan
- Cardiology Department, Liverpool University Hospitals NHS Foundation TrustLiverpool, UK
- Liverpool Centre for Cardiovascular Science, University of LiverpoolLiverpool, UK
| |
Collapse
|
9
|
Du Y, Duan Y, Zhao J, Liu C, Zhang Z, Zhang J, Meng Z, Wang X, Lau WB, Xie D, Lopez BL, Christopher TA, Gao E, Koch WW, Liu H, Liu D, Ma XL, Gu G, Wang Y. Dysfunctional APPL1-Mediated Epigenetic Regulation in Diabetic Vascular Injury. Arterioscler Thromb Vasc Biol 2023; 43:e491-e508. [PMID: 37795615 DOI: 10.1161/atvbaha.122.318752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 09/21/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND APN (adiponectin) and APPL1 (adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1) are potent vasculoprotective molecules, and their deficiency (eg, hypoadiponectinemia) contributes to diabetic vascular complications. However, the molecular mechanisms that govern their vasculoprotective genes as well as their alteration by diabetes remain unknown. METHODS Diabetic medium-cultured rat aortic endothelial cells, mouse aortic endothelial cells from high-fat-diet animals, and diabetic human aortic endothelial cells were used for molecular/cellular investigations. The in vivo concept-prove demonstration was conducted using diabetic vascular injury and diabetic hindlimb ischemia models. RESULTS In vivo animal experiments showed that APN replenishment caused APPL1 nuclear translocation, resulting in an interaction with HDAC (histone deacetylase) 2, which inhibited HDAC2 activity and increased H3Kac27 levels. Based on transcriptionome pathway-specific real-time polymerase chain reaction profiling and bioinformatics analysis, Angpt1 (angiopoietin 1), Ocln (occludin), and Cav1 (caveolin 1) were found to be the top 3 vasculoprotective genes suppressed by diabetes and rescued by APN in an APPL1-dependent manner. APN reverses diabetes-induced inhibition of Cav1 interaction with APPL1. APN-induced Cav1 expression was not affected by Angpt1 or Ocln deficiency, whereas APN-induced APPL1 nuclear translocation or upregulation of Angpt1/Ocln expression was abolished in the absence of Cav1 both in vivo and in vitro, suggesting Cav1 is upstream molecule of Angpt1/Ocln in response to APN administration. Chromatin immunoprecipitation-qPCR (quantitative polymerase chain reaction) demonstrated that APN caused significant enrichment of H3K27ac in Angpt1 and Ocln promoter region, an effect blocked by APPL1/Cav1 knockdown or HDAC2 overexpression. The protective effects of APN on the vascular system were attenuated by overexpression of HDAC2 and abolished by knocking out APPL1 or Cav1. The double knockdown of ANGPT1/OCLN blunted APN vascular protection both in vitro and in vivo. Furthermore, in diabetic human endothelial cells, HDAC2 activity is increased, H3 acetylation is decreased, and ANGPT1/OCLN expression is reduced, suggesting that the findings have important translational implications. CONCLUSIONS Hypoadiponectinemia and dysregulation of APPL1-mediated epigenetic regulation are novel mechanisms leading to diabetes-induced suppression of vasculoprotective gene expression. Diabetes-induced pathological vascular remodeling may be prevented by interventions promoting APPL1 nuclear translocation and inhibiting HDAC2.
Collapse
Affiliation(s)
- Yunhui Du
- Beijing Key Laboratory of Upper Airway Dysfunction-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases. Beijing Anzhen Hospital (Y. Du, Y. Duan), Capital Medical University, China
| | - Yanru Duan
- Beijing Key Laboratory of Upper Airway Dysfunction-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases. Beijing Anzhen Hospital (Y. Du, Y. Duan), Capital Medical University, China
| | - Jianli Zhao
- Department of Biomedical Engineering, UAB, Birmingham (J.Z., Y.W.)
| | - Caihong Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Zhen Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - John Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Zhijun Meng
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Xiaoliang Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Bernard L Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Theodore A Christopher
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA (E.G., W.W.K.)
| | - Walter W Koch
- Center for Translational Medicine, Temple University, Philadelphia, PA (E.G., W.W.K.)
| | - Huirong Liu
- Department of Physiology and Pathophysiology (H.L.), Capital Medical University, China
| | - Demin Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (D.L., G.G.)
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| | - Guoqiang Gu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (D.L., G.G.)
| | - Yajing Wang
- Department of Biomedical Engineering, UAB, Birmingham (J.Z., Y.W.)
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (C.L., Z.Z., J.Z., Z.M., X.W., W.B.L., D.X., B.L.L., T.A.C., X.-L.M., Y.W.)
| |
Collapse
|
10
|
Yang NI, Kuo LT, Lee CC, Ting MK, Wu IW, Chen SW, Hsu KH. Associations of Three-Dimensional Anthropometric Body Surface Scanning Measurements and Coronary Artery Disease. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:570. [PMID: 36984571 PMCID: PMC10056801 DOI: 10.3390/medicina59030570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Background and Objectives: The relationship between three-dimensional (3D) scanning-derived body surface measurements and biomarkers in patients with coronary artery disease (CAD) were assessed. Methods and Methods: The recruitment of 98 patients with CAD confirmed by cardiac catheterization and 98 non-CAD patients were performed between March 2016 and December 2017. A health questionnaire on basic information, life style variables, and past medical and family history was completed. 3D body surface measurements and biomarkers were obtained. Differences between the two groups were assessed and multivariable analysis performed. Results: It was found that chest width (odds ratio [OR] 0.761, 95% confidence interval [CI] = 0.586-0.987, p = 0.0399), right arm length (OR 0.743, 95% CI = 0.632-0.875, p = 0.0004), waist circumference (OR 1.119, 95% CI = 1.035-1.21, p = 0.0048), leptin (OR 1.443, 95% CI = 1.184-1.76, p = 0.0003), adiponectin (OR 0.978, 95% CI = 0.963-0.994, p = 0.006), and interleukin 6 (OR 1.181, 95% CI = 1.021-1.366, p = 0.0254) were significantly associated with CAD. The combination of biomarker scores and body measurement scores had the greatest area under the curve and best association with CAD (area under the curve of 0.8049 and 95% CI = 0.7440-0.8657). Conclusions: Our study suggests that 3D derived body surface measurements in combination with leptin, adiponectin, and interleukin 6 levels may direct us to those at risk of CAD, allowing a non-invasive approach to identifying high-risk patients.
Collapse
Affiliation(s)
- Ning-I Yang
- Division of Cardiology, Department of Internal Medicine, Chang Gung University College of Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Li-Tang Kuo
- Division of Cardiology, Department of Internal Medicine, Chang Gung University College of Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Chin-Chan Lee
- Division of Nephrology, Department of Internal Medicine, Chang Gung University College of Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Ming-Kuo Ting
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung University College of Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - I-Wen Wu
- Division of Nephrology, Department of Internal Medicine, Chang Gung University College of Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Shuo-Wei Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung University College of Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Kuang-Hung Hsu
- Laboratory for Epidemiology, Department of Health Care Management, Chang Gung University, Taoyuan 333, Taiwan
- Department of Health Care Management, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Safety, Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City 243, Taiwan
| |
Collapse
|
11
|
Gasbarrino K, Hafiane A, Gianopoulos I, Zheng H, Mantzoros CS, Daskalopoulou SS. Relationship between circulating adipokines and cholesterol efflux in subjects with severe carotid atherosclerosis. Metabolism 2023; 140:155381. [PMID: 36566801 DOI: 10.1016/j.metabol.2022.155381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/02/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
AIMS Cholesterol efflux capacity (CEC) as a measure of high-density lipoprotein functionality is independently and inversely associated with increased risk of cardiovascular events and mortality, and advanced plaque morphology. Adipokines, adipose tissue-derived factors, can influence systemic lipoprotein metabolism, and participate in the regulation of vascular function and inflammation. We aimed to investigate the association between CEC and circulating adipokine levels (anti-inflammatory adiponectin, and pro-inflammatory chemerin and resistin) in subjects with severe carotid atherosclerotic disease and evaluate its impact on post-surgical outcomes. METHODS AND RESULTS This is a cross-sectional study with a 5-year follow-up component. Consecutive patients with severe carotid atherosclerosis scheduled for a carotid endarterectomy were recruited from hospital-based centres in Montreal, Canada (n = 285). Fasting blood samples were collected pre-operatively and used to measure plasma total and high-molecular weight (HMW) adiponectin, chemerin, and resistin, and to perform cholesterol efflux assays in J774 macrophage-like cells. Five-year post-surgery outcomes were obtained through medical chart review. Subjects had a mean age of 70.1 ± 9.4, were 67.0 % male, had various comorbidities (hypercholesterolemia [85.3 %], hypertension [83.5 %], type 2 diabetes [34.5 %], coronary artery disease [38.6 %]), and previously experienced cerebrovascular symptomatology (77.9 %). CEC was independently and positively associated with total and HMW adiponectin levels (ß [95 % confidence interval]; 0.216 [0.134-0.298] and 0.107 [0.037-0.176], respectively) but not with chemerin or resistin. Total adiponectin had the greatest association accounting for 8.3 % of the variance in CEC. Interaction regression models demonstrated a significant interaction between adiponectin and chemerin in increasing CEC. Notably, with each unit increase in CEC there was a 93.9 % decrease in the odds of having an ischemic cerebrovascular event 5 years post-surgery (0.061 [0.007-0.561]). CONCLUSIONS Our findings demonstrated circulating adiponectin to have a strong association with increased CEC in subjects with severe carotid atherosclerosis and high CEC to be associated with more favourable post-surgical outcomes. These findings reflect the importance of adipose tissue health in influencing CEC levels and atherosclerotic cardiovascular disease risk.
Collapse
Affiliation(s)
- Karina Gasbarrino
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada
| | - Anouar Hafiane
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada
| | - Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada
| | - Huaien Zheng
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA, United States
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada; Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, McGill University Montreal, Canada.
| |
Collapse
|
12
|
Tseng SY, Chang HY, Li YH, Chao TH. Effects of Cilostazol on Angiogenesis in Diabetes through Adiponectin/Adiponectin Receptors/Sirtuin1 Signaling Pathway. Int J Mol Sci 2022; 23:14839. [PMID: 36499166 PMCID: PMC9739574 DOI: 10.3390/ijms232314839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Cilostazol is an antiplatelet agent with vasodilating effects that functions by increasing the intracellular concentration of cyclic adenosine monophosphate. We have previously shown that cilostazol has favorable effects on angiogenesis. However, there is no study to evaluate the effects of cilostazol on adiponectin. We investigated the effects of cilostazol on angiogenesis in diabetes in vitro and in vivo through adiponectin/adiponectin receptors (adipoRs) and the sirtuin 1 (SIRT1)/AMP-activated protein kinase (AMPK) signaling pathway. Human umbilical vein endothelial cells (HUVECs) and human aortic smooth muscle cells (HASMCs) were cocultured under high glucose (HG) conditions. Adiponectin concentrations in the supernatants were significantly increased when HASMCs were treated with cilostazol but not significantly changed when only HUVECs were treated with cilostazol. Cilostazol treatment enhanced the expression of SIRT1 and upregulated the phosphorylation of AMPK in HG-treated HUVECs. By sequential knockdown of adipoRs, SIRT1, and AMPK, our data demonstrated that cilostazol prevented apoptosis and stimulated proliferation, chemotactic motility, and capillary-like tube formation in HG-treated HUVECs through the adipoRs/SIRT1/AMPK signaling pathway. The phosphorylation of downstream signaling molecules, including acetyl-CoA carboxylase (ACC) and endothelial nitric oxide synthase (eNOS), was downregulated when HUVECs were treated with a SIRT1 inhibitor. In streptozotocin-induced diabetic mice, cilostazol treatment could improve blood flow recovery 21-28 days after inducing hindlimb ischemia as well as increase the circulating of CD34+CD45dim cells 14-21 days after operation; moreover, these effects were significantly attenuated by the knockdown of adipoR1 but not adipoR2. The expression of SIRT1 and phosphorylation of AMPK/ACC and Akt/eNOS in ischemic muscles were significantly attenuated by the gene knockdown of adipoRs. Cilostazol improves HG-induced endothelial dysfunction in vascular endothelial cells and enhances angiogenesis in diabetic mice by upregulating the expression of adiponectin/adipoRs and its SIRT1/AMPK downstream signaling pathway.
Collapse
Affiliation(s)
- Shih-Ya Tseng
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Hsien-Yuan Chang
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Yi-Heng Li
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Ting-Hsing Chao
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Health Management Center, National Cheng Kung University Hospital, Tainan 704, Taiwan
| |
Collapse
|
13
|
Causal associations of circulating adiponectin with cardiometabolic diseases and osteoporotic fracture. Sci Rep 2022; 12:6689. [PMID: 35461346 PMCID: PMC9035157 DOI: 10.1038/s41598-022-10586-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 04/07/2022] [Indexed: 02/05/2023] Open
Abstract
Circulating adiponectin shows some relationships with the occurrence of cardiometabolic diseases and osteoporotic fracture, but little is known about their causal associations. This two-sample Mendelian randomization (MR) study aims to explore the causal roles of circulating adiponectin in cardiometabolic diseases and osteoporotic fracture. We used 15 single nucleotide polymorphisms associated with circulating adiponectin as the instrumental variables. Inverse variance weighted, weighted median and MR-Egger regression methods were applied to study the causal associations. The results found that high circulating adiponectin was causally associated with reduced risk of type 2 diabetes (beta-estimate: -0.030, 95% CI: -0.048 to -0.011, SE: 0.009, P-value = 0.002) and may be the risk factor of coronary artery disease (beta-estimate: 0.012, 95% CI: 0.001 to 0.023, SE: 0.006, P-value = 0.030). No causal associations were seen between circulating adiponectin and other outcomes including heart failure, atrial fibrillation, cerebral ischemia, intracerebral hemorrhage or osteoporotic fracture. This study found the potential causal roles of high circulating adiponectin in reduced risk of type 2 diabetes and increased risk of coronary artery disease, which may help prevent and treat these two diseases.
Collapse
|
14
|
Salagre D, Chayah M, Molina-Carballo A, Oliveras-López MJ, Munoz-Hoyos A, Navarro-Alarcón M, Fernández-Vázquez G, Agil A. Melatonin induces fat browning by transdifferentiation of white adipocytes and de novo differentiation of mesenchymal stem cells. Food Funct 2022; 13:3760-3775. [PMID: 35274657 DOI: 10.1039/d1fo04360a] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The role of melatonin in obesity control is extensively accepted, but its mechanism of action is still unclear. Previously we demonstrated that chronic oral melatonin acts as a brown-fat inducer, driving subcutaneous white adipose tissue (sWAT) into a brown-fat-like function (beige) in obese diabetic rats. However, immunofluorescence characterization of beige depots in sWAT and whether melatonin is a beige-fat inducer by de novo differentiation and/or transdifferentiation of white adipocytes are still undefined. Lean (ZL) and diabetic fatty (ZDF) Zücker rats were subdivided into two groups, control (C) and oral melatonin-supplemented (M, 10 mg kg-1 day-1) for 6 weeks. Mesenchymal stem cells (MSCs) were isolated from both rat inguinal fat and human lipoaspirates followed by adipogenesis assays with or without melatonin (50 nM for 12 h in a 24 h period, 12 h+/12 h-) mimicking the light/dark cycle. Immunofluorescence and western-blot assays showed the partial transdifferentiation of white adipocytes in both ZL and ZDF rats, with increasing thermogenic and beige markers, UCP1 and CITED1 and decreasing white adipocyte marker ASC-1 expression. In addition, melatonin increased UCP1, CITED1, and PGC1-α expression in differentiated adipocytes in both rats and humans. These results demonstrate that melatonin increases brown fat in obese diabetic rats by both adipocyte transdifferentiation and de novo differentiation. Furthermore, it promotes beige MSC adipogenesis in humans. This may contribute to the control of body weight attributed to melatonin and its metabolic benefits in human diabesity.
Collapse
Affiliation(s)
- Diego Salagre
- Department of Pharmacology and Neurosciences Institute, School of Medicine & Biomedical Research Center, University of Granada, 18016 Granada, Spain.
| | - Meriem Chayah
- Department of Pharmacology and Neurosciences Institute, School of Medicine & Biomedical Research Center, University of Granada, 18016 Granada, Spain.
| | - Antonio Molina-Carballo
- Department of Pediatrics, School of Medicine, University of Granada (Spain). Unit of Pediatric Neurology and Neurodevelopment, Clínico San Cecilio University Hospital, the Andalusian Health Service, Granada, Spain.
| | - María-Jesús Oliveras-López
- Department of Molecular Biology and Biochemical Engineering, University Pablo de Olavide, 41013, Seville, Spain
| | - Antonio Munoz-Hoyos
- Department of Pediatrics, School of Medicine, University of Granada (Spain). Unit of Pediatric Neurology and Neurodevelopment, Clínico San Cecilio University Hospital, the Andalusian Health Service, Granada, Spain.
| | - Miguel Navarro-Alarcón
- Department of Nutrition and Bromatology, School of Pharmacy, University of Granada, 18071 Granada, Spain
| | | | - Ahmad Agil
- Department of Pharmacology and Neurosciences Institute, School of Medicine & Biomedical Research Center, University of Granada, 18016 Granada, Spain.
| |
Collapse
|
15
|
Okocha CE, Manafa PO, Igwe CN, Okite UP, Onah CE, Efobi C. Adiponectin and Disease Severity in Sickle Cell Anemia Patients Attending a Tertiary Health Institution in Nnewi, Southeast Nigeria. Front Genet 2022; 13:799425. [PMID: 35281808 PMCID: PMC8905498 DOI: 10.3389/fgene.2022.799425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Hemoglobin polymerization in sickle cell anemia (SCA) leads to abnormally rigid and adhesive erythrocytes that obstruct blood vessels, leading to poor tissue perfusion, hence provoking inflammation and damage of surrounding tissues. Adiponectin, a protein hormone, presumptively has anti-inflammatory characteristics, hence may be an important therapeutic target in SCA. Aim: The aim of the study was to evaluate the status of adiponectin and its correlation with disease severity in SCA. Patients and Methods: A total of 84 subjects were recruited for the study comprising 34 homozygous sickle cell (HbSS) subjects (25 in the steady state and nine in the resolving crisis state) and 50 controls (25 heterozygous sickle cell [HbAS] and 25 hemoglobin phenotype AA subjects). The hemoglobin phenotype, adiponectin levels, and full blood counts were evaluated. Anthropometric measurements were also conducted. Results: A significant difference was observed in the mean body mass index between the different hemoglobin phenotype groups and also between the SCA in crisis resolution patients and the control group (p < 0.05). There was no significant difference in the median serum levels of adiponectin in the different hemoglobin phenotype groups and between SCA patients in the steady state compared with those in the crisis resolution state. Also, there was no correlation between disease severity and adiponectin in SCA patients in the steady state (p = 0.87). Conclusion: Our study seems to suggest that in our data set of sickle cell anemia patients in the steady state, adiponectin does not constitute part of the endocrinopathy that affects these patients.
Collapse
Affiliation(s)
| | - Patrick O Manafa
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, Nnamdi Azikiwe University, Nnewi, Nigeria
| | - Chioma Nkechinyere Igwe
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, Nnamdi Azikiwe University, Nnewi, Nigeria
| | | | - Christian Ejike Onah
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, Nnamdi Azikiwe University, Nnewi, Nigeria
| | - Chilota Efobi
- Haematology Department, Nnamdi Azikiwe University Teaching Hospital, Nnewi, Nigeria
| |
Collapse
|
16
|
Lizcano F. Roles of estrogens, estrogen-like compounds, and endocrine disruptors in adipocytes. Front Endocrinol (Lausanne) 2022; 13:921504. [PMID: 36213285 PMCID: PMC9533025 DOI: 10.3389/fendo.2022.921504] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Women are subject to constitutional changes after menopause, which increases conditions and diseases prone to cardiovascular risks such as obesity and diabetes mellitus. Both estrogens and androgens influence the individual's metabolic mechanism, which controls the fat distribution and the hypothalamic organization of the regulatory centers of hunger and satiety. While androgens tend to accumulate fat in the splanchnic and the visceral region with an increase in cardiovascular risk, estrogens generate more subcutaneous and extremity distribution of adipose tissue. The absence of estrogen during menopause seems to be the main factor that gives rise to the greater predisposition of women to suffer cardiovascular alterations. However, the mechanisms by which estrogens regulate the energy condition of people are not recognized. Estrogens have several mechanisms of action, which mainly include the modification of specific receptors that belong to the steroid receptor superfamily. The alpha estrogen receptors (ERα) and the beta receptors (ERβ) have a fundamental role in the metabolic control of the individual, with a very characteristic corporal distribution that exerts an influence on the metabolism of lipids and glucose. Despite the significant amount of knowledge in this field, many of the regulatory mechanisms exerted by estrogens and ER continue to be clarified. This review will discuss the role of estrogens and their receptors on the central regulation of caloric expenditure and the influence they exert on the differentiation and function of adipocytes. Furthermore, chemical substances with a hormonal activity that cause endocrine disruption with affectation on estrogen receptors will be considered. Finally, the different medical therapies for the vasomotor manifestations of menopause and their role in reducing obesity, diabetes, and cardiovascular risk will be analyzed.
Collapse
|
17
|
El Atab O, Ghantous CM, El-Zein N, Farhat R, Agouni A, Korashy HM, Djouhri L, Kamareddine L, Zibara K, Zeidan A. Involvement of caveolae in hyperglycemia-induced changes in adiponectin and leptin expressions in vascular smooth muscle cells. Eur J Pharmacol 2021; 919:174701. [PMID: 34954233 DOI: 10.1016/j.ejphar.2021.174701] [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: 06/20/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 11/03/2022]
Abstract
Hyperglycemia exerts various harmful effects on the vasculature. Studies have shown an association between the levels of the adipokines leptin and adiponectin (APN) and vascular complications in diabetes mellitus. The aim of our study was to investigate the molecular mechanisms mediated by APN and leptin that are involved in hyperglycemia-induced vascular remodeling, especially at the level of oxidative stress and actin cytoskeleton dynamics. Rat aorta organ culture was used to investigate the effect of hyperglycemia on APN and leptin protein expression in vascular smooth muscle cells (VSMCs) using Western blot analysis and immunohistochemistry. Hyperglycemia lead to a significant increase in APN synthesis in VSMCs, mainly through caveolae, but this increase failed to provide vascular protection because of the decreased expression of APN receptors, especially AdipoR2, which was assessed by qPCR. In addition, hyperglycemia significantly upregulated leptin expression in VSMCs through caveolae and the RhoA/ROCK pathway. These variations lead to a marked increase in reactive oxygen species (ROS) production, detected by dihydroethidium (DHE) staining, and in NADPH oxidase type 4 (Nox4) expression. Moreover, Nox4 mediated the synthesis of APN in hyperglycemia in VSMCs. Finally, hyperglycemia activated the RhoA/ROCK pathway and subsequently induced the polymerization of globular actin (G-actin) into filamentous actin (F-actin), decreasing the G/F-actin ratio. Taken together, these data show that hyperglycemia increases oxidative stress and changes actin cytoskeleton dynamics in the aorta via caveolae, favoring vascular remodeling.
Collapse
Affiliation(s)
- Ola El Atab
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut, Beirut, Lebanon; PRASE and Department of Biology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Crystal M Ghantous
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut, Beirut, Lebanon; Department of Nursing and Health Sciences, Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Keserwan, Lebanon
| | - Nabil El-Zein
- PRASE and Department of Biology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Rima Farhat
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut, Beirut, Lebanon
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Qatar
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Qatar
| | - Laiche Djouhri
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Qatar; Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Layla Kamareddine
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Qatar; Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, P.O. Box 2713, Qatar
| | - Kazem Zibara
- PRASE and Department of Biology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Asad Zeidan
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Qatar; Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
18
|
Chondrogianni M, Lambadiari V, Katsanos AH, Stefanou MI, Palaiodimou L, Triantafyllou AS, Karagiannis G, Konstantakos V, Ioakeimidis M, Triantafyllou S, Zompola C, Liantinioti C, Pappa A, Rizos I, Voumvourakis K, Tsivgoulis G, Boutati E. Omentin Is Independently Associated with Stroke Severity and Ipsilateral Carotid Artery Stenosis in Patients with Acute Cerebral Ischemia. J Clin Med 2021; 10:jcm10245797. [PMID: 34945092 PMCID: PMC8703878 DOI: 10.3390/jcm10245797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/27/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022] Open
Abstract
Mounting evidence indicates an association between adipokines and inflammation-related atherosclerosis. Here, we sought to investigate the association of vaspin and omentin with clinical characteristics and outcomes of patients with acute cerebral ischemia (ACI). Consecutive ACI patients were evaluated within 24 h from symptom-onset. Stroke aetiology was classified using TOAST criteria. Adipokines were assayed using quantikine enzyme immunoassay commercially available kits. Stroke severity was assessed by NIHSS-score, and ipsilateral carotid stenosis (≥50% by NASCET criteria) by ultrasound and CT/MR angiography. Major cerebrovascular events were assessed at three months. We included 135 ACI patients (05 (78%) and 30 (22%) with acute ischemic stroke and transient ischemic attack, respectively; mean age ± SD: 59 ± 10 years; 68% men; median NIHSS-score: 3 (IQR:1–7)). Omentin was strongly correlated to admission stroke severity (Spearman rho coefficient: +0.303; p < 0.001). Patients with ipsilateral carotid stenosis had higher omentin levels compared to patients without stenosis (13.3 ± 8.9 ng/mL vs. 9.5 ± 5.5 ng/mL, p = 0.014). Increasing omentin levels were independently associated with higher stroke severity (linear regression coefficient = 0.290; 95%CI: 0.063–0.516; p = 0.002) and ipsilateral carotid stenosis (linear regression coefficient = 3.411; 95%CI: 0.194–6.628; p = 0.038). No association of vaspin with clinical characteristics and outcomes was found. Circulating omentin may represent a biomarker for the presence of atherosclerotic plaque, associated with higher stroke severity in ACI patients.
Collapse
Affiliation(s)
- Maria Chondrogianni
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Vaia Lambadiari
- Second Department of Internal Medicine, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (V.L.); (E.B.)
| | - Aristeidis H. Katsanos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
- Division of Neurology, McMaster University and Population Health Research Institute, Hamilton, ON L8S 3L8, Canada
| | - Maria Ioanna Stefanou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Lina Palaiodimou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Alexandros Stavros Triantafyllou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Georgios Karagiannis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Vasileios Konstantakos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Michael Ioakeimidis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Sokratis Triantafyllou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Christina Zompola
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Chryssa Liantinioti
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Alexandra Pappa
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Ioannis Rizos
- Second Department of Cardiology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece;
| | - Konstantinos Voumvourakis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (M.C.); (A.H.K.); (M.I.S.); (L.P.); (A.S.T.); (G.K.); (V.K.); (M.I.); (S.T.); (C.Z.); (C.L.); (A.P.); (K.V.)
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Correspondence: ; Tel.: +30-6937178635; Fax: +30-2105832471
| | - Eleni Boutati
- Second Department of Internal Medicine, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (V.L.); (E.B.)
| |
Collapse
|
19
|
de Alencar AKN, Wang H, de Oliveira GMM, Sun X, Zapata-Sudo G, Groban L. Crossroads between Estrogen Loss, Obesity, and Heart Failure with Preserved Ejection Fraction. Arq Bras Cardiol 2021; 117:1191-1201. [PMID: 34644788 PMCID: PMC8757160 DOI: 10.36660/abc.20200855] [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: 07/31/2020] [Revised: 12/16/2020] [Accepted: 01/27/2021] [Indexed: 11/24/2022] Open
Abstract
The prevalence of obesity and heart failure with preserved ejection fraction (HFpEF) increases significantly in postmenopausal women. Although obesity is a risk factor for left ventricular diastolic dysfunction (LVDD), the mechanisms that link the cessation of ovarian hormone production, and particularly estrogens, to the development of obesity, LVDD, and HFpEF in aging females are unclear. Clinical, and epidemiologic studies show that postmenopausal women with abdominal obesity (defined by waist circumference) are at greater risk for developing HFpEF than men or women without abdominal obesity. The study presents a review of clinical data that support a mechanistic link between estrogen loss plus obesity and left ventricular remodeling with LVDD. It also seeks to discuss potential cell and molecular mechanisms for estrogen-mediated protection against adverse adipocyte cell types, tissue depots, function, and metabolism that may contribute to LVDD and HFpEF.
Collapse
Affiliation(s)
| | - Hao Wang
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
- Wake Forest School of MedicineWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Internal Medicine-Section of Molecular Medicine, Winston-Salem, North Carolina - Estados Unidos da América
| | - Gláucia Maria Moraes de Oliveira
- Universidade Federal do Rio de JaneiroDepartamento de Clínica MédicaFaculdade de MedicinaRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Departamento de Clínica Médica, Faculdade de Medicina, Rio de Janeiro, RJ - Brasil
| | - Xuming Sun
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
| | - Gisele Zapata-Sudo
- Universidade Federal do Rio de JaneiroInstituto de Ciências BiomédicasRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Instituto de Ciências Biomédicas, Rio de Janeiro, RJ - Brasil
- Universidade Federal do Rio de JaneiroInstituto de Cardiologia Edson SaadFaculdade de MedicinaRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Instituto de Cardiologia Edson Saad, Faculdade de Medicina, Rio de Janeiro, RJ - Brasil
| | - Leanne Groban
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
- Wake Forest School of MedicineWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Internal Medicine-Section of Molecular Medicine, Winston-Salem, North Carolina - Estados Unidos da América
| |
Collapse
|
20
|
Antioxidant Potential of Adiponectin and Full PPAR- γ Agonist in Correcting Streptozotocin-Induced Vascular Abnormality in Spontaneously Hypertensive Rats. PPAR Res 2021; 2021:6661181. [PMID: 34691163 PMCID: PMC8531825 DOI: 10.1155/2021/6661181] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 07/15/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress, which is associated with metabolic and anthropometric perturbations, leads to reactive oxygen species production and decrease in plasma adiponectin concentration. We investigated pharmacodynamically the pathophysiological role and potential implication of exogenously administered adiponectin with full and partial peroxisome proliferator-activated receptor-gamma (PPAR-γ) agonists on modulation of oxidative stress, metabolic dysregulation, and antioxidant potential in streptozotocin-induced spontaneously hypertensive rats (SHR). Group I (WKY) serves as the normotensive control, whereas 42 male SHRs were randomized equally into 7 groups (n = 6); group II serves as the SHR control, group III serves as the SHR diabetic control, and groups IV, V, and VI are treated with irbesartan (30 mg/kg), pioglitazone (10 mg/kg), and adiponectin (2.5 μg/kg), whereas groups VII and VIII received cotreatments as irbesartan+adiponectin and pioglitazone+adiponectin, respectively. Diabetes was induced using an intraperitoneal injection of streptozotocin (40 mg/kg). Plasma adiponectin, lipid contents, and arterial stiffness with oxidative stress biomarkers were measured using an in vitro and in vivo analysis. Diabetic SHRs exhibited hyperglycemia, hypertriglyceridemia, hypercholesterolemia, and increased arterial stiffness with reduced plasma adiponectin and antioxidant enzymatic levels (P < 0.05). Diabetic SHRs pretreated with pioglitazone and adiponectin separately exerted improvements in antioxidant enzyme activities, abrogated arterial stiffness, and offset the increased production of reactive oxygen species and dyslipidemic effects of STZ, whereas the blood pressure values were significantly reduced in the irbesartan-treated groups (all P < 0.05). The combined treatment of exogenously administered adiponectin with full PPAR-γ agonist augmented the improvement in lipid contents and adiponectin concentration and restored arterial stiffness with antioxidant potential effects, indicating the degree of synergism between adiponectin and full PPAR-γ agonists (pioglitazone).
Collapse
|
21
|
T-Cadherin and the Ratio of Its Ligands as Predictors of Carotid Atherosclerosis: A Pilot Study. Biomedicines 2021; 9:biomedicines9101398. [PMID: 34680515 PMCID: PMC8533356 DOI: 10.3390/biomedicines9101398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/27/2022] Open
Abstract
In the cardiovascular system, atherogenic low-density lipoproteins (LDL) and the protective hormone adiponectin bind to the same receptor, T-cadherin. In this study, we tested the hypothesis that the ratio of circulating LDL to high-molecular weight (HMW) adiponectin could predict the development of atherosclerosis. Using enzyme-linked immunosorbent assay, we measured the level of circulating HMW adiponectin in the blood of donors together with ultrasound measuring of intima-media thickness (IMT) of carotid arteries. Single-nucleotide polymorphisms in the T-cadherin gene were identified using polymerase chain reaction. We found that carotid artery IMT is inversely correlated with the level of HMW in male subjects. We also found that the G allele of rs12444338 SNP in the T-cadherin gene correlates with a lower level of circulating T-cadherin and thinner IMT and therefore could be considered as an atheroprotective genotype. Despite our data, we could not provide direct evidence for the initial study hypothesis. However, we did uncover an important correlation between circulating T-cadherin and thinner carotid IMT.
Collapse
|
22
|
Afzal S, Sattar MA, Johns EJ, Eseyin OA. Peroxisome proliferator-activated receptor agonist (pioglitazone) with exogenous adiponectin ameliorates arterial stiffness and oxidative stress in diabetic Wistar Kyoto rats. Eur J Pharmacol 2021; 907:174218. [PMID: 34111396 DOI: 10.1016/j.ejphar.2021.174218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 11/17/2022]
Abstract
Oxidative stress causes hypoadiponectemia and reactive oxygen species production. This study investigates the pathophysiological role and potential effects of adiponectin with partial and full peroxisome proliferator-activated receptor-gamma agonists on modulation of metabolic dysregulation and oxidative stress in diabetic model of Wistar Kyoto rats (WKY). Forty two male WKY rats were randomized equally into 7 groups (n = 6), Group I serve as control, group II as WKY diabetic control, groups III, IV and V treated with irbesartan (30 mg/kg), pioglitazone (10 mg/kg) and adiponectin (2.5 μg/kg), groups VI and VII were co-treated as: irbesartan + adiponectin, pioglitazone + adiponectin, respectively. Streptozotocin @ 40 mg/kg was administered intraperitoneally to induce diabetes. Plasma adiponectin, metabolic indices, pulse wave velocity, oxidative stress and antioxidant enzymatic activities were measured. Streptozotocin induced WKYs expressed hyperglycaemia, hypertriglyceridemia, hypercholesterolemia, hypoadiponectemia, increased arterial stiffness and decreased antioxidant enzymatic levels (P<0.05). Treatment with adiponectin or pioglitazone alone showed improvements in metabolic indices, antioxidant enzymes, and abrogated arterial stiffness, attenuated generation of reactive oxygen species and dyslipidaemic effects of streptozotocin better as compared to irbesartan sets of treatment (all P<0.05). Co-treatment of adiponectin with pioglitazone significantly amplified the improvement in plasma triglycerides, adiponectin concentration, pulse wave velocity and antioxidant enzymatic activities indicating synergistic effects of adiponectin with full PPAR-γ agonist.
Collapse
Affiliation(s)
- Sheryar Afzal
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia; Faculty of Pharmacy, MAHSA University, Selangor, Malaysia.
| | | | | | - Olorunfemi A Eseyin
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia; Faculty of Pharmacy, University of Uyo, Uyo, Akwa Ibom State, Nigeria
| |
Collapse
|
23
|
Moosavian SP, Rahimlou M, Asbaghi O, Moradi S, Marx W, Paknahad Z. The effect of soy products on circulating adiponectin and leptin concentration in adults: A systematic review and meta-analysis of randomised controlled trials. Int J Clin Pract 2021; 75:e14100. [PMID: 33619795 DOI: 10.1111/ijcp.14100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/18/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Human clinical trials that have investigated the effect of soy product consumption on adipokines have reported inconsistent results. Our objective was to elucidate the role of soy product consumption on adiponectin and leptin in adults through a systematic review and meta-analysis of available randomised placebo-controlled trials (RCTs). METHODS The systematic search included PubMed, Scopus, Web of Science, EmBase, Google Scholar and Cochrane database from inception to July 2020. Human clinical trials that reported the effect of soy product consumption on leptin and adiponectin were included. The pooled weighted mean difference (WMD) was calculated by the random-effects model. Heterogeneity, sensitivity analysis, and publication bias were reported using standard methods. Quality assessment was performed using Cochrane risk of bias assessment tool. RESULTS Overall, 13 RCTs with 824 participants were included in this meta-analysis. Our analysis showed that soy product consumption did not significantly affect leptin (WMD: 0.01 ng/mL; 95% CI, -0.16, 0.18; P = .88) and adiponectin (WMD: -0.09 ng/mL; 95% CI, -0.29, 0.12; P = .39) concentration in comparison with control. Furthermore, subgroup analysis indicated that the effect remained non-significant when analysed by study design, participant demographics and intervention characteristics. Based on the Cochrane Collaboration Risk of Bias tool, seven studies were considered good quality and six studies were fair. CONCLUSION The present systematic review and meta-analysis suggest that soy product consumption had no significant effect on leptin and adiponectin levels in adults. However, future larger and well-designed trials are still needed to further explore this research area and to address the heterogeneous study design used in the existing literature.
Collapse
Affiliation(s)
- Seyedeh Parisa Moosavian
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehran Rahimlou
- Department of Nutrition, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Omid Asbaghi
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Sajjad Moradi
- Halal Research Center of IRI, FDA, Tehran, Iran
- Nutritional Sciences Department, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Wolfgang Marx
- iMPACT (the Institute for Mental and Physical Health and Clinical Translation), Food & Mood Centre, Deakin University, Geelong, VIC, Australia
| | - Zamzam Paknahad
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
24
|
Močnik M, Marčun Varda N. Cardiovascular Risk Factors in Children with Obesity, Preventive Diagnostics and Possible Interventions. Metabolites 2021; 11:551. [PMID: 34436493 PMCID: PMC8398426 DOI: 10.3390/metabo11080551] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/30/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022] Open
Abstract
The increasing burden of obesity plays an essential role in increased cardiovascular morbidity and mortality. The effects of obesity on the cardiovascular system have also been demonstrated in childhood, where prevention is even more important. Obesity is associated with hormonal changes and vascular dysfunction, which eventually lead to hypertension, hyperinsulinemia, chronic kidney disease, dyslipidemia and cardiac dysfunction-all associated with increased cardiovascular risk, leading to potential cardiovascular events in early adulthood. Several preventive strategies are being implemented to reduce the cardiovascular burden in children. This paper presents a comprehensive review of obesity-associated cardiovascular morbidity with the preventive diagnostic workup at our hospital and possible interventions in children.
Collapse
Affiliation(s)
- Mirjam Močnik
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
| | - Nataša Marčun Varda
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
- Medical Faculty, University of Maribor, Taborska 8, 2000 Maribor, Slovenia
| |
Collapse
|
25
|
Silva-Boghossian CM, Dezonne RS. What Are the Clinical and Systemic Results of Periodontitis Treatment in Obese Individuals? ACTA ACUST UNITED AC 2021; 8:48-65. [PMID: 34367878 PMCID: PMC8327900 DOI: 10.1007/s40496-021-00295-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/24/2022]
Abstract
Purpose of Review Periodontitis and obesity are characterized by a dysregulated inflammatory state. Obese individuals have a higher chance of presenting periodontitis. Clinical studies in different populations demonstrate that individuals with obesity have worse periodontal conditions. This current review aims to explore recent literature to understand what the impacts of obesity on periodontal treatment outcomes are and to learn whether periodontal treatment can improve systemic biomarkers in obese individuals. Recent Findings Short- and long-term evaluations demonstrated that non-surgical periodontal treatment could improve clinical parameters in obese individuals, represented as the reduction in mean probing depth, sites with probing depth ≥ 4 mm, and extension of bleeding on probing. However, obese individuals may have less clinical improvement when compared to normal-weight individuals with a similar periodontal profile. Additionally, periodontal treatment may contribute to a reduction in systemic levels of retinol-binding protein 4 and leptin, while promoting an increase in systemic levels of adiponectin. Summary Overall, obese individuals with periodontitis can significantly benefit from non-surgical periodontal treatment. However, clinical improvements seem to be less prominent in obese individuals with periodontitis compared to non-obese individuals with similar periodontal status. Nevertheless, periodontal treatment may impact significantly on the reduction of several biochemical biomarkers of obesity with or without weight reduction. Further investigations are needed to improve our comprehension of the mechanisms underlying those findings.
Collapse
Affiliation(s)
- Carina M. Silva-Boghossian
- Periodontics, School of Dentistry, Federal University of Rio de Janeiro, Rua Professor Rodolpho Paulo Rocco, 325, Cidade Universitaria, Rio de Janeiro, RJ CEP 21941-617 Brazil
| | - Romulo S. Dezonne
- Postgraduate Program in Translational Biomedicine, University of Grande Rio, Duque de Caxias, RJ Brazil
| |
Collapse
|
26
|
Lake JE, La K, Erlandson KM, Adrian S, Yenokyan G, Scherzinger A, Dubé MP, Stanley T, Grinspoon S, Falutz J, Mamputu JC, Marsolais C, McComsey GA, Brown TT. Tesamorelin improves fat quality independent of changes in fat quantity. AIDS 2021; 35:1395-1402. [PMID: 33756511 PMCID: PMC8243807 DOI: 10.1097/qad.0000000000002897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Fat quality and quantity may affect health similarly or differently. Fat quality can be assessed by measuring fat density on CT scan (greater density = smaller, higher quality adipocytes). We assessed the effects of tesamorelin, a growth hormone-releasing hormone analogue that reduces visceral fat (VAT) quantity in some people living with HIV (PWH), on fat density. DESIGN Participants from two completed, placebo-controlled, randomized trials of tesamorelin for central adiposity treatment in PWH were included if they had either a clinical response to tesamorelin (VAT decrease ≥8%, ≈70% of participants) or were placebo-treated. METHODS CT VAT and subcutaneous fat (SAT) density (Hounsfield Units, HU) were measured by a central blinded reader. RESULTS Participants (193 responders, 148 placebo) were 87% male and 83% white. Baseline characteristics were similar across arms, including VAT (-91 HU both arms, P = 0.80) and SAT density (-94 HU tesamorelin, -95 HU placebo, P = 0.29). Over 26 weeks, mean (SD) VAT and SAT density increased in tesamorelin-treated participants only [VAT: +6.2 (8.7) HU tesamorelin, +0.3 (4.2) HU placebo, P < 0.0001; SAT: +4.0 (8.7) HU tesamorelin, +0.3 (4.8) HU placebo, P < 0.0001]. The tesamorelin effects persisted after controlling for baseline VAT or SAT HU and area, and VAT [+2.3 HU, 95% confidence interval (4.5-7.3), P = 0.001) or SAT (+3.5 HU, 95% confidence interval (2.3-4.7), P < 0.001] area change. CONCLUSION In PWH with central adiposity who experienced VAT quantity reductions on tesamorelin, VAT and SAT density increased independent of changes in fat quantity, suggesting that tesamorelin also improves VAT and SAT quality in this group.
Collapse
Affiliation(s)
- Jordan E Lake
- University of Texas Health Science Center, Houston, Texas
| | - Kristen La
- University of Texas Health Science Center, Houston, Texas
| | | | | | | | | | - Michael P Dubé
- University of Southern California, Los Angeles, California
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Sousa AS, Sponton ACS, Delbin MA. Perivascular adipose tissue and microvascular endothelial dysfunction in obese mice: Beneficial effects of aerobic exercise in adiponectin receptor (AdipoR1) and peNOS Ser1177. Clin Exp Pharmacol Physiol 2021; 48:1430-1440. [PMID: 34260769 DOI: 10.1111/1440-1681.13550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 11/28/2022]
Abstract
In the present study, we aim to investigate the effects of aerobic physical training on perivascular adipose tissue (PVAT)-induced microvascular dysfunction of the femoral artery in obese mice. Microvascular reactivity was evaluated in control sedentary (c-SD), obese sedentary (o-SD) and obese trained (o-TR) male mice (C57BL6/JUnib), in the absence (PVAT-) or the presence (PVAT+) of femoral artery PVAT. We also analyzed protein expression, vascular nitric oxide (NO) production and reactive oxygen species (ROS) generation in PVAT. The blood glucose, triglycerides and total cholesterol levels were increased in the o-SD group, when compared with the c-SD group. The maximal responses and the potency to acetylcholine (ACh) were decreased in PVAT+ compared with PVAT- rings in the o-SD group, accompanied by a decrease in vascular protein expression of peNOSSer1177 , Cu/Zn-SOD, leptin receptor (Ob-R) and adiponectin receptor (AdipoR1). The protein expression of leptin increased and that of adiponectin decreased in PVAT. Additionally, vascular NO production was reduced and ROS generation was enhanced in PVAT in the o-SD group. Aerobic exercise training was effective for normalizing ACh relaxation response, vascular NO production and ROS generation in the o-TR group. It partially re-established the vascular protein expression of peNOSSer1177 and the PVAT leptin; normalized the vascular Cu/Zn-SOD and AdipoR1 protein expressions. In obese sedentary mice, the presence of PVAT is involved in the process of microvascular dysfunction of the femoral artery in a pathway associated with increased inflammation and ROS generation. The aerobic exercise training normalized the vascular response, the NO production and/or bioavailability and oxidative stress, with improved vascular expressions of Cu/Zn-SOD, peNOSser1177 , and AdipoR1.
Collapse
Affiliation(s)
- Andressa S Sousa
- Laboratory of Vascular Biology, Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Amanda C S Sponton
- Laboratory of Vascular Biology, Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Maria A Delbin
- Laboratory of Vascular Biology, Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
28
|
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: 1.5] [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
|
29
|
da Silva Rosa SC, Liu M, Sweeney G. Adiponectin Synthesis, Secretion and Extravasation from Circulation to Interstitial Space. Physiology (Bethesda) 2021; 36:134-149. [PMID: 33904786 PMCID: PMC8461789 DOI: 10.1152/physiol.00031.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adiponectin, an adipokine that circulates as multiple multimeric complexes at high levels in serum, has antidiabetic, anti-inflammatory, antiatherogenic, and cardioprotective properties. Understanding the mechanisms regulating adiponectin's physiological effects is likely to provide critical insight into the development of adiponectin-based therapeutics to treat various metabolic-related diseases. In this review, we summarize our current understanding on adiponectin action in its various target tissues and in cellular models. We also focus on recent advances in two particular regulatory aspects; namely, the regulation of adiponectin gene expression, multimerization, and secretion, as well as extravasation of circulating adiponectin to the interstitial space and its degradation. Finally, we discuss some potential therapeutic approaches using adiponectin as a target and the current challenges facing adiponectin-based therapeutic interventions.
Collapse
Affiliation(s)
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Jichitu A, Bungau S, Stanescu AMA, Vesa CM, Toma MM, Bustea C, Iurciuc S, Rus M, Bacalbasa N, Diaconu CC. Non-Alcoholic Fatty Liver Disease and Cardiovascular Comorbidities: Pathophysiological Links, Diagnosis, and Therapeutic Management. Diagnostics (Basel) 2021; 11:689. [PMID: 33921359 PMCID: PMC8069361 DOI: 10.3390/diagnostics11040689] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has a growing prevalence in recent years. Its association with cardiovascular disease has been intensively studied, and certain correlations have been identified. The connection between these two entities has lately aroused interest regarding therapeutic management. In order to find the best therapeutic options, a detailed understanding of the pathophysiology that links (NAFLD) to cardiovascular comorbidities is needed. This review focuses on the pathogenic mechanisms that are behind these two diseases and on the therapeutic management available at this time.
Collapse
Affiliation(s)
- Alexandra Jichitu
- Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania; (A.J.); (C.C.D.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - Ana Maria Alexandra Stanescu
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.M.V.); (C.B.)
| | - Mirela Marioara Toma
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - Cristiana Bustea
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.M.V.); (C.B.)
| | - Stela Iurciuc
- Department of Cardiology, Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Marius Rus
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Nicolae Bacalbasa
- Department 13, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Surgery, “Ion Cantacuzino” Clinical Hospital, 030167 Bucharest, Romania
| | - Camelia Cristina Diaconu
- Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania; (A.J.); (C.C.D.)
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
31
|
Sultan HK, El-Ayat WM, AbouGhalia AH, Lasheen NN, Moustafa AS. Study of long non-coding RNA and mitochondrial dysfunction in diabetic rats. Tissue Cell 2021; 71:101516. [PMID: 33744742 DOI: 10.1016/j.tice.2021.101516] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/02/2021] [Accepted: 02/17/2021] [Indexed: 12/29/2022]
Abstract
Diabetes mellitus (DM) is a worldwide health problem. The Micro- and macro-vascular complications are the major causes of morbidity and mortality of DM. Molecular regulation of mitochondrial fission/fusion cycles is being studied, but the results were not conclusive. The aim of this study is to investigate the possible functional role of lncRNA H19 and its relation to mitofusin-2 (Mfn-2) gene expression in diabetic rats with cardiac and renal complications. Streptozotocin-induced diabetic male, albino rats and a matched control group were investigated. Cardiac weights, blood pressure and ECG were recorded. Biochemical evaluation of cardiac and renal functions was performed. Molecular determination of lncRNA H19 and Mfn-2 gene expression and histological examination by light and electron microscopy for cardiac and renal tissues were performed. Diabetic rats showed a significant increase of left ventricle weight/whole body weight ratio, R wave voltage, and a significant decrease of blood pressure, heart rate, and P wave voltage. At the molecular level, lncRNA H19 and Mfn-2 mRNA showed altered expression with a statistically significant downregulation of Mfn-2 mRNA expression in renal tissues. In conclusion, the changes in lncRNA H19 and Mfn-2 mRNA expression may help better understanding of the pathogenesis of cardiac and renal dysfunctions associated with type 1 DM.
Collapse
Affiliation(s)
- Haytham K Sultan
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, P.O. Box 11381, Egypt.
| | - Wael M El-Ayat
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, P.O. Box 11381, Egypt
| | - Azza H AbouGhalia
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, P.O. Box 11381, Egypt
| | - Noha N Lasheen
- Medical Physiology Department, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, P.O. Box 11381, Egypt
| | - Amr S Moustafa
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, P.O. Box 11381, Egypt
| |
Collapse
|
32
|
Kowalska K, Dembczyński R, Gołąbek A, Olkowicz M, Olejnik A. ROS Modulating Effects of Lingonberry ( Vaccinium vitis-idaea L.) Polyphenols on Obese Adipocyte Hypertrophy and Vascular Endothelial Dysfunction. Nutrients 2021; 13:nu13030885. [PMID: 33803343 PMCID: PMC7999824 DOI: 10.3390/nu13030885] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/28/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress and dysregulated adipocytokine secretion accompanying hypertrophied adipose tissue induce chronic inflammation, which leads to vascular endothelial dysfunction. The present study investigated the ability of anthocyanin (ACN) and non-anthocyanin polyphenol (PP) fractions from lingonberry fruit to mitigate adipose tissue hypertrophy and endothelial dysfunction using 3T3-L1 adipocytes and human umbilical vein endothelial cells (HUVECs). This study showed that the PP fraction decreased intracellular ROS generation in hypertrophied adipocytes by enhancing antioxidant enzyme expression (SOD2) and inhibiting oxidant enzyme expression (NOX4, iNOS). Moreover, PP and ACN fractions reduced triglyceride content in adipocytes accompanied by downregulation of the expression of lipogenic genes such as aP2, FAS, and DAGT1. Treatment with both fractions modulated the mRNA expression and protein secretion of key adipokines in hypertrophied adipocytes. Expression and secretion of leptin and adiponectin were, respectively, down- and upregulated. Furthermore, PP and ACN fractions alleviated the inflammatory response in TNF-α-induced HUVECs by inhibiting the expression of pro-inflammatory genes (IL-6, IL-1β) and adhesion molecules (VCAM-1, ICAM-1, SELE). The obtained results suggest that consuming polyphenol-rich lingonberry fruit may help prevent and treat obesity and endothelial dysfunction due to their antioxidant and anti-inflammatory actions.
Collapse
Affiliation(s)
- Katarzyna Kowalska
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (K.K.); (R.D.); (A.G.)
| | - Radosław Dembczyński
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (K.K.); (R.D.); (A.G.)
| | - Agata Gołąbek
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (K.K.); (R.D.); (A.G.)
| | - Mariola Olkowicz
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, 14 Bobrzynskiego St., 30-348 Krakow, Poland;
| | - Anna Olejnik
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland; (K.K.); (R.D.); (A.G.)
- Correspondence:
| |
Collapse
|
33
|
Bai J, Ma Q, Lan Y, Chen Y, Ma S, Li J, Liu C, Fu Z, Lu X, Huang Y, Li Y. Mitochondrial tRNA Mutation and Regulation of the Adiponectin Pathway in Maternally Inherited Hypertension in Chinese Han. Front Cell Dev Biol 2021; 8:623450. [PMID: 33553162 PMCID: PMC7862570 DOI: 10.3389/fcell.2020.623450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/01/2020] [Indexed: 01/11/2023] Open
Abstract
Some essential hypertension (EH) patients show maternal inheritance, which is the mode of mitochondrial DNA inheritance. This study examines the mechanisms by which mitochondrial mutations cause EH characterized by maternal inheritance. The study enrolled 115 volunteers, who were divided into maternally inherited EH (group A, n = 17), non-maternally inherited EH (group B, n = 65), and normal control (group C, n = 33) groups. A mitochondrial tRNA (15910 C>T) gene mutation was significantly correlated with EH and may play an important role in the pathogenesis of maternally inherited EH. Examining two families carrying the mitochondrial tRNA 15910 C>T mutation, which disrupted base pairing and may affect the stability and function of mitochondrial tRNAThr, we find that the overall incidence of EH was 59.3% in the maternal family members and 90% in males, significantly higher than in the general population in China (23.2%), and that the EH began at a younger age in those carrying mitochondrial tRNA 15910 C>T. To reveal the mechanism through which mitochondrial tRNA 15910 C>T causes maternally inherited EH, we cultured human peripheral blood mononuclear cells from family A2 in vitro. We find that cells carrying mitochondrial tRNA 15910 C>T were more viable and proliferative, and the increased ATP production resulted in raised intracellular reactive oxygen species (ROS). Moreover, the mitochondrial dysfunction resulted in reduced APN levels, causing hypoadiponectinemia, which promoted cell proliferation, and produced more ROS. This vicious cycle promoted the occurrence of EH with maternally inherited mitochondrial tRNA 15910 C>T. The mitochondrial tRNA 15910 C>T mutation may induce hypertension by changing the APN, AdipoR1, PGC-1α, and ERRα signaling pathways to elevate blood pressure. We discover a new mitochondrial mutation (tRNA 15910 C>T) related to EH, reveal part of the mechanism by which mitochondrial mutations lead to the occurrence and development of maternally inherited EH, and discuss the role of APN in it.
Collapse
Affiliation(s)
- Jing Bai
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Qiang Ma
- Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yunfeng Lan
- Hainan LANBO Health Management Co. Ltd., Sanya, China
| | - Yating Chen
- Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Shanshan Ma
- Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jiaxin Li
- Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Chuanbin Liu
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zihao Fu
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xu Lu
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yun Huang
- Department of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Li
- Department of Cardiology, The Sixth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
34
|
Komici K, Dello Iacono A, De Luca A, Perrotta F, Bencivenga L, Rengo G, Rocca A, Guerra G. Adiponectin and Sarcopenia: A Systematic Review With Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:576619. [PMID: 33935962 PMCID: PMC8082154 DOI: 10.3389/fendo.2021.576619] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sarcopenia is a progressive loss of skeletal muscle mass whose pathophysiology has been proposed to possibly involve mechanisms of altered inflammatory status and endocrine function. Adiponectin has been shown to modulate inflammatory status and muscle metabolism. However, the possible association between adiponectin levels and sarcopenia is poorly understood. In order to fill this gap, in the present manuscript we aimed to summarize the current evidence with a systematic review and a meta-analysis of studies reporting serum adiponectin levels in patients with sarcopenia compared to non-sarcopenic controls. METHODS An electronic search through Medline/PubMed, Cochrane Library, and Science Direct was performed till March 1, 2020. From the included papers, meta-analysis of cross-sectional studies comparing serum levels of adiponectin between patients with sarcopenia and controls was performed. RESULTS Out of 1,370 initial studies, seven studies were meta-analyzed. Sarcopenic participants had significantly higher levels of adiponectin Hedges' g with 95% confidence interval (CI): 1.20 (0.19-2.22), p = 0.02 than controls. Subgroup analysis, performed in Asian population and focused on identification of the condition based on AWGS criteria, reported higher adiponectin levels in sarcopenic population (2.1 (0.17-4.03), p = 0.03 and I2 = 98.98%. Meta-regression analysis revealed female gender to significantly influence the results as demonstrated by beta = 0.14 (95% CI (0.010-0.280), p = 0.040). CONCLUSIONS Our meta-analysis found evidence that sarcopenia is associated with higher adiponectin levels. However, caution is warranted on the interpretation of these findings, and future longitudinal research is required to disentangle and better understand the topic.
Collapse
Affiliation(s)
- Klara Komici
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
- *Correspondence: Klara Komici,
| | - Antonio Dello Iacono
- School of Health and Life Sciences, University of the West of Scotland, Hamilton, United Kingdom
| | - Antonio De Luca
- Section of Human Anatomy, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Fabio Perrotta
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
- Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), Telese Terme, Italy
| | - Aldo Rocca
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| |
Collapse
|
35
|
Role of Adiponectin and Brain Derived Neurotrophic Factor in Metabolic Regulation Involved in Adiposity and Body Fat Browning. J Clin Med 2020; 10:jcm10010056. [PMID: 33375318 PMCID: PMC7794779 DOI: 10.3390/jcm10010056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/16/2022] Open
Abstract
Obesity, characterized by excessive fat mass, has been emerging as a major global epidemic and contributes to the increased risk of morbidity around the world. Thus, the necessity to find effective therapy and specific regulatory mechanisms is increasing for controlling obesity. Lately, many researchers have been interested in the linkage between obesity and adipokines/myokines, particularly adiponectin and brain-derived neurotrophic factor (BDNF). However, the role of adiponectin and BDNF in adiposity has not been clearly defined yet. We examined the association of adiposity with adiponectin and BDNF through human study (observational study) with Korean women and in vitro experiments. In the human study, we found a negative relationship between adiposity and circulating adiponectins but irregular patterns in the relationship between adiposity and circulating BDNFs. In the in vitro study using 3T3-L1 adipocytes, adiponectin treatment strongly promoted adipocyte differentiation and the fat browning process, whereas BDNF treatment attenuated adipocyte differentiation and the fat browning process in differentiated adipocytes. Our results demonstrate that adiponectin and BDNF play an important role in regulating fat mass and the expression of fat-browning markers in different ways, and also suggest that circulating adiponectin may be used as an important monitoring index for obesity status.
Collapse
|
36
|
Hafiane A, Daskalopoulou SS. Adiponectin's mechanisms in high-density lipoprotein biogenesis and cholesterol efflux. Metabolism 2020; 113:154393. [PMID: 33058851 DOI: 10.1016/j.metabol.2020.154393] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/18/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022]
Abstract
AIM Among adiponectin's beneficial properties is its ability to promote cellular cholesterol efflux, thereby generating high-density lipoprotein (HDL) particles. However, adiponectin's role in the regulation of macrophage lipid metabolism, a crucial process in atherogenesis, remains poorly investigated. The aim of this study was to characterize the adiponectin's role in HDL biogenesis. METHODS AND RESULTS We perform kinetics studies in baby hamster kidney (BHK) and Tamm-Horsfall protein 1 (THP-1) cell lines to elucidate adiponectin's role in HDL biogenesis. In cholesterol-enriched cells, specific molar doses of adiponectin stimulated cholesterol efflux with high efficiency to apoA-I. In the presence of adiponectin, BHK cells expressing ATP binding cassette transporter A1 (ABCA1) or ABCG1 generated lipidated particles having α electrophoretic mobility (α-HDL) and a molecular size of 7.5-20 nm. Interestingly, in THP-1 macrophages, cholesterol efflux was associated with more lipidated preβ1-HDL particles. Direct molecular interaction of adiponectin with apoA-I enhanced the affinity of apoA-I to free cholesterol and resulted in an increase in preβ1-HDL particles from plasma ex vivo. Adiponectin increased ABCA1 and ABCG1 protein expression and activated the formation of ABCA1-linked cholesterol oxidase sensitive plasma membrane domains. CONCLUSION Adiponectin upregulated ABCA1 and ABCG1 protein expression, reduced lipid accumulation, and efficiently promoted nascent HDL formation. These results highlight that these cellular processes are interconnected through adiponectin and ABCA1- and ABCG1-dependent. In this pathway, adiponectin increased the affinity of apoA-I to cholesterol and effectively accelerated cholesterol removal from the plasma membrane to HDL particles. Thus, by accelerating HDL biogenesis, adiponectin may have therapeutic potential for atherosclerotic cardiovascular disease prevention and management.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01.3370H, Montréal, Qc H4A 3J1, Canada.
| | - Stella S Daskalopoulou
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01.3370H, Montréal, Qc H4A 3J1, Canada; Department of Medicine, Division of Internal Medicine, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, EM1.2230, Montreal, Quebec H4A 3J1, Canada.
| |
Collapse
|
37
|
Aparecida Silveira E, Vaseghi G, de Carvalho Santos AS, Kliemann N, Masoudkabir F, Noll M, Mohammadifard N, Sarrafzadegan N, de Oliveira C. Visceral Obesity and Its Shared Role in Cancer and Cardiovascular Disease: A Scoping Review of the Pathophysiology and Pharmacological Treatments. Int J Mol Sci 2020; 21:E9042. [PMID: 33261185 PMCID: PMC7730690 DOI: 10.3390/ijms21239042] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
The association between obesity, cancer and cardiovascular disease (CVD) has been demonstrated in animal and epidemiological studies. However, the specific role of visceral obesity on cancer and CVD remains unclear. Visceral adipose tissue (VAT) is a complex and metabolically active tissue, that can produce different adipokines and hormones, responsible for endocrine-metabolic comorbidities. This review explores the potential mechanisms related to VAT that may also be involved in cancer and CVD. In addition, we discuss the shared pharmacological treatments which may reduce the risk of both diseases. This review highlights that chronic inflammation, molecular aspects, metabolic syndrome, secretion of hormones and adiponectin associated to VAT may have synergistic effects and should be further studied in relation to cancer and CVD. Reductions in abdominal and visceral adiposity improve insulin sensitivity, lipid profile and cytokines, which consequently reduce the risk of CVD and some cancers. Several medications have shown to reduce visceral and/or subcutaneous fat. Further research is needed to investigate the pathophysiological mechanisms by which visceral obesity may cause both cancer and CVD. The role of visceral fat in cancer and CVD is an important area to advance. Public health policies to increase public awareness about VAT's role and ways to manage or prevent it are needed.
Collapse
Affiliation(s)
- Erika Aparecida Silveira
- Department of Epidemiology & Public Health, Institute of Epidemiology & Health Care, University College London, London WC1E 6BT, UK;
- Postgraduate Program in Health Sciences, Faculty of Medicine, Federal University of Goiás, Goiânia 74690-900, Goiás, Brazil; (A.S.d.C.S.); (M.N.)
| | - Golnaz Vaseghi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Annelisa Silva de Carvalho Santos
- Postgraduate Program in Health Sciences, Faculty of Medicine, Federal University of Goiás, Goiânia 74690-900, Goiás, Brazil; (A.S.d.C.S.); (M.N.)
- United Faculty of Campinas, Goiânia 74525-020, Goiás, Brazil
| | - Nathalie Kliemann
- Nutritional Epidemiology Group, Nutrition and Metabolism Section, International Agency for Research on Cancer, World Health Organization, 69372 Lyon, France;
| | - Farzad Masoudkabir
- Cardiac Primary Prevention Research Center, Tehran Heart Center, Tehran University of Medical Sciences, Tehran 1416753955, Iran;
- Department of Cardiology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran 1411713138, Iran
| | - Matias Noll
- Postgraduate Program in Health Sciences, Faculty of Medicine, Federal University of Goiás, Goiânia 74690-900, Goiás, Brazil; (A.S.d.C.S.); (M.N.)
- Instituto Federal Goiano, Ceres 76300-000, Goiás, Brazil
| | - Noushin Mohammadifard
- Hypertension Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran
- School of Population and Public Health, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Cesar de Oliveira
- Department of Epidemiology & Public Health, Institute of Epidemiology & Health Care, University College London, London WC1E 6BT, UK;
| |
Collapse
|
38
|
Francischetti EA, Dezonne RS, Pereira CM, de Moraes Martins CJ, Celoria BMJ, de Oliveira PAC, de Abreu VG. Insights Into the Controversial Aspects of Adiponectin in Cardiometabolic Disorders. Horm Metab Res 2020; 52:695-707. [PMID: 32927496 DOI: 10.1055/a-1239-4349] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In 2016, the World Health Organization estimated that more than 1.9 billion adults were overweight or obese. This impressive number shows that weight excess is pandemic. Overweight and obesity are closely associated with a high risk of comorbidities, such as insulin resistance and its most important outcomes, including metabolic syndrome, type 2 diabetes mellitus, and cardiovascular disease. Adiponectin has emerged as a salutary adipocytokine, with insulin-sensitizing, anti-inflammatory, and cardiovascular protective properties. However, under metabolically unfavorable conditions, visceral adipose tissue-derived inflammatory cytokines might reduce the transcription of the adiponectin gene and consequently its circulating levels. Low circulating levels of adiponectin are negatively associated with various conditions, such as insulin resistance, type 2 diabetes mellitus, metabolic syndrome, and cardiovascular disease. In contrast, several recent clinical trials and meta-analyses have reported high circulating adiponectin levels positively associated with cardiovascular mortality and all-cause mortality. These results are biologically intriguing and counterintuitive, and came to be termed "the adiponectin paradox". Adiponectin paradox is frequently associated with adiponectin resistance, a concept related with the downregulation of adiponectin receptors in insulin-resistant states. We review this contradiction between the apparent role of adiponectin as a health promoter and the recent evidence from Mendelian randomization studies indicating that circulating adiponectin levels are an unexpected predictor of increased morbidity and mortality rates in several clinical conditions. We also critically review the therapeutic perspective of synthetic peptide adiponectin receptors agonist that has been postulated as a promising alternative for the treatment of metabolic syndrome and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Emilio Antonio Francischetti
- Laboratory of Clinical and Experimental Pathophysiology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Rômulo Sperduto Dezonne
- Postgraduate Program in Translational Biomedicine, Grande Rio University, Duque de Caxias, Brazil
| | - Cláudia Maria Pereira
- Postgraduate Program in Translational Biomedicine, Grande Rio University, Duque de Caxias, Brazil
| | - Cyro José de Moraes Martins
- Laboratory of Clinical and Experimental Pathophysiology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | | | - Virgínia Genelhu de Abreu
- Laboratory of Clinical and Experimental Pathophysiology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
39
|
Maximus PS, Al Achkar Z, Hamid PF, Hasnain SS, Peralta CA. Adipocytokines: Are they the Theory of Everything? Cytokine 2020; 133:155144. [PMID: 32559663 PMCID: PMC7297161 DOI: 10.1016/j.cyto.2020.155144] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/08/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Adipose tissue secretes various bioactive peptides/proteins, immune molecules and inflammatory mediators which are known as adipokines or adipocytokines. Adipokines play important roles in the maintenance of energy homeostasis, appetite, glucose and lipid metabolism, insulin sensitivity, angiogenesis, immunity and inflammation. Enormous number of studies from all over the world proved that adipocytokines are involved in the pathogenesis of diseases affecting nearly all body systems, which raises the question whether we can always blame adipocytokines as the triggering factor of every disease that may hit the body. OBJECTIVE Our review targeted the role played by adipocytokines in the pathogenesis of different diseases affecting different body systems including diabetes mellitus, kidney diseases, gynecological diseases, rheumatologic disorders, cancers, Alzheimer's, depression, muscle disorders, liver diseases, cardiovascular and lung diseases. METHODOLOGY We cited more than 33 recent literature reviews that discussed the role played by adipocytokines in the pathogenesis of different diseases affecting different body systems. CONCLUSION More evidence is being discovered to date about the role played by adipocytokines in more diseases and extra research is needed to explore hidden roles played by adipokine imbalance on disease pathogenesis.
Collapse
Affiliation(s)
- Pierre S Maximus
- California Institute of Behavioral Neurosciences and Psychology, United States.
| | - Zeina Al Achkar
- California Institute of Behavioral Neurosciences and Psychology, United States
| | - Pousette F Hamid
- California Institute of Behavioral Neurosciences and Psychology, United States
| | - Syeda S Hasnain
- California Institute of Behavioral Neurosciences and Psychology, United States
| | - Cesar A Peralta
- California Institute of Behavioral Neurosciences and Psychology, United States
| |
Collapse
|
40
|
Heydari M, Cornide-Petronio ME, Jiménez-Castro MB, Peralta C. Data on Adiponectin from 2010 to 2020: Therapeutic Target and Prognostic Factor for Liver Diseases? Int J Mol Sci 2020; 21:5242. [PMID: 32718097 PMCID: PMC7432057 DOI: 10.3390/ijms21155242] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
The review describes the role of adiponectin in liver diseases in the presence and absence of surgery reported in the literature in the last ten years. The most updated therapeutic strategies based on the regulation of adiponectin including pharmacological and surgical interventions and adiponectin knockout rodents, as well as some of the scientific controversies in this field, are described. Whether adiponectin could be a potential therapeutic target for the treatment of liver diseases and patients submitted to hepatic resection or liver transplantation are discussed. Furthermore, preclinical and clinical data on the mechanism of action of adiponectin in different liver diseases (nonalcoholic fatty disease, alcoholic liver disease, nonalcoholic steatohepatitis, liver cirrhosis and hepatocellular carcinoma) in the absence or presence of surgery are evaluated in order to establish potential targets that might be useful for the treatment of liver disease as well as in the practice of liver surgery associated with the hepatic resections of tumors and liver transplantation.
Collapse
Affiliation(s)
- Misaq Heydari
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.H.); (M.E.C.-P.)
| | | | - Mónica B. Jiménez-Castro
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.H.); (M.E.C.-P.)
| | - Carmen Peralta
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.H.); (M.E.C.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
| |
Collapse
|
41
|
Pétursdóttir Maack H, Larsson A, Axelsson O, Olovsson M, Wikström AK, Sundström Poromaa I. Pregnancy in metabolic healthy and unhealthy obese women. Acta Obstet Gynecol Scand 2020; 99:1640-1648. [PMID: 32491196 DOI: 10.1111/aogs.13929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 05/06/2020] [Accepted: 05/28/2020] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Obesity-associated adverse obstetric outcomes are well-known but still relatively rare and difficult to predict. Not all obese individuals are unhealthy; some have metabolically healthy obesity (MHO) and others have metabolic unhealthy obesity (MUO). Our aim was to investigate whether the MHO and MUO classification could be used for prediction of obesity-associated complications in pregnancy. MATERIAL AND METHODS We included 547 pregnant women with obesity, 2302 pregnant women with overweight and 5852 normal weight pregnant women. The women with obesity were classified as MUO (n = 181, 33.1%) or MHO (n = 366, 66.9%) based on first trimester blood pressure, lipoprotein levels and nonfasting glucose levels. The risk of obstetric outcomes was evaluated by multivariable logistic regression. We compared cardiovascular risk markers in blood between obese pregnant women with MUO and MHO. RESULTS Overall, 45.9% of women with MUO developed at least one adverse obesity-associated obstetric outcome, whereas corresponding proportions in women with MHO, overweight and normal weight were 35.0%, 27.6% and 21.2%, respectively. Following adjustment, the overall risk of at least one obesity-associated obstetric or perinatal complication in women with MUO, vs women with MHO, was increased (adjusted odds ratio 1.49, 95% confidence interval 1.03-2.15), but the predictive value of the model was low. Women with MUO had altered levels of cardiovascular risk markers such as cathepsin S, adiponectin, and cystatin C in comparison with women with MHO. CONCLUSIONS Women with MUO had an overall increased risk of adverse obstetric outcomes in comparison to women with MHO, but the classification had little relevance for risk prediction beyond that of body mass index.
Collapse
Affiliation(s)
| | - Anders Larsson
- Department of Medical Science, Uppsala University, Uppsala, Sweden
| | - Ove Axelsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.,Center for Clinical Research Sörmland, Uppsala University, Eskilstuna, Sweden
| | - Matts Olovsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Anna-Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
42
|
Artimani T, Najafi R. APPL1 as an important regulator of insulin and adiponectin‐signaling pathways in the PCOS: A narrative review. Cell Biol Int 2020; 44:1577-1587. [DOI: 10.1002/cbin.11367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/09/2020] [Accepted: 04/25/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Tayebe Artimani
- Endometrium and Endometriosis Research CenterHamadan University of Medical Sciences Hamadan Iran
| | - Rezvan Najafi
- Research Center for Molecular MedicineHamadan University of Medical Sciences Hamadan Iran
| |
Collapse
|
43
|
Leffler KE, Abdel-Rahman AA. Restoration of Adiponectin-Connexin43 Signaling Mitigates Myocardial Inflammation and Dysfunction in Diabetic Female Rats. J Cardiovasc Pharmacol 2020; 75:259-267. [PMID: 31868825 PMCID: PMC7537147 DOI: 10.1097/fjc.0000000000000789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ur preclinical findings replicated women's hypersensitivity to type-2 diabetes mellitus (T2DM)-evoked cardiac dysfunction along with demonstrating estrogen (E2)-dependent disruption of the cardiac adiponectin (APN)-connexin43 (Cx43) signaling. Whether the latter molecular anomaly underlies this women's cardiovascular health problem remains unknown. We hypothesized that restoration of the disrupted APN-Cx43 signaling alleviates this sex/E2-dependent cardiac dysfunction in diabetic female rats. To test this hypothesis, we administered the adiponectin receptor 1 (AdipoR1) agonist AdipoRon (30 mg/kg/d for 10 days) to female sham operated (SO) and ovariectomized (OVX) rats, which exhibited and lacked the T2DM left ventricular (LV) dysfunction, respectively, when fed high-fat diet and received low dose streptozotocin regimen; nondiabetic control SO and OVX rats received control diet and vehicle for streptozotocin. In T2DM SO rats, LV dysfunction, AdipoRon mitigated: (1) LV hypertrophy, (2) reductions in fractional shortening, LV developed pressure, dP/dtmax, dP/dtmin, and Tau. In LV tissues of the same rats, AdipoRon reversed reduction in Cx43 and elevations in TNFα, heme-oxygenase 1 (HO-1), and circulating cardiovascular risk factor asymmetric dimethylarginine. The findings also revealed ovarian hormones independent effects of AdipoRon, which included dampening of the pro-oxidant enzyme HO-1. These novel findings yield new insight into a causal role for compromised APN-Cx43 signaling in the E2-dependent hypersensitivity to T2DM-evoked cardiac inflammation and dysfunction. Equally important, the findings identify restoration of Cx43 signaling as a viable therapeutic modality for alleviating this women's cardiovascular health-related problem.
Collapse
MESH Headings
- Adiponectin/metabolism
- Animals
- Arginine/analogs & derivatives
- Arginine/metabolism
- Connexin 43/metabolism
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/prevention & control
- Estradiol/metabolism
- Estrogen Receptor alpha/metabolism
- Female
- Heme Oxygenase (Decyclizing)/metabolism
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Ovariectomy
- Piperidines/pharmacology
- Rats, Wistar
- Receptors, Adiponectin/agonists
- Receptors, Adiponectin/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Tumor Necrosis Factor-alpha/metabolism
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Korin E Leffler
- Department of Pharmacology and Toxicology, East Carolina University, Brody School of Medicine, Greenville, NC
| | | |
Collapse
|
44
|
Haghighatdoost F, Gholami A, Hariri M. Alpha-lipoic acid effect on leptin and adiponectin concentrations: a systematic review and meta-analysis of randomized controlled trials. Eur J Clin Pharmacol 2020; 76:649-657. [PMID: 32040596 DOI: 10.1007/s00228-020-02844-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/02/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND New evidence suggests that dysregulation of adipocytokines caused by excess adiposity plays an important role in the pathogenesis of various obesity comorbidities. Our aim in this meta-analysis was to determine the effect of alpha-lipoic acid (ALA) supplementation on serum levels of leptin and adiponectin. METHODS We searched Scopus, PubMed, Google Scholar, and ISI Web of Science from inception up to July 2019. Mean difference for leptin and adiponectin were calculated by subtracting the change from baseline in each study group. Summary estimates for the overall effect of ALA on serum leptin and adiponectin concentrations were calculated using random effects model. Results were presented as weighted mean difference (WMD) and their 95% confidence intervals (CI). Between-study heterogeneity was examined using the I2 statistics. RESULT Eight studies were included in systematic review and seven studies in meta-analysis. The overall effect suggested a significant decrement in serum leptin concentrations (WMD = - 3.63; 95% CI, - 5.63, - 1.64 μg/ml; I2 = 80.7%) and a significant increase in serum levels of adiponectin (WMD = 1.98 μg/ml; 95% CI, 0.92, 3.04; I2 = 95.7%). Subgroup analyses based on age showed a significant reduction in leptin levels only in younger adults, and subgroup analysis based on duration indicated in studies with a duration of more than 8 weeks adiponectin levels increased significantly and leptin levels decreased significantly. CONCLUSION Our results revealed ALA decreased leptin and increased adiponectin especially in studies lasted more than 8 weeks. We still need more studies with different ALA dose, intervention duration, and separately on male and female.
Collapse
Affiliation(s)
- Fahimeh Haghighatdoost
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Gholami
- Department of Epidemiology & Biostatistics, School of Public Health, Neyshabur University of Medical Sciences, Neyshabur, Iran.,Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Mitra Hariri
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
45
|
Adiyaman SC, Ozer M, Saydam BO, Akinci B. The Role of Adiponectin in Maintaining Metabolic Homeostasis. Curr Diabetes Rev 2020; 16:95-103. [PMID: 31267874 DOI: 10.2174/1573399815666190702155733] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/22/2019] [Accepted: 06/20/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Adiponectin is an adipocyte-derived cytokine closely associated with obesity, altered body adipose tissue distribution, insulin resistance, and cardiovascular diseases. INTRODUCTION Evidence from animal and human studies demonstrate that adiponectin plays an important role in the regulation of glucose and lipid metabolism. Adiponectin increases insulin sensitivity and improves systemic lipid metabolism. Although research efforts on adiponectin mostly aim towards its endocrine functions, this adipocyte-derived molecule also has profound autocrine and paracrine functions. CONCLUSION In this review, our aim is to discuss the role of adiponectin in maintaining metabolic homeostasis and its association with cardiovascular health. The proper identification of these roles is of great importance, which has the potential to identify a wealth of novel targets for the treatment of diabetes and related cardio-metabolic diseases.
Collapse
Affiliation(s)
| | - Muhammet Ozer
- Department of Internal Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Basak Ozgen Saydam
- Division of Endocrinology and Metabolism, Dokuz Eylul University, Izmir, Turkey
| | - Baris Akinci
- Division of Endocrinology and Metabolism, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
46
|
Lian K, Feng YN, Li R, Liu HL, Han P, Zhou L, Li CX, Wang Q. Middle- and high-molecular weight adiponectin levels in relation to nonalcoholic fatty liver disease. J Clin Lab Anal 2019; 34:e23148. [PMID: 31880002 PMCID: PMC7171302 DOI: 10.1002/jcla.23148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 10/21/2019] [Accepted: 11/15/2019] [Indexed: 12/14/2022] Open
Abstract
Objective Adiponectin (APN) circulates as high‐molecular weight (HMW), medium‐molecular weight (MMW), and low‐molecular weight (LMW) forms. Nonalcoholic fatty liver disease (NAFLD) is a common cause of chronic liver disease. Currently, the role of LMW, MMW, and HMW APN remains largely unclear in NAFLD. Methods We examined the variation of these forms and analyzed the related clinical characteristics in NAFLD. A total of 63 male NAFLD patients (mean age: 43.00 ± 6.10 years) and 70 healthy male subjects (mean age: 42.53 ± 7.98 years) were included in the study. Total APN and other clinical characteristics were measured. The changes in HMW, MMW, and LMW APN were determined in NAFLD patients and NAFLD patients on a high‐fat diet, and the association between the groups was further analyzed. Results Decreased levels of total APN and three APN isoforms were found in NAFLD. Significantly decreased levels of HMW (P < .01) and MMW (P < .001) were observed in NAFLD of high‐fat diet patients. In NAFLD patients, height (R = −.270, P = .032) and N‐epsilon‐(carboxymethyl) lysine (R = −.259, P = .040) significantly correlated with total APN. HMW APN was significantly associated with fasting plasma glucose (R = .350, P = .016), alanine aminotransferase (R = −.321, P = .029), and aspartate aminotransferase (R = −.295, P = .045). Additionally, MMW APN was significantly associated with total cholesterol (R = .357, P = .014) and high‐density lipoprotein (R = .556, P < .0001). Low‐density lipoprotein (R = −.283, P = .054) was also clearly associated with LMW APN in NAFLD patients. Conclusion These results suggest that HMW and MMW APN may be involved in the pathogenesis and progression of NAFLD.
Collapse
Affiliation(s)
- Kun Lian
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yu-Nan Feng
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Rong Li
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hao-Lin Liu
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Peng Han
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lei Zhou
- Department of Clinical Laboratory Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Cheng-Xiang Li
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qin Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
47
|
Adelmidrol protects against non-alcoholic steatohepatitis in mice. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:777-784. [PMID: 31853615 DOI: 10.1007/s00210-019-01785-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is a more serious condition of non-alcoholic fatty liver disease (NAFLD), a widely spread type of chronic liver disease. Oxidative stress and inflammation induced by lipotoxicity are involved in hepatic injury of NASH. This work aimed to investigate the possible protective effect of adelmidrol (PPAR α and γ agonist) against NASH in mice.Thirty-six mice were divided into six equal groups: sham-operated, NASH, vehicle, adelmidrol 5 mg/kg, adelmidrol 10 mg/kg, and adelmidrol 20 mg /kg. Adelmidrol was injected intraperitoneally once daily for 7 weeks along with high-fat diet (HFD). Mice were euthanized, and livers were removed for light microscopic examination and detection of reduced glutathione (GSH) content while blood samples were collected for assessment of transaminases (ALT& AST), tumor necrosis factor (TNF-α), matrix metalloproteinase-1 (MMP-1), adiponectin, cholesterol, high-density lipoprotein (HDL), and triglyceride. NASH mice had increments in MMP-1, TNF-α, AST, ALT, triglyceride, and cholesterol levels while HDL, adiponectin levels, and GSH content were decreased with vesicular steatosis, lobular inflammation, hepatocyte ballooning, and degeneration. Administration of adelmidrol decreased MMP-1, TNF-α, AST, ALT, triglyceride, and cholesterol levels while increased HDL, adiponectin levels, and GSH content as well as ameliorated the histopathological changes. Adelmidrol protected mice from NASH; an effect could be attributed to its anti-inflammatory and antioxidant actions.
Collapse
|
48
|
Abstract
PURPOSE OF THE REVIEW Osteoarthritis (OA) is an aging-associated and injury-induced joint disease characterized by cartilage degradation, bone sclerosis, and persistent low-grade inflammation in the joint. Aging and injury are triggers of joint pathological changes mediated by pro-inflammatory factors, some of which are secreted by white adipose tissue. Adipokines including adiponectin, leptin, resistin, chemerin, IL-6, and TNF-α are major players not only during inflammation but also in metabolic regulation of joint cells including chondrocytes, osteoblasts, osteoclasts as well as mesenchymal stem cells. The purpose of this review is to summarize the signal transduction pathways of adipokines in the articular joint to provide new information on potential targets for intervention of OA. RECENT FINDINGS The risk of knee osteoarthritis is associated with adipokine gene polymorphism. While the infrapatellar fat pad is a major source of adipokines in knee synovial fluid, adipocytes also accumulate in the bone marrow during aging and obesity. Adipokines can act as SASPs (senescence associated secretory phenotype factors) that participate in cellular senescence of chondrocytes, but they also regulate energy metabolism impacting bone remodeling. Thus, adipokines are closely related to the metabolic syndrome and degenerative pathological changes in cartilage and bone during OA. Modulating the effects of adipokines on different cell types in the intra-articular joint will be a promising new option for OA intervention.
Collapse
Affiliation(s)
- Chenxi Xie
- Bone and Joint Research Center, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qian Chen
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, 02903, USA.
| |
Collapse
|
49
|
Saghebjoo M, Farrokhi-Fard M, Hedayati M, Sadeghi-Tabas S. The effect of high-intensity interval training and L-arginine supplementation on the serum levels of adiponectin and lipid profile in overweight and obese young men. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.obmed.2019.100139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Hafiane A, Gasbarrino K, Daskalopoulou SS. The role of adiponectin in cholesterol efflux and HDL biogenesis and metabolism. Metabolism 2019; 100:153953. [PMID: 31377319 DOI: 10.1016/j.metabol.2019.153953] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/27/2022]
Abstract
Cholesterol efflux is the initial step in the reverse cholesterol transport pathway by which excess cholesterol in peripheral cells is exported and subsequently packaged into high-density lipoprotein (HDL) particles. Adiponectin is the most abundantly secreted adipokine that possesses anti-inflammatory and vasculoprotective properties via interaction with transmembrane receptors, AdipoR1 and AdipoR2. Evidence suggests that low levels of adiponectin may be a useful marker for atherosclerotic disease. A proposed anti-atherogenic mechanism of adiponectin involves its ability to promote cholesterol efflux. We performed a systematic review of the role of adiponectin in cholesterol efflux and HDL biogenesis, and of the proteins and receptors believed to be implicated in this process. Nineteen eligible studies (7 clinical, 11 fundamental, 1 clinical + fundamental) were identified through Ovid Medline, Ovid Embase, and Pubmed, that support the notion that adiponectin plays a key role in promoting ABCA1-dependent cholesterol efflux and in modulating HDL biogenesis via activation of the PPAR-γ/LXR-α signalling pathways in macrophages. AdipoR1 and AdipoR2 are suggested to also be implicated in this process, however the data are conflicting/insufficient to establish any firm conclusions. Once the exact mechanisms are unravelled, adiponectin may be critical in defining future treatment strategies directed towards increasing HDL functionality and ultimately reducing atherosclerotic disease.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| | - Karina Gasbarrino
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| | - Stella S Daskalopoulou
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|