1
|
Xuan X, Zhang S. Targeting the programmed cell death (PCD) signaling mechanism with natural substances for the treatment of diabetic cardiomyopathy (DCM). Phytother Res 2023; 37:5495-5508. [PMID: 37622685 DOI: 10.1002/ptr.7992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/27/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023]
Abstract
Diabetic cardiomyopathy (DCM) is one of the severe complications of diabetes, characterized by structural and functional abnormalities in the hearts of diabetic patients without hypertension, coronary heart disease, or valvular heart disease. DCM can progress to heart failure, which is a significant cause of death in diabetic patients, but currently, there is no effective treatment available. Programmed cell death (PCD) is a genetically regulated form of cell death that includes apoptosis, autophagy, necroptosis, ferroptosis, and pyroptosis. PCD is essential for tissue homeostasis and normal development of the body. DCM is a complex condition, and abnormalities in the cascade of PCD signaling have been observed in its pathological process, suggesting that targeting PCD could be a potential therapeutic strategy. Studies have shown that natural substances can effectively modulate PCD to intervene in the treatment of DCM, and their use is safe. This review explores the role of different forms of PCD in the pathogenesis of DCM and summarizes the research progress in targeting PCD with natural substances to treat DCM. It can serve as a basis for further research and drug development to provide new treatment strategies for DCM patients.
Collapse
Affiliation(s)
- Xiaoyu Xuan
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shiliang Zhang
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
2
|
Peng J, Chen Q, Wu C. The role of adiponectin in cardiovascular disease. Cardiovasc Pathol 2023; 64:107514. [PMID: 36634790 DOI: 10.1016/j.carpath.2022.107514] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease (CVD) is a common disease that seriously threatens the health of human beings, especially middle-aged and elderly people over 50 years old. It has the characteristics of high prevalence, high disability rate and high mortality rate. Previous studies have shown that adiponectin has therapeutic effects on a variety of CVDs. As a key adipokine, adiponectin, is an abundant peptide-regulated hormone that is mainly released by adipocytes and cardiomyocytes, as well as endothelial and skeletal cells. Adiponectin can protect against CVD by improving lipid metabolism, protecting vascular endothelial cells and inhibiting foam cell formation and vascular smooth muscle cell proliferation. Further investigation of the molecular and cellular mechanisms underlying the adiponectin system may provide new ideas for the treatment of CVD. Herein, this review aims to describe the structure and function of adiponectin and adiponectin receptors, introduce the function of adiponectin in the protection of cardiovascular disease and analyze the potential use and clinical significance of this hormone in the protection and treatment of cardiovascular disease, which shows that adiponectin can be expected to become a new therapeutic target and biomarker for the diagnosis and treatment of CVD.
Collapse
Affiliation(s)
- Jin Peng
- Clinical Medical Research Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qian Chen
- Clinical Medical Research Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Chuncao Wu
- Insititution of Chinese Materia Medica Preparation, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China.
| |
Collapse
|
3
|
Zhou Y, Yang Y, Zhou T, Li B, Wang Z. Adiponectin and Thyroid Cancer: Insight into the Association between Adiponectin and Obesity. Aging Dis 2021; 12:597-613. [PMID: 33815885 PMCID: PMC7990371 DOI: 10.14336/ad.2020.0919] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 09/13/2020] [Indexed: 12/14/2022] Open
Abstract
In recent decades, the incidence and diagnosis of thyroid cancer have risen dramatically, and thyroid cancer has now become the most common endocrine cancer in the world. The onset of thyroid cancer is insidious, and its progression is slow and difficult to detect. Therefore, early prevention and treatment have important strategic significance. Moreover, an in-depth exploration of the pathogenesis of thyroid cancer is key to early prevention and treatment. Substantial evidence supports obesity as an independent risk factor for thyroid cancer. Adipose tissue dysfunction in the obese state is accompanied by dysregulation of a variety of adipocytokines. Adiponectin (APN) is one of the most pivotal adipocytokines, and its connection with obesity and obesity-related disease has gradually become a hot topic in research. Recently, the association between APN and thyroid cancer has received increasing attention. The purpose of this review is to systematically review previous studies, give prominence to APN, focus on the relationship between APN, obesity and thyroid cancer, and uncover the underlying pathogenic mechanisms.
Collapse
Affiliation(s)
- Yuanyuan Zhou
- 1Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province, Fourth Affiliated Hospital of Kunming Medical University, Kunming, China.,2Department of Endocrinology and Metabolism, Sixth Affiliated Hospital of Kunming Medical University, The People's Hospital of Yuxi City, Yuxi, China
| | - Ying Yang
- 1Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province, Fourth Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Taicheng Zhou
- 1Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province, Fourth Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bai Li
- 3School of Medicine, Yunnan University, Kunming, China
| | - Zhanjian Wang
- 4Department of Endocrinology and Metabolism, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
4
|
Chen L, Liu S, Xu W, Zhang Y, Bai J, Li L, Cui M, Sun L. Association of Plasma C1q/TNF-Related Protein 3 (CTRP3) in Patients with Atrial Fibrillation. Mediators Inflamm 2020; 2020:8873152. [PMID: 33424438 PMCID: PMC7781729 DOI: 10.1155/2020/8873152] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/12/2020] [Indexed: 01/03/2023] Open
Abstract
Atrial fibrillation (AF) is a highly prevalent cardiac arrhythmia characterized by atrial remodeling. Complement C1q tumor necrosis factor-related protein 3 (CTRP3) is one of the adipokines associated with obesity, diabetes, and coronary heart disease. The association between plasma CTRP3 levels and AF is uncertain. The aim of this study was to investigate whether plasma CTRP3 concentrations were correlated with AF. Our study included 75 AF patients who underwent catheter ablation at our hospital and 47 sinus rhythm patients to determine the difference in plasma CTRP3 concentrations. Blood samples before the ablation were collected, and ELISA was used to measure the concentrations of CTRP3. Plasma CTRP3 concentrations were significantly lower in AF patients compared with control group (366.9 ± 105.2 ng/ml vs. 429.1 ± 100.1 ng/ml, p = 0.002). In subgroup studies, patients with persistent AF had lower plasma CTRP3 concentrations than those with paroxysmal AF (328.3 ± 83.3 ng/ml vs. 380.0 ± 109.2 ng/ml, p = 0.037). The concentrations of plasma CTRP3 in the recurrence group after radiofrequency catheter ablation of AF were lower than those in the nonrecurrence group (337.9 ± 77.3 ng/ml vs. 386.6 ± 108.1 ng/ml, p = 0.045). Multivariate regression analysis revealed the independent correlation between plasma CTRP3 level and AF. Plasma CTRP3 concentrations were correlated with the presence of AF and AF recurrence.
Collapse
Affiliation(s)
- Liwen Chen
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Shuwang Liu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Wei Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Yuan Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Jin Bai
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Lei Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Ming Cui
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Lijie Sun
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, 49 Huayuan-Bei Road, Haidian District, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| |
Collapse
|
5
|
Kita S, Maeda N, Shimomura I. Interorgan communication by exosomes, adipose tissue, and adiponectin in metabolic syndrome. J Clin Invest 2020; 129:4041-4049. [PMID: 31483293 DOI: 10.1172/jci129193] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue plays important roles in regulating whole-body energy metabolism through its storage function in white adipocytes and its dissipating function in brown and beige adipocytes. Adipose tissue also produces a variety of secreted factors called adipocytokines, including leptin and adiponectin. Furthermore, recent studies have suggested the important roles of extracellular vesicles of endosomal origin termed exosomes, which are secreted from adipocytes and other cells in adipose tissue and influence whole-body glucose and lipid metabolism. Adiponectin is known to be a pleiotropic organ-protective protein that is exclusively produced by adipocytes and decreased in obesity. Adiponectin accumulates in tissues such as heart, muscle, and vascular endothelium through binding with T-cadherin, a glycosylphosphatidylinositol-anchored (GPI-anchored) cadherin. Recently, adiponectin was found to enhance exosome biogenesis and secretion, leading to a decrease in cellular ceramides, excess of which is known to cause insulin resistance and cardiovascular disease phenotypes. These findings support the hypothesis that adipose tissue metabolism systemically regulates exosome production and whole-body metabolism through exosomes. This review focuses on intra-adipose and interorgan communication by exosomes, adiponectin-stimulated exosome production, and their dysregulation in metabolic diseases.
Collapse
Affiliation(s)
- Shunbun Kita
- Department of Metabolic Medicine.,Department of Adipose Management, and
| | - Norikazu Maeda
- Department of Metabolic Medicine.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | |
Collapse
|
6
|
Hamjane N, Benyahya F, Nourouti NG, Mechita MB, Barakat A. Cardiovascular diseases and metabolic abnormalities associated with obesity: What is the role of inflammatory responses? A systematic review. Microvasc Res 2020; 131:104023. [PMID: 32485192 DOI: 10.1016/j.mvr.2020.104023] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Obesity is a chronic disease responsible for a high morbidity and mortality rate, with an increasing worldwide prevalence. Obesity is associated with immune responses characterized by chronic systemic inflammation. This article focuses on the mechanisms that explain the proposed link between obesity-associated diseases and inflammation. Also, it describes the role of inflammatory molecules in obesity-associated metabolic abnormalities. METHODS More than 200 articles were selected and consulted by an online English search using various electronic search databases. Predefined key-words for the pathogenesis of obesity-induced inflammation and associated diseases, as well as the role of various inflammatory molecules, were used. RESULTS We have summarized the data of the articles consulted in this research and we have found that obesity is associated with a low-grade inflammation resulting from the change of adipose tissue (AT). The AT produces a variety of inflammatory molecules called adipocytokines that are involved in the onset of systemic low-grade inflammation which is the link between obesity and associated-chronic abnormalities; such as insulin resistance, metabolic syndrome, cardiovascular disease (CVD), hypertension, diabetes, and some cancers. Also, we have searched all the inflammatory molecules involved in this pathogenesis and we have briefly described the role of 16 of them which are the most related to obesity-associated inflammation. The results have shown that there are inflammatory molecules that have a positive relationship with the pathogenesis of obesity-related diseases and others have a negative relationship with this pathogenesis. CONCLUSION Inflammation plays a crucial role in the development of various metabolic-abnormalities related to obesity. In this regard, the management of obesity may help reduce the risk of cardiovascular disease and other metabolic complications by inhibiting inflammatory mechanisms.
Collapse
Affiliation(s)
- Nadia Hamjane
- Laboratory of Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology, Abdelmalek Essaadi University, Tangier, Morocco.
| | | | - Naima Ghailani Nourouti
- Laboratory of Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology, Abdelmalek Essaadi University, Tangier, Morocco
| | - Mohcine Bennani Mechita
- Laboratory of Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology, Abdelmalek Essaadi University, Tangier, Morocco
| | - Amina Barakat
- Laboratory of Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology, Abdelmalek Essaadi University, Tangier, Morocco
| |
Collapse
|
7
|
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
|
8
|
Liu Y, Vu V, Sweeney G. Examining the Potential of Developing and Implementing Use of Adiponectin-Targeted Therapeutics for Metabolic and Cardiovascular Diseases. Front Endocrinol (Lausanne) 2019; 10:842. [PMID: 31920962 PMCID: PMC6918867 DOI: 10.3389/fendo.2019.00842] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiometabolic diseases encompass those affecting the heart and vasculature as well as other metabolic problems, such as insulin resistance, diabetes, and non-alcoholic fatty liver disease. These diseases tend to have common risk factors, one of which is impaired adiponectin action. This may be due to reduced bioavailability of the hormone or resistance to its effects on target tissues. A strong negative correlation between adiponectin levels and cardiometabolic diseases has been well-documented and research shown that adiponectin has cardioprotective, insulin sensitizing and direct beneficial metabolic effects. Thus, therapeutic approaches to enhance adiponectin action are widely considered to be desirable. The complexity of adiponectin structure and function has so far made progress in this area less than ideal. In this article we will review the effects and mechanism of action of adiponectin on cardiometabolic tissues, identify scenarios where enhancing adiponectin action would be of clinical value and finally discuss approaches via which this can be achieved.
Collapse
Affiliation(s)
- Ying Liu
- Metabolic Disease Research Division, iCarbonX Co. Ltd., Shenzhen, China
- *Correspondence: Ying Liu
| | - Vivian Vu
- Department of Biology, York University, Toronto, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
- Gary Sweeney
| |
Collapse
|
9
|
Dludla PV, Nkambule BB, Jack B, Mkandla Z, Mutize T, Silvestri S, Orlando P, Tiano L, Louw J, Mazibuko-Mbeje SE. Inflammation and Oxidative Stress in an Obese State and the Protective Effects of Gallic Acid. Nutrients 2018; 11:nu11010023. [PMID: 30577684 PMCID: PMC6356415 DOI: 10.3390/nu11010023] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/08/2018] [Accepted: 11/17/2018] [Indexed: 12/12/2022] Open
Abstract
Metabolic complications in an obese state can be aggravated by an abnormal inflammatory response and enhanced production of reactive oxygen species. Pro-inflammatory response is known to be associated with the formation of toxic reactive oxygen species and subsequent generation of oxidative stress. Indeed, adipocytes from obese individuals display an altered adipokine profile, with upregulated expression and secretion of pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin (IL-6). Interestingly, natural compounds, including phenolic enriched foods are increasingly explored for their ameliorative effects against various metabolic diseases. Of interest is gallic acid, a trihydroxybenzoic acid that has progressively demonstrated robust anti-obesity capabilities in various experimental models. In addition to reducing excessive lipid storage in obese subjects, gallic acid has been shown to specifically target the adipose tissue to suppress lipogenesis, improve insulin signaling, and concomitantly combat raised pro-inflammatory response and oxidative stress. This review will revise mechanisms involved in the pathophysiological effects of inflammation and oxidative stress in an obese state. To better inform on its therapeutic potential and improvement of human health, available evidence reporting on the anti-obesity properties of gallic acid and its derivatives will be discussed, with emphases on its modulatory effect on molecular mechanisms involved in insulin signaling, inflammation and oxidative stress.
Collapse
Affiliation(s)
- Phiwayinkosi V Dludla
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy.
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| | - Babalwa Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
| | - Zibusiso Mkandla
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| | - Tinashe Mutize
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy.
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy.
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy.
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa.
| | - Sithandiwe E Mazibuko-Mbeje
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.
| |
Collapse
|
10
|
Ge J, Cheng B, Qi B, Peng W, Wen H, Bai L, Liu Y, Zhai W. Cloning and functional analysis of human acyl coenzyme A: Cholesterol acyltransferase1 gene P1 promoter. Mol Med Rep 2016; 14:831-8. [PMID: 27220725 DOI: 10.3892/mmr.2016.5295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 04/13/2016] [Indexed: 11/05/2022] Open
Abstract
Acyl-coenzyme A: cholesterol acyltransferase 1 (ACAT1) catalyzes the conversion of free cholesterol (FC) to cholesterol ester. The human ACAT1 gene P1 promoter has been cloned. However, the activity and specificity of the ACAT1 gene P1 promoter in diverse cell types remains unclear. The P1 promoter fragment was digested with KpnI/XhoI from a P1 promoter cloning vector, and was subcloned into the multiple cloning site of the Firefly luciferase vector pGL3‑Enhancer to obtain the construct P1E‑1. According to the analysis of biological information, the P1E‑1 plasmid was used to generate deletions of the ACAT1 gene P1 promoter with varying 5' ends and an identical 3' end at +65 by polymerase chain reaction (PCR). All the 5'‑deletion constructs of the P1 promoter were identified by PCR, restriction enzyme digestion mapping and DNA sequencing. The transcriptional activity of each construct was detected after transient transfection into THP‑1, HepG2, HEK293 and Hela cells using DEAE‑dextran and Lipofectamine 2000 liposome transfection reagent. Results showed that the transcriptional activity of the ACAT1 gene P1 promoter and deletions of P1 promoter in THP‑1 and HepG2 cells was higher than that in HEK293 and HeLa cells. Moreover, the transcriptional activity of P1E‑9 was higher compared with those of other deletions in THP‑1, HepG2, HEK293 and HeLa cells. These findings indicate that the transcriptional activity of the P1 promoter and the effects of deletions vary with different cell lines. Thus, the P1 promoter may drive ACAT1 gene expression with cell‑type specificity. In addition, the core sequence of ACAT1 gene P1 promoter was suggested to be between -125 and +65 bp.
Collapse
Affiliation(s)
- Jing Ge
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Bei Cheng
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Benling Qi
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wen Peng
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hui Wen
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lijuan Bai
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wei Zhai
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
11
|
Yamamoto H, Kuroda N, Uekita H, Kochi I, Matsumoto A, Niinaga R, Funahashi T, Shimomura I, Kihara S. E-selectin ligand-1 (ESL-1) is a novel adiponectin binding protein on cell adhesion. Biochem Biophys Res Commun 2016; 470:425-430. [PMID: 26792720 DOI: 10.1016/j.bbrc.2016.01.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/05/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Adiponectin (APN) is an adipocyte-derived bioactive molecule with anti-diabetic and anti-atherogenic properties. Although anti-diabetic effects are mostly mediated by the adiponectin receptors AdipoR1 and AdipoR2, the anti-atherogenic mechanisms have not been fully elucidated. METHODS AND RESULTS In this study, we identified E-selectin ligand (ESL)-1 as a novel APN-binding protein by mass spectrometry analysis of HepG2 cell-derived immunoprecipitant with an anti-APN antibody. Cell adhesion assays using fluorescence-labelled monocyte cell line THP-1 cells and human umbilical vein endothelial cells (HUVECs) revealed that APN-pre-treated THP-1 cells had reduced binding ability to HUVECs. This APN-mediated suppressive effect on monocyte binding to endothelial cells was partially abrogated by targeting ESL-1 with shRNA in THP-1 cells. In addition, serial mutagenesis analysis disclosed that five extracellular amino acids close to the N-terminus of ESL-1 were essential for binding with APN. CONCLUSION Our results highlight the fact that interaction between APN and ESL-1 could provide a fundamental mechanism underlying the anti-atherogenic properties of APN.
Collapse
Affiliation(s)
- Hiroyasu Yamamoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nana Kuroda
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiromi Uekita
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ikoi Kochi
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akane Matsumoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryu Niinaga
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tohru Funahashi
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinji Kihara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
12
|
Ebrahimi-Mamaeghani M, Mohammadi S, Arefhosseini SR, Fallah P, Bazi Z. Adiponectin as a potential biomarker of vascular disease. Vasc Health Risk Manag 2015; 11:55-70. [PMID: 25653535 PMCID: PMC4303398 DOI: 10.2147/vhrm.s48753] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The increasing prevalence of diabetes and its complications heralds an alarming situation worldwide. Obesity-associated changes in circulating adiponectin concentrations have the capacity to predict insulin sensitivity and are a link between obesity and a number of vascular diseases. One obvious consequence of obesity is a decrease in circulating levels of adiponectin, which are associated with cardiovascular disorders and associated vascular comorbidities. Human and animal studies have demonstrated decreased adiponectin to be an independent risk factor for cardiovascular disease. However, in animal studies, increased circulating adiponectin alleviates obesity-induced endothelial dysfunction and hypertension, and also prevents atherosclerosis, myocardial infarction, and diabetic cardiac tissue disorders. Further, metabolism of a number of foods and medications are affected by induction of adiponectin. Adiponectin has beneficial effects on cardiovascular cells via its antidiabetic, anti-inflammatory, antioxidant, antiapoptotic, antiatherogenic, vasodilatory, and antithrombotic activity, and consequently has a favorable effect on cardiac and vascular health. Understanding the molecular mechanisms underlying the regulation of adiponectin secretion and signaling is critical for designing new therapeutic strategies. This review summarizes the recent evidence for the physiological role and clinical significance of adiponectin in vascular health, identification of the receptor and post-receptor signaling events related to the protective effects of the adiponectin system on vascular compartments, and its potential use as a target for therapeutic intervention in vascular disease.
Collapse
MESH Headings
- Adiponectin/immunology
- Adiponectin/metabolism
- Adipose Tissue/immunology
- Adipose Tissue/metabolism
- Adipose Tissue/physiopathology
- Animals
- Biomarkers/metabolism
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Humans
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Prognosis
- Protective Factors
- Receptors, Adiponectin/metabolism
- Risk Factors
- Signal Transduction
- Vascular Diseases/immunology
- Vascular Diseases/metabolism
- Vascular Diseases/physiopathology
- Vascular Diseases/prevention & control
Collapse
Affiliation(s)
| | - Somayeh Mohammadi
- Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Rafie Arefhosseini
- Department of Food Technology, Faculty of Nutrition Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Fallah
- Department of Molecular Biology and Genetic Engineering, Stem Cell Technology Research Center, Tehran, Iran
| | - Zahra Bazi
- Department of Biotechnology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Arnaboldi L, Corsini A. Could changes in adiponectin drive the effect of statins on the risk of new-onset diabetes? The case of pitavastatin. ATHEROSCLEROSIS SUPP 2015; 16:1-27. [DOI: 10.1016/s1567-5688(14)70002-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
14
|
Obesity and atherosclerosis: mechanistic insights. Can J Cardiol 2014; 31:177-83. [PMID: 25661552 DOI: 10.1016/j.cjca.2014.11.031] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 11/25/2014] [Accepted: 11/25/2014] [Indexed: 12/19/2022] Open
Abstract
Obesity is a multifactorial chronic disease characterized by an accumulation of visceral and subcutaneous fat, which leads to a predisposition toward cardiometabolic diseases. A plethora of mechanisms, including abnormalities in lipid metabolism, insulin resistance, inflammation, endothelial dysfunction, adipokine imbalance, and inflammasome activation have been suggested to underlie the relationship between obesity and atherosclerosis. More recent data point toward an emerging role of impaired autophagy and altered gut microbiome homeostasis as potentially contributing factors. This review provides an overview of this area.
Collapse
|
15
|
Troglitazone and Δ2Troglitazone enhance adiponectin expression in monocytes/macrophages through the AMP-activated protein kinase pathway. Mediators Inflamm 2014; 2014:726068. [PMID: 25328285 PMCID: PMC4189946 DOI: 10.1155/2014/726068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/28/2014] [Indexed: 01/01/2023] Open
Abstract
Accumulating evidence indicates that the regimen to increase adiponectin will provide a novel therapeutic strategy for inflammation and cardiovascular disorders. Here, we tested the effect of troglitazone (TG) and its newly synthesized derivative, 5-[4-(6-hydroxy-2,5,7,8-tetramethyl-chroman-2-yl-methoxy)-benzylidene]-2,4-thiazolidinedione (Δ2troglitazone, (Δ2TG)), on the adiponectin expression in monocytes/macrophages and the relative mechanisms. The expression of adiponectin was located in macrophages of atherosclerotic lesions from patients and cholesterol-fed rabbits. TG and Δ2TG enhanced adiponectin mRNA and protein expression in THP-1 cells by quantitative real-time PCR, Western blot, and immunocytochemistry. TG induced adiponectin mRNA expression through a PPARγ-dependent pathway whereas Δ2TG enhanced adiponectin mRNA expression through a PPARγ-independent pathway in THP-1 cells. Both TG and Δ2TG enhanced adiponectin mRNA expression through AMP-activated protein kinase (AMPK) activation. TG and Δ2TG decreased the adhesion of THP-1 cells to TNF-α-treated HUVECs and the inhibitory effect was abolished by specific antiadiponectin antibodies. TG- and Δ2TG-induced suppression on monocyte adhesion were inhibited by a selective AMPK inhibitor compound C. Our data suggest that the inhibitory effect of TG and Δ2TG on monocyte adhesion might be at least in part through de novo adiponectin expression and activation of an AMPK-dependent pathway, which might play an important role in anti-inflammation and antiatherosclerosis.
Collapse
|
16
|
Long W, Hui Ju Z, Fan Z, Jing W, Qiong L. The effect of recombinant adeno-associated virus-adiponectin (rAAV2/1-Acrp30) on glycolipid dysmetabolism and liver morphology in diabetic rats. Gen Comp Endocrinol 2014; 206:1-7. [PMID: 25019654 DOI: 10.1016/j.ygcen.2014.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 06/29/2014] [Accepted: 07/03/2014] [Indexed: 01/11/2023]
Abstract
Adiponectin is an adipocytokine derived from adipocytes with insulin resistance-improving and anti-inflammatory activities. The level of Adiponectin is decreased in obesity, insulin resistance and Type 2 diabetes mellitus. The administration of recombinant adiponectin has been shown to improve hyperglycemia and insulin resistance in diabetic mice. Therefore, we investigated the effects of recombinant adeno-associated virus-adiponectin (rAAV2/1-Acrp30) on the glycolipid profile and liver morphology in streptozotocin-induced diabetic rats. Animals were fed a high-fat/high-glucose diet for 4weeks and diabetes induced by intraperitoneal administration of streptozotocin. The animals were divided randomly into four groups: diabetes control group, rAAV2/1-Acrp30 treatment group, vacuity virus group, and normal control group. Compared with diabetic rats and those in the vacuity virus group, animals treated with rAAV2/1-Acrp30 exhibited significantly lower values for glycaemic and lipidic profiles, and significantly higher levels of HDL. Although APN expression increased in the liver tissue, serum levels were not significantly increased. However, the rAAV2/1-Acrp30 treated animals showed amelioration of hepatic disease, accompanied by marked reduction in the expression of NF-κBp65 and IκBα. The results suggest that rAAV2/1-Acrp30 ameliorates glycolipid dysmetabolism and hepatic disease in streptozotocin-induced diabetic rats. These observations indicate that the function of rAAV2/1-Acrp30 is mediated by downregulated expression of NF-κBp65 and IκBα.
Collapse
Affiliation(s)
- Wen Long
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China.
| | - Zhong Hui Ju
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Zhang Fan
- Department of Anesthesia, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Wang Jing
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Li Qiong
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| |
Collapse
|
17
|
Piao L, Han Y, Li D. Correlation study on adiponectin gene SNP45 and long-term oxidative stress in patients with diabetes and carotid atherosclerosis. Exp Ther Med 2014; 8:707-712. [PMID: 25120586 PMCID: PMC4113525 DOI: 10.3892/etm.2014.1808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 03/05/2014] [Indexed: 11/23/2022] Open
Abstract
The aim of the present study was to investigate the correlation between the adiponectin gene single nucleotide polymorphism (SNP)45 T/G and long-term oxidative stress in type II diabetes mellitus (T2DM) patients with carotid atherosclerosis. Patients with T2DM were divided into non-carotid atherosclerosis and carotid atherosclerosis groups, which were then subsequently divided into TT and TG + GG groups according to the adiponectin SNP45 T/G genotypes. Enzyme-linked immunosorbent assay, TaqMan probe quantitative polymerase chain reaction (PCR), PCR-TaqMan, color Doppler and other methods were used to determine the adiponectin levels, gene polymorphisms, acquired mitochondrial DNA (mtDNA) A3243G somatic cell mutation rates and the carotid intima-media thickness. The somatic cell mutation rate of acquired mtDNA A3243A/G in the T2DM carotid atherosclerosis group was significantly higher compared with the group without carotid atherosclerosis. In addition, the acquired mtDNA A3243A/G somatic cell mutation rate in the T2DM carotid atherosclerosis group with the adiponectin gene SNP45 TT genotype was significantly lower compared with the SNP45 TG/GG genotype group. T2DM combined with carotid atherosclerosis was associated with long-term oxidative stress. In addition, adiponectin gene SNP45 T/G was associated with increased mtDNA A3243A/G somatic mutation rates in T2DM patients with carotid atherosclerosis. Therefore, adiponectin gene polymorphisms may lead to diabetes atherosclerosis through oxidative stress.
Collapse
Affiliation(s)
- Lianshan Piao
- Department of Endocrinology, The Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Yanhua Han
- Department of Endocrinology, The Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Dan Li
- Department of Endocrinology, The Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
18
|
Eguchi A, Kaneko Y, Murakami A, Ohigashi H. Zerumbone Suppresses Phorbol Ester-Induced Expression of Multiple Scavenger Receptor Genes in THP-1 Human Monocytic Cells. Biosci Biotechnol Biochem 2014; 71:935-45. [PMID: 17420607 DOI: 10.1271/bbb.60596] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Unregulated uptake of oxidized low-density lipoproteins (ox-LDL) via macrophage scavenger receptors (SRs), such as lectin-like ox-LDL receptor-1 (LOX-1), is a key event in atherosclerosis. In the present study, we used differentiated Caco-2 cells as a model of the human small intestine to evaluate the suppressive effects of 16 traditional food items selected from Okinawa on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced LOX-1 mRNA expression in THP-1 human monocyte-like cells. Three Zingiberaceae plants, Curcuma aromatica Salisbury, Curcuma longa L., and Zingiber zerumbet Smith, markedly suppressed that expression. When added to the apical sides of Caco-2 monolayers, zerumbone, a sesquiterpene from Z. zerumbet Smith, was found to permeate into the basolateral medium as an intact structure in a time-dependent manner. alpha-Humulene, a structural analog of zerumbone lacking the alpha,beta-unsaturated carbonyl group, did not suppress LOX-1 mRNA expression, indicating that its electrophilic moiety might play pivotal roles in its activities. Further, zerumbone attenuated the expression of SR-A, SR-PSOX, and CD36, but not that of CD68 or CLA-1, leading to a blockade of DiI-acLDL uptake, while it also inhibited the transcriptional activities of activator protein-1 and nuclear factor-kappaB. Together, our results indicate that zerumbone is a potential phytochemical for regulating atherosclerosis with reasonable action mechanisms.
Collapse
Affiliation(s)
- Ai Eguchi
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | | | | | | |
Collapse
|
19
|
Caselli C, D'Amico A, Cabiati M, Prescimone T, Del Ry S, Giannessi D. Back to the heart: the protective role of adiponectin. Pharmacol Res 2014; 82:9-20. [PMID: 24657240 DOI: 10.1016/j.phrs.2014.03.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/25/2014] [Accepted: 03/06/2014] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide and the prevalence of obesity and diabetes are increasing. In obesity, adipose tissue increases the secretion of bioactive mediators (adipokines) that may represent a key mechanism linking obesity to CVD. Adiponectin, extensively studied in metabolic diseases, exerts anti-diabetic, anti-atherogenic and anti-inflammatory activities. Due to these positive actions, the role of adiponectin in cardiovascular protection has been evaluated in recent years. In particular, for its potential therapeutic benefits in humans, adiponectin has become the subject of intense preclinical research. In the cardiovascular context, understanding of the cellular and molecular mechanisms underlying the adiponectin system, throughout its secretion, regulation and signaling, is critical for designing new drugs that target adiponectin system molecules. This review focused on recent advances regarding molecular mechanisms related to protective effects of the adiponectin system on both cardiac and vascular compartments and its potential use as a target for therapeutic intervention of CVD.
Collapse
Affiliation(s)
- C Caselli
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy.
| | - A D'Amico
- Scuola Superiore S. Anna, Pisa, Italy
| | - M Cabiati
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy
| | - T Prescimone
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy
| | - S Del Ry
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy
| | - D Giannessi
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy
| |
Collapse
|
20
|
Fu Y. Adiponectin Signaling and Metabolic Syndrome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:293-319. [DOI: 10.1016/b978-0-12-800101-1.00009-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Association of circulating levels of nicotinamide phosphoribosyltransferase (NAMPT/Visfatin) and of a frequent polymorphism in the promoter of the NAMPT gene with coronary artery disease in diabetic and non-diabetic subjects. Cardiovasc Diabetol 2013; 12:119. [PMID: 23968400 PMCID: PMC3765274 DOI: 10.1186/1475-2840-12-119] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 08/12/2013] [Indexed: 12/02/2022] Open
Abstract
Background Nicotinamide phosphoribosyltransferase (NAMPT) is the limiting enzyme in one of pathways of synthesis of Nicotinamide Adenine Dinucleotide, a redox coenzyme. NAMPT is considered as an insulin-mimetic factor and a potential regulatory factor in inflammatory and immune processes. Associations of circulating NAMPT levels with cardiovascular disease (CVD) and insulin resistance have been reported. We investigated association of circulating NAMPT levels and the rs9770242 NAMPT gene polymorphism with coronary artery disease (CAD). Methods We studied 594 Brazilian subjects undergoing a coronary angiography (49% of whom had type 2 diabetes). CAD, defined as stenosis greater than 50% in one major coronary vessel or branch, was observed in 68% of subjects. Genetic studies were also performed in 858 North-American Non-Hispanic White subjects with type 2 diabetes (49% with CAD). Results We observed an interaction between glycemic and CAD status on the comparison of NAMPT levels by CAD status. NAMPT levels were higher in type 2 diabetic patients with CAD as compared to those without CAD: 5.27 ± 2.93 ng/ml vs. 4.43 ± 2.94 ng/ml, p = 0.006 (mean ± SD). NAMPT levels were not significantly different in non-diabetic subjects with or without CAD. The T-allele of rs9770242 was associated with CAD in the Brazilian cohort (OR 1.46, 95% CI 1.06 - 2.01, p = 0.02) while no association was observed in the North-American cohort. Conclusions Our data suggest that circulating NAMPT levels are associated with CAD in type 2 diabetic patients. NAMPT rs9770242 polymorphism may be associated with CAD in some populations.
Collapse
|
22
|
Impact of adiponectin overexpression on allergic airways responses in mice. J Allergy (Cairo) 2013; 2013:349520. [PMID: 23861690 PMCID: PMC3686156 DOI: 10.1155/2013/349520] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/07/2013] [Indexed: 11/18/2022] Open
Abstract
Obesity is an important risk factor for asthma. Obese individuals have decreased circulating adiponectin, an adipose-derived hormone with anti-inflammatory properties. We hypothesized that transgenic overexpression of adiponectin would attenuate allergic airways inflammation and mucous hyperplasia in mice. To test this hypothesis, we used mice overexpressing adiponectin (Adipo Tg). Adipo Tg mice had marked increases in both serum adiponectin and bronchoalveolar lavage (BAL) fluid adiponectin. Both acute and chronic ovalbumin (OVA) sensitization and challenge protocols were used. In both protocols, OVA-induced increases in total BAL cells were attenuated in Adipo Tg versus WT mice. In the acute protocol, OVA-induced increases in several IL-13 dependent genes were attenuated in Adipo Tg versus WT mice, even though IL-13 per se was not affected. With chronic exposure, though OVA-induced increases in goblet cells numbers per millimeter of basement membrane were greater in Adipo Tg versus WT mice, mRNA abundance of mucous genes in lungs was not different. Also, adiponectin overexpression did not induce M2 polarization in alveolar macrophages. Our results indicate that adiponectin protects against allergen-induced inflammatory cell recruitment to the airspaces, but not development of goblet cell hyperplasia.
Collapse
|
23
|
Luo N, Chung BH, Wang X, Klein RL, Tang CK, Garvey WT, Fu Y. Enhanced adiponectin actions by overexpression of adiponectin receptor 1 in macrophages. Atherosclerosis 2013; 228:124-35. [PMID: 23510830 PMCID: PMC3640696 DOI: 10.1016/j.atherosclerosis.2013.02.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 01/29/2013] [Accepted: 02/21/2013] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Adiponectin is one of several important, metabolically active cytokines secreted from adipose tissue. Epidemiologic studies have associated low circulating levels of this adipokine with multiple metabolic disorders including obesity, insulin resistance, type II diabetes, and cardiovascular disease. To investigate how enhanced adiponectin-mediated changes in metabolism in vivo, we generated transgenic mice which specifically overexpress the gene coding for adiponectin receptor 1 (AdipoR1) in mouse macrophages using the human scavenger receptor A-I gene (SR-AI) enhancer/promoter. We found that macrophage-specific AdipoR1 transgenic mice (AdR1-TG) presented reduced whole body weight, fat accumulation and liver steatosis when these transgenic mice were fed with a high fat diet. Moreover, these macrophage AdR1-TG mice exhibited enhanced whole-body glucose tolerance and insulin sensitivity with reduced proinflammatory cytokines, MCP-1 and TNF-α, both in the serum and in the insulin target metabolic tissues. Additional studies demonstrated that these macrophage AdR1-TG animals exhibited reduced macrophage foam cell formation in the arterial wall when these transgenic mice were crossed with a low-density lipoprotein receptor (Ldlr) deficient mouse model. CONCLUSIONS These results suggest that AdipoR1 overexpressed in macrophages can physiologically modulate metabolic activities in vivo by enhancing adiponectin actions in distal metabolically active tissues. The AdipoR1 modified macrophages provide unique interactions with the residented tissues/cells, suggesting a novel role of macrophage adiponectin receptor in improving metabolic disorders in vivo.
Collapse
MESH Headings
- Adiponectin/blood
- Adipose Tissue/metabolism
- Animals
- Blood Glucose/metabolism
- Carrier Proteins/genetics
- Cells, Cultured
- Cholesterol/blood
- Disease Models, Animal
- Fatty Liver/genetics
- Fatty Liver/metabolism
- Foam Cells/cytology
- Foam Cells/physiology
- Glucose Intolerance/genetics
- Glucose Intolerance/metabolism
- Humans
- Inflammation/genetics
- Inflammation/metabolism
- Insulin/blood
- Liver/metabolism
- Macrophages, Peritoneal/cytology
- Macrophages, Peritoneal/physiology
- Metabolic Syndrome/genetics
- Metabolic Syndrome/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscle, Skeletal/metabolism
- Promoter Regions, Genetic/genetics
- Receptors, Adiponectin/genetics
- Receptors, Adiponectin/metabolism
- Serine-Arginine Splicing Factors
- Triglycerides/blood
Collapse
Affiliation(s)
- Nanlan Luo
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-3360
| | - B Hong Chung
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-3360
| | - Xiangdong Wang
- Institute of Cell Biology, Shandong University, Jinan, China 250012
| | - Richard L. Klein
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Chao-Ke Tang
- Institute of Cardiovascular Research, University of South China, Hengyang, China, 421001
| | - W. Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-3360
- Birmingham VA Medical Center, Birmingham, AL 35233
| | - Yuchang Fu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-3360
| |
Collapse
|
24
|
AdR1-TG/TALLYHO mice have improved lipid accumulation and insulin sensitivity. Biochem Biophys Res Commun 2013; 433:567-72. [PMID: 23523784 DOI: 10.1016/j.bbrc.2013.03.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 03/13/2013] [Indexed: 11/24/2022]
Abstract
BACKGROUND Overexpression of adiponectin receptor 1 in macrophages can physiologically modulate metabolic activities in vivo by enhancing adiponectin actions in distal metabolically active tissues. To investigate the effects of enhanced adiponectin actions in TALLYHO (TH) diabetic mouse model, we crossed the adiponectin receptor 1 macrophage-specific transgenic mice (AdR1-TG) with the TALLYHO diabetic mice (TH) to examine the changes of lipid accumulation and insulin sensitivity in these mice. METHODS AdR1-TG/TH and the control WT/TH mice were fed either normal diet or high fat diet for 28weeks. Whole body weights of these mice were measured and mouse sera were analyzed for the levels of cholesterol, triglyceride, and free fatty acids. Glucose tolerance testing (GTT) and insulin tolerance testing (ITT) in these mice were performed to investigate systemic insulin sensitivity in vivo. Molecular markers for insulin signaling pathway in mouse skeletal muscle tissues, IRS-1 and AKT, were examined. Mouse serum insulin levels were measured and Sirt1 gene expression in mouse pancreatic tissues was also quantified related to the insulin secretion. The Caspase 3 protein levels were analyzed by Western blot methods. RESULTS Compared to the control WT/TH mice, AdR1-TG/TH mice showed significantly lower body weights under either normal diet or high fat diet and the mouse serum levels of cholesterol, triglyceride and free fatty acids were significantly decreased in the transgenic crossed mice when compared to those from the control mice. Improved GTT and ITT tests indicating increased systemic insulin sensitivity in the transgenic crossed mice demonstrated the enhanced adiponectin actions on the systemic metabolism in vivo. The increases of insulin secretion and its related gene expression were also detected in the transgenic crossed mice. In contrast, the control mice showed hypertrophy pancreases companying with high apoptosis gene expression. These results suggest that enhanced adiponectin actions by overexpressing adiponectin receptor 1 in macrophages can provide unique interactions with the metabolic tissues/cells, improving lipid accumulation and insulin sensitivity in TALLYHO diabetic mice.
Collapse
|
25
|
Gemfibrozil and its combination with metformin on pleiotropic effect on IL-10 and adiponectin and anti-atherogenic treatment in insulin resistant type 2 diabetes mellitus rats. Inflammopharmacology 2012; 21:137-45. [DOI: 10.1007/s10787-012-0154-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Accepted: 10/08/2012] [Indexed: 11/25/2022]
|
26
|
Lin HL, Shen KP, Chang WT, Lin JC, An LM, Chen IJ, Wu BN. Eugenosedin-A prevents high-fat diet increased adhesion molecules through inhibition of MAPK- and p65-mediated NF-κB pathway in rat model. J Pharm Pharmacol 2012; 65:300-9. [DOI: 10.1111/j.2042-7158.2012.01597.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 08/30/2012] [Indexed: 12/23/2022]
Abstract
Abstract
Objectives
Previous studies have shown eugenosedin-A, a 5-HT1B/2A and α1/α2/β1-adrenergic blocker, is able to decrease cholesterol levels, hyperglycaemia and inflammation in hyperlipidaemic mice induced by high-fat diet (HFD). The aim of this study is to examine the effects of eugenosedin-A on the inhibition of adhesion molecules of platelets, the aorta and acyl-coenzymeA:cholesterol acyltransferase-1 (ACAT-1) of macrophages in a hyperlipidaemic rat model.
Methods
Six-week-old Sprague–Dawley rats were randomly divided into two control and treatment groups. The control rats received either a regular diet or HFD and the treatment groups were fed HFD with either 5 mg/kg eugenosedin-A or atorvastatin for a 10-week period.
Key findings
Compared with the two control groups, the HFD group had lower levels of high-density lipoprotein, higher concentrations of triglycerides, total cholesterol, low-density lipoprotein and insulin. The expression of adhesion molecules in platelets, aorta and monocyte-macrophage were enhanced by HFD. HFD also increased upstream proteins and their phosphorylated form in the aorta. In treatment groups, eugenosedin-A and atorvastatin improved HFD-induced hyperlipidaemia and levels of insulin. Eugenosedin-A reduced the upregulation of P-selectin, ICAM-1, ICAM-2, ICAM-3, VCAM, PECAM in platelets and inhibited E-selectin, ICAM-1, ICAM-2, ICAM-3, VCAM and PECAM protein levels in the aorta. Eugenosedin-A reduced the ACAT-1 protein expression of monocyte-macrophages. The expression of PKCα, MAPKs, IKKα and p65 and their phosphorylated form were reduced in treatment groups.
Conclusions
Taken together, hyperlipidaemia enhances the expression of adhesion molecules and ACAT-1 protein, and eugenosedin-A ameliorates those increases. Through inhibition of MAPK- and p-65-mediated NF-κB pathway, eugenosedin-A decreases the quantity of adhesion molecules.
Collapse
Affiliation(s)
- Hui-Li Lin
- Department of Food Science and Nutrition, Meiho University, Pingtung, Taiwan
| | - Kuo-Ping Shen
- Department of Nursing, Meiho University, Pingtung, Taiwan
| | - Wen-Tsan Chang
- Division of Hepatobiliarypancreatic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jou-Chun Lin
- Department of Ophthalmology, Kaohsiung Municipal United Hospital, Kaohsiung, Taiwan
| | - Li-Mei An
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ing-Jun Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bin-Nan Wu
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
27
|
Park YM, Kashyap S, Major J, Silverstein RL. Insulin promotes macrophage foam cell formation: potential implications in diabetes-related atherosclerosis. J Transl Med 2012; 92:1171-80. [PMID: 22525426 PMCID: PMC3407326 DOI: 10.1038/labinvest.2012.74] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The prevalence of atherosclerotic cardiovascular disease is higher in patients with type 2 diabetes, a disorder characterized by hyperinsulinemia and insulin resistance. The role of hyperinsulinemia as an independent participant in the atherogenic process has been controversial. In the current study, we tested the effect of insulin and the insulin sensitizer, adiponectin, on human macrophage foam cell formation. We found that both insulin and adiponectin increased the expression of the type 2 scavenger receptor CD36 by approximately twofold and decreased the expression of the ATP-binding cassette transporter ABCA1 by >80%. In both cases regulation was post-transcriptional. As a consequence of these changes, we found that oxidized LDL (oxLDL) uptake was increased by 80% and cholesterol efflux to apolipoprotein A1 (apoA1) was decreased by ∼25%. This led to two- to threefold more cholesterol accumulation over a 16-h period. As reported previously in studies of murine systems, scavenger receptor-A (SR-A) expression on human macrophages was downregulated by insulin and adiponectin. Insulin and adiponectin did not affect oxLDL-induced secretion of monocyte attractant protein-1 (MCP-1) and interleukin-6 (IL-6). These studies suggest that hyperinsulinemia could promote macrophage foam cell formation and thus may contribute to atherosclerosis in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Young Mi Park
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea,Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sangeeta Kashyap
- Department of Endocrinology, Cleveland Clinic, Cleveland, OH, USA
| | - Jennifer Major
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Roy L. Silverstein
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
28
|
Dalamaga M, Diakopoulos KN, Mantzoros CS. The role of adiponectin in cancer: a review of current evidence. Endocr Rev 2012; 33:547-94. [PMID: 22547160 PMCID: PMC3410224 DOI: 10.1210/er.2011-1015] [Citation(s) in RCA: 456] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Excess body weight is associated not only with an increased risk of type 2 diabetes and cardiovascular disease (CVD) but also with various types of malignancies. Adiponectin, the most abundant protein secreted by adipose tissue, exhibits insulin-sensitizing, antiinflammatory, antiatherogenic, proapoptotic, and antiproliferative properties. Circulating adiponectin levels, which are determined predominantly by genetic factors, diet, physical activity, and abdominal adiposity, are decreased in patients with diabetes, CVD, and several obesity-associated cancers. Also, adiponectin levels are inversely associated with the risk of developing diabetes, CVD, and several malignancies later in life. Many cancer cell lines express adiponectin receptors, and adiponectin in vitro limits cell proliferation and induces apoptosis. Recent in vitro studies demonstrate the antiangiogenic and tumor growth-limiting properties of adiponectin. Studies in both animals and humans have investigated adiponectin and adiponectin receptor regulation and expression in several cancers. Current evidence supports a role of adiponectin as a novel risk factor and potential diagnostic and prognostic biomarker in cancer. In addition, either adiponectin per se or medications that increase adiponectin levels or up-regulate signaling pathways downstream of adiponectin may prove to be useful anticancer agents. This review presents the role of adiponectin in carcinogenesis and cancer progression and examines the pathophysiological mechanisms that underlie the association between adiponectin and malignancy in the context of a dysfunctional adipose tissue in obesity. Understanding of these mechanisms may be important for the development of preventive and therapeutic strategies against obesity-associated malignancies.
Collapse
Affiliation(s)
- Maria Dalamaga
- Laboratory of Clinical Biochemistry, Attikon General University Hospital, University of Athens, School of Medicine, 12462 Athens, Greece
| | | | | |
Collapse
|
29
|
Abstract
UNLABELLED The global epidemic of obesity is accompanied by an increased prevalence of cardiovascular disease (CVD), in particular stroke and heart attack. Dysfunctional adipose tissue links obesity to CVD by secreting a multitude of bioactive lipids and pro-inflammatory factors (adipokines) with detrimental effects on the cardiovascular system. Adiponectin is one of the few adipokines that possesses multiple salutary effects on insulin sensitivity and cardiovascular health. Clinical investigations have identified adiponectin deficiency (hypoadiponectinaemia) as an independent risk factor for CVD. In animals, elevation of plasma adiponectin by either pharmacological or genetic approaches alleviates obesity-induced endothelial dysfunction and hypertension, and also prevents atherosclerosis, myocardial infarction and diabetic cardiomyopathy. Furthermore, many therapeutic benefits of the peroxisome-proliferator activated receptor gamma agonists, the thiazolidinediones, are mediated by induction of adiponectin. Adiponectin protects cardiovascular health through its vasodilator, anti-apoptotic, anti-inflammatory and anti-oxidative activities in both cardiac and vascular cells. This review summarizes recent findings in the understanding of the physiological role and clinical relevance of adiponectin in cardiovascular health, and in the identification of the receptor and postreceptor signalling events that mediate the cardiovascular actions of adiponectin. It also discusses adiponectin-targeted drug discovery strategies for treating obesity, diabetes and CVD. LINKED ARTICLES This article is part of a themed section on Fat and Vascular Responsiveness. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-3.
Collapse
Affiliation(s)
- Xiaoyan Hui
- Department of Medicine, the University of Hong Kong, Hong Kong
| | | | | | | |
Collapse
|
30
|
Watanabe T, Sato K, Itoh F, Wakabayashi K, Shichiri M, Hirano T. Endogenous bioactive peptides as potential biomarkers for atherosclerotic coronary heart disease. SENSORS 2012; 12:4974-85. [PMID: 22666071 PMCID: PMC3355454 DOI: 10.3390/s120404974] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/22/2012] [Accepted: 04/16/2012] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide, with high medical costs and rates of disability. It is therefore important to evaluate the use of cardiovascular biomarkers in the early diagnosis of coronary artery disease (CAD). We have screened a variety of recently identified bioactive peptides candidates in anticipation that they would allow detection of atherosclerotic CAD. Especially, we have focused on novel anti-atherogenic peptides as indicators and negative risk factors for CAD. In vitro, in vivo and clinical studies indicated that human adiponectin, heregulin-β1, glucagon-like peptide-1 (GLP-1), and salusin-α, peptides of 244, 71, 30, and 28 amino acids, respectively, attenuate the development and progression of atherosclerotic lesions by suppressing macrophage foam cell formation via down-regulation of acyl-coenzyme A: cholesterol acyltransferase-1. Circulating levels of these peptides in the blood are significantly decreased in patients with CAD compared to patients without CAD. Receiver operating characteristic analyses showed that salusin-α is a more useful biomarker, with better sensitivity and specificity, compared with the others for detecting CAD. Therefore, salusin-α, heregulin-β1, adiponectin, and/or GLP-1, alone or in various combinations, may be useful as biomarkers for atherosclerotic CAD.
Collapse
Affiliation(s)
- Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan; E-Mails: (K.S.); (F.I.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-42-676-6983; Fax: +81-42-676-4323
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan; E-Mails: (K.S.); (F.I.)
| | - Fumiko Itoh
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan; E-Mails: (K.S.); (F.I.)
| | - Kohei Wakabayashi
- Division of Cardiology, Showa University Fujigaoka Hospital, Yokohama, Kanagawa 227-8501, Japan; E-Mail:
| | - Masayoshi Shichiri
- Department of Endocrinology, Diabetes and Metabolism, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan; E-Mail:
| | - Tsutomu Hirano
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8555, Japan; E-Mail:
| |
Collapse
|
31
|
Oliveira CSV, Saddi-Rosa P, Crispim F, Canani LH, Gerchman F, Giuffrida FMA, Vieira JGH, Velho G, Reis AF. Association of ADIPOQ variants, total and high molecular weight adiponectin levels with coronary artery disease in diabetic and non-diabetic Brazilian subjects. J Diabetes Complications 2012; 26:94-8. [PMID: 22459242 DOI: 10.1016/j.jdiacomp.2012.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 01/20/2012] [Accepted: 02/24/2012] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To investigate the association of ADIPOQ variants, total and high molecular weight adiponectin (HMW) adiponectin levels with the prevalence of diabetes mellitus and coronary artery disease (CAD) diagnosed by coronary angiography in Brazilian subjects with high cardiovascular risk. METHODS 603 subjects undergoing coronary angiography were studied in regard to their glycemic status and presence of CAD (lesions >0%). We evaluated baseline concentrations of total and HMW adiponectin and three ADIPOQ variants: -11391G>A (rs17300539), +45T>G (rs2241766) and+276G>T (rs1501299). RESULTS The G-allele of rs2241766 was associated with higher levels of total and HMW adiponectin, and the A-allele of rs17300539 was associated with higher levels of HMW adiponectin. Lower levels of total and HMW adiponectin were independently associated with CAD. The G-allele of rs2241766 (OR 2.45, 95% C.I. 1.05-6.04, p=0.04) and the G-allele of rs1501299 (OR 1.89, 95% C.I. 1.04-3.45, p=0.03) were associated with CAD, and these associations were independent of circulating levels of adiponectin. CONCLUSIONS In Brazilian subjects with high cardiovascular risk, CAD was associated with lower total and HMW adiponectin levels. The rs2241766 and rs1501299 polymorphisms were associated with CAD. The rs2241766 variant was associated with total and HMW adiponectin levels, while rs17300539 was associated with HMW adiponectin levels.
Collapse
|
32
|
Tian L, Luo N, Zhu X, Chung BH, Garvey WT, Fu Y. Adiponectin-AdipoR1/2-APPL1 signaling axis suppresses human foam cell formation: differential ability of AdipoR1 and AdipoR2 to regulate inflammatory cytokine responses. Atherosclerosis 2011; 221:66-75. [PMID: 22227293 DOI: 10.1016/j.atherosclerosis.2011.12.014] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 12/13/2011] [Accepted: 12/14/2011] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Adiponectin is an adipokine that exerts anti-inflammatory and anti-atherogenic effects during macrophage transformation into foam cells. To further understand the signaling pathways of adiponectin involved in macrophage foam cell transformation, we investigated the roles of two adiponectin receptors (AdipoR1 and AdipoR2) and their downstream adaptor protein, phosphotyrosine interaction, PH domain and leucine zipper containing 1 (APPL1) in mediating adiponectin action on foam cell transformation. METHODS AND RESULTS Transfections were performed to overexpress or knockdown AdipoR1 or AdipoR2 genes in human THP-1 monocytes. Lentiviral-shRNAs were also used to knockdown APPL1 gene in these cells. Foam cell transformation was induced via exposure to oxidized low-density lipoprotein (oxLDL). Our results showed that both AdipoR1 and AdipoR2 were critical for transducing the adiponectin signal that suppresses lipid accumulation and inhibits transformation from macrophage to foam cell. However, AdipoR1 and AdipoR2 were found to have differential effects in diminishing proinflammatory responses. While AdipoR1 was required by adiponectin to suppress tumor necrosis factor alpha (TNFα) and monocyte chemotactic protein 1 (MCP-1) gene expression, AdipoR2 served as the dominant receptor for adiponectin suppression of scavenger receptor A type 1 (SR-AI) and upregulation of interleukin-1 receptor antagonist (IL-1Ra). Knockdown of APPL1 significantly abrogated the ability of adiponectin to inhibit lipid accumulation, SR-AI and nuclear factor-κB (NF-κB) gene expression, and Akt phosphorylation in macrophage foam cells. CONCLUSIONS In current studies, we have demonstrated that adiponectin's abilty to suppress macrophage lipid accumulation and foam cell formation is mediated through AdipoR1 and AdipoR2 and the APPL1 docking protein. However, AdipoR1 and AdipoR2 exhibited a differential ability to regulate inflammatory cytokines and SR-A1. These novel data support the idea that the adiponectin-AdipoR1/2-APPL1 axis may serve as a potential therapeutic target for preventing macrophage foam cell formation and atherosclerosis.
Collapse
Affiliation(s)
- Ling Tian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-3360, United States
| | | | | | | | | | | |
Collapse
|
33
|
Luo N, Wang X, Chung BH, Lee MH, Klein RL, Garvey WT, Fu Y. Effects of macrophage-specific adiponectin expression on lipid metabolism in vivo. Am J Physiol Endocrinol Metab 2011; 301:E180-6. [PMID: 21505149 PMCID: PMC3129839 DOI: 10.1152/ajpendo.00614.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epidemiological studies have associated low circulating levels of the adipokine adiponectin with multiple metabolic disorders, including metabolic syndrome, obesity, insulin resistance, type II diabetes, and cardiovascular disease. Recently, we reported that adiponectin selectively overexpressed in mouse macrophages can improve insulin sensitivity and protect against inflammation and atherosclerosis. To further investigate the role of adiponectin and macrophages on lipid and lipometabolism in vivo, we engineered the expression of adiponectin in mouse macrophages (Ad-TG mice) and examined effects on plasma lipoproteins and on the expression levels of genes involved in lipoprotein metabolism in tissues. Compared with the wild-type (WT) mice, Ad-TG mice exhibited significantly lower levels of plasma total cholesterol (-21%, P < 0.05) due to significantly decreased LDL (-34%, P < 0.05) and VLDL (-32%, P < 0.05) cholesterol concentrations together with a significant increase in HDL cholesterol (+41%, P < 0.05). Further studies investigating potential mechanisms responsible for the change in lipoprotein cholesterol profile revealed that adiponectin-producing macrophages altered expression of key genes in liver tissue, including apoA1, apoB, apoE, the LDL receptor, (P < 0.05), and ATP-binding cassette G1 (P < 0.01). In addition, Ad-TG mice also exhibited higher total and high-molecular-weight adipnection levels in plasma and increased expression of the anti-inflammatory cytokine IL-10 as well as a decrease in the proinflammatory cytokine IL-6 in adipose tissue. These results indicate that macrophages engineered to produce adiponectin can influence in vivo gene expression in adipose tissue in a manner that reduces inflammation and macrophage infiltration and in liver tissue in a manner that alters the circulating lipoprotein profile, resulting in a decrease in VLDL and LDL and an increase in HDL cholesterol. The data support further study addressing the use of genetically manipulated macrophages as a novel therapeutic approach for treatment of cardiometabolic disease.
Collapse
Affiliation(s)
- Nanlan Luo
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama 35294-0012, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Meilleur KG, Doumatey A, Huang H, Charles B, Chen G, Zhou J, Shriner D, Adeyemo A, Rotimi C. Circulating adiponectin is associated with obesity and serum lipids in West Africans. J Clin Endocrinol Metab 2010; 95:3517-21. [PMID: 20382687 PMCID: PMC2928908 DOI: 10.1210/jc.2009-2765] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Adiponectin, a hormone secreted by adipose tissue, has both metabolic and antiinflammatory properties. Although multiple studies have described the relationship between adiponectin and obesity in several human populations, no large studies have evaluated this relationship in Africans. OBJECTIVE We investigated the relationship between adiponectin and measures of obesity, serum lipids, and insulin resistance in a large African cohort. DESIGN Participants are from the Africa America Diabetes Mellitus (AADM) Study, a case-control study of genetic and other risk factors associated with development of type 2 diabetes in Africans. SETTING Patients were recruited from five academic medical centers in Nigeria and Ghana (Accra and Kumasi in Ghana and Enugu, Ibadan, and Lagos in Nigeria) over 10 yr. MAIN OUTCOME MEASURES Circulating adiponectin levels were measured in 690 nondiabetic controls using an ELISA. The correlation between log-transformed circulating adiponectin levels and age, gender, measures of obesity (body mass index, waist circumference, and percent fat mass), and serum lipid levels was assessed. Linear regression was used to explore the association between adiponectin levels and measures of obesity, lipids, and insulin resistance as measured by homeostasis model assessment. RESULTS Significant negative associations were observed between log-adiponectin levels and measures of obesity after adjusting for age and gender. Similarly, log-adiponectin levels were significantly negatively associated with serum triglycerides and insulin resistance but positively associated with high-density lipoprotein-cholesterol and total cholesterol after adjusting for age, gender, and body mass index. CONCLUSIONS Circulating adiponectin is significantly associated with measures of obesity, serum lipids, and insulin resistance in this study of West African populations.
Collapse
Affiliation(s)
- Katherine G Meilleur
- The Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Luo N, Liu J, Chung BH, Yang Q, Klein RL, Garvey WT, Fu Y. Macrophage adiponectin expression improves insulin sensitivity and protects against inflammation and atherosclerosis. Diabetes 2010; 59:791-9. [PMID: 20350970 PMCID: PMC2844826 DOI: 10.2337/db09-1338] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Adiponectin is one of several important metabolically active cytokines secreted from adipose tissue. Epidemiologic studies have associated low-circulating levels of this adipokine with multiple metabolic disorders including obesity, insulin resistance, type 2 diabetes, and cardiovascular disease. To investigate adiponectin-mediated changes in metabolism in vivo, we generated transgenic mice that specifically express the gene coding for human adiponectin in mouse macrophages using the human scavenger receptor A-I gene enhancer/promoter. METHODS AND RESULTS Using this transgenic mouse model, we found that adiponectin expression was associated with reduced whole-animal body and fat-pad weight and an improved lipid accumulation in macrophages when these transgenic mice were fed with a high-fat diet. Moreover, these macrophage Ad-TG mice exhibit enhanced whole-body glucose tolerance and insulin sensitivity with reduced proinflammatory cytokines, MCP-1 and TNF-a (both in the serum and in the metabolic active macrophage), adipose tissue, and skeletal muscle under the high-fat diet condition. Additional studies demonstrated that these macrophage adiponectin transgenic animals exhibit reduced macrophage foam cell formation in the arterial wall when these transgenic mice were crossed with an LDL receptor-deficient mouse model and were fed a high-fat diet. CONCLUSIONS These results suggest that adiponectin expressed in macrophages can physiologically modulate metabolic activities in vivo by improving metabolism in distal tissues. The use of macrophages as carriers for adiponectin, a molecule with antidiabetes, anti-inflammatory, and antiatherogenic properties, provides a novel and unique strategy for studying the mechanisms of adiponectin-mediated alterations in body metabolism in vivo.
Collapse
Affiliation(s)
- Nanlan Luo
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jian Liu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - B. Hong Chung
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Qinglin Yang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Richard L. Klein
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, and the Ralph H. Johnson Department of Veteran Affairs Medical Center, Charleston, South Carolina
| | - W. Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Yuchang Fu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
- Corresponding author: Yuchang Fu,
| |
Collapse
|
36
|
Antiatherosclerotic and anti-insulin resistance effects of adiponectin: basic and clinical studies. Prog Cardiovasc Dis 2009; 52:126-40. [PMID: 19732605 DOI: 10.1016/j.pcad.2009.06.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Adiponectin is a protein secreted by adipose cells that may couple regulation of insulin sensitivity with energy metabolism and serve to link obesity with insulin resistance. Obesity-related disorders characterized by insulin resistance including the metabolic syndrome, diabetes, atherosclerosis, hypertension, and coronary artery disease are associated with both decreased adiponectin levels and endothelial dysfunction. Recent studies demonstrate that adiponectin has insulin-sensitizing effects as well as antiatherogenic properties. Lifestyle modifications and some drug therapies to treat atherosclerosis, hypertension, diabetes, and coronary heart disease have important effects in increasing adiponectin levels, decreasing insulin resistance, and improving endothelial dysfunction. In this review, we discuss insights into the relationships between adiponectin levels, insulin resistance, and endothelial dysfunction that are derived from various therapeutic interventions. The effects of lifestyle modifications and cardiovascular drugs on adiponectin levels and insulin resistance suggest plausible mechanisms that may be important for understanding and treating atherosclerosis and coronary heart disease.
Collapse
|
37
|
Hongo S, Watanabe T, Arita S, Kanome T, Kageyama H, Shioda S, Miyazaki A. Leptin modulates ACAT1 expression and cholesterol efflux from human macrophages. Am J Physiol Endocrinol Metab 2009; 297:E474-82. [PMID: 19625677 DOI: 10.1152/ajpendo.90369.2008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Leptin is an adipose tissue-derived hormone implicated in atherosclerosis and macrophage foam cell formation. The current study was conducted to examine the effect of leptin on cholesteryl ester accumulation in human monocytes/macrophages. Exogenously added leptin at 5 nM during differentiation of monocytes into macrophages for 7 days accelerated acetylated LDL (acetyl-LDL)-induced cholesteryl ester accumulation by 30-50%. Leptin did not affect endocytic uptake of acetyl-LDL; however, it increased ACAT activity 1.8-fold and ACAT-1 protein expression 1.9-fold. Among the four ACAT-1 mRNA transcripts, two shorter transcripts (2.8 and 3.6 kb) were upregulated approximately 1.7-fold upon leptin treatment. The enhanced expression of ACAT-1 protein by leptin was suppressed by inhibitors of Janus-activated kinase2 (JAK2) and phosphatidylinositol 3-kinase (PI3K). HDL-mediated cholesterol efflux was suppressed by leptin, which was canceled by K-604, an ACAT-1 inhibitor. Expression of long form of leptin receptor was upregulated during monocytic differentiation into macrophages and sustained after differentiation. Thus, the results suggest that leptin accelerates cholesteryl ester accumulation in human monocyte-derived macrophages by increasing ACAT-1 expression via JAK2 and PI3K, thereby suppressing cholesterol efflux.
Collapse
Affiliation(s)
- Shigeki Hongo
- Department of Biochemistry, Showa University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The enzymes acyl-coenzyme A (CoA):cholesterol acyltransferases (ACATs) are membrane-bound proteins that utilize long-chain fatty acyl-CoA and cholesterol as substrates to form cholesteryl esters. In mammals, two isoenzymes, ACAT1 and ACAT2, encoded by two different genes, exist. ACATs play important roles in cellular cholesterol homeostasis in various tissues. This chapter summarizes the current knowledge on ACAT-related research in two areas: 1) ACAT genes and proteins and 2) ACAT enzymes as drug targets for atherosclerosis and for Alzheimer's disease.
Collapse
Affiliation(s)
- Ta-Yuan Chang
- Department of Biochemistry, Dartmouth Medical School, 1 Rope Ferry Rd., Hanover, NH 03755-1404, USA.
| | | | | | | |
Collapse
|
39
|
Lei L, Xiong Y, Chen J, Yang JB, Wang Y, Yang XY, Chang CCY, Song BL, Chang TY, Li BL. TNF-alpha stimulates the ACAT1 expression in differentiating monocytes to promote the CE-laden cell formation. J Lipid Res 2009; 50:1057-67. [PMID: 19189937 DOI: 10.1194/jlr.m800484-jlr200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
High levels of the inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) are present in atherosclerotic lesions. TNF-alpha regulates expression of multiple genes involved in various stages of atherosclerosis, and it exhibits proatherosclerotic and antiatherosclerotic properties. ACAT catalyzes the formation of cholesteryl esters (CE) in monocytes/macrophages, and it promotes the foam cell formation at the early stage of atherosclerosis. We hypothesize that TNF-alpha may be involved in regulating the ACAT gene expression in monocytes/macrophages. In this article, we show that in cultured, differentiating human monocytes, TNF-alpha enhances the expression of the ACAT1 but not ACAT2 gene, increases the cholesteryl ester accumulation, and promotes the lipid-laden cell formation. Several other proinflammatory cytokines tested do not affect the ACAT1 gene expression. The stimulation effect is consistent with a receptor-dependent process, and is blocked by using nuclear factor-kappa B (NF-kappa B) inhibitors. A functional and unique NF-kappa B element located within the human ACAT1 gene proximal promoter is required to mediate the action of TNF-alpha. Our data demonstrate that TNF-alpha, through the NF-kappa B pathway, specifically enhances the expression of human ACAT1 gene to promote the CE-laden cell formation from the differentiating monocytes, and our data support the hypothesis that TNF-alpha is proatherosclerotic during early phase of lesion development.
Collapse
Affiliation(s)
- Lei Lei
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kanome T, Watanabe T, Nishio K, Takahashi K, Hongo S, Miyazaki A. Angiotensin II upregulates acyl-CoA:cholesterol acyltransferase-1 via the angiotensin II Type 1 receptor in human monocyte-macrophages. Hypertens Res 2009; 31:1801-10. [PMID: 18971559 DOI: 10.1291/hypres.31.1801] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Angiotensin II (Ang II) is known to accelerate the progression of macrophage-driven atherosclerotic lesions. Acyl-CoA:cholesterol acyltransferase-1 (ACAT1) converts intracellular free cholesterol into cholesterol ester (CE) for storage in lipid droplets, and promotes foam cell formation in atherosclerotic lesions. The present study explored the effect of Ang II on ACAT1 expression as a molecular mechanism of foam cell formation in primary cultured human monocyte-macrophages. Ang II significantly increased ACAT1 protein expression in a time- or concentration-dependent manner. Application of an Ang II type 1 (AT(1)) receptor agonist (L162313), but not an Ang II type 2 (AT(2)) receptor agonist (CGP42112A), mimicked the effects of Ang II treatment in inducing ACAT1 protein expression. ACAT activity and ACAT1 mRNA levels were also significantly increased by Ang II. Two-fold increases in ACAT1 protein expression and ACAT activity with Ang II treatment were completely inhibited by AT(1) receptor antagonists (candesartan, [Sar(1),Ile(8)]-Ang II), but not by an AT(2) receptor antagonist (PD123319). Treatment with a G-protein inactivator (GDP-beta-S), a c-Src tyrosine kinase inhibitor (PP2), a protein kinase C (PKC) inhibitor (rottlerin), or a mitogen activated protein kinase (MAPK) kinase inhibitor (PD98059) significantly reduced Ang II-induced ACAT1 protein expression. Macrophage foam cell formation assessed using acetylated low-density lipoprotein (LDL)-induced CE accumulation was significantly enhanced by Ang II, which was completely inhibited by treatment with candesartan. These results suggested that Ang II enhances foam cell formation by upregulating ACAT1 expression predominantly through the actions of AT(1) receptor via the G protein/c-Src/PKC/MAPK pathway in human monocyte-macrophages.
Collapse
Affiliation(s)
- Tomoko Kanome
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Ferrarezi DAF, Cheurfa N, Reis AF, Fumeron F, Velho G. Adiponectin gene and cardiovascular risk in type 2 diabetic patients: a review of evidences. ACTA ACUST UNITED AC 2008; 51:153-9. [PMID: 17505621 DOI: 10.1590/s0004-27302007000200003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Accepted: 01/04/2007] [Indexed: 12/11/2022]
Abstract
Diabetic patients have a 3-fold higher risk of developing atherosclerosis and its clinical complications as compared to non-diabetic individuals. Part of the cardiovascular risk associated with diabetes is probably due to genetic determinants influencing both glucose homeostasis and the development of atherosclerosis. However, type 2 diabetes frequently coexists with other cardiovascular risk factors like arterial hypertension, central obesity and dyslipidemia. Genetic variability affecting many areas such as lipid and energy metabolisms, hypertension and haemodynamic mechanisms, blood clotting homeostasis, inflammation, and matrix turnover in the vascular wall will have an impact on the development of macrovascular complications in diabetic patients. Adiponectin is abundantly secreted by adipocytes. It plays important roles in lipid and glucose metabolisms and has direct anti-inflammatory and anti-atherogenic effects. In this review, we summarize recent data from the literature suggesting an implication of allelic variations of the adiponectin gene (ADIPOQ) in the genetic determinants of cardiovascular disease in diabetic subjects.
Collapse
Affiliation(s)
- Daniela A F Ferrarezi
- Laboratory of Cellular and Molecular Endocrinology, São Paulo University, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
42
|
Cheng M, Hashmi S, Mao X, Zeng QT. Relationships of adiponectin and matrix metalloproteinase-9 to tissue inhibitor of metalloproteinase-1 ratio with coronary plaque morphology in patients with acute coronary syndrome. Can J Cardiol 2008; 24:385-90. [PMID: 18464944 DOI: 10.1016/s0828-282x(08)70602-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
OBJECTIVES Adiponectin, an adipocyte-specific protein, matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) play a crucial role in arteriosclerosis and plaque disruption. The present study was designed to elucidate the relationship of adiponectin and the ratio of MMP-9/TIMP-1 and their effects on the stability of plaque in acute coronary syndrome (ACS). METHODS The concentrations of adiponectin, MMP-9, TIMP-1 and interleukin-10 were analyzed using ELISA in 56 consecutive unselected patients divided into two groups, stable angina (n=13) and ACS (n=43), and were compared with 19 healthy control subjects. The 56 patients were also angiographically studied and divided into two groups, simple lesion (n=22) and complex lesion (n=34), based on coronary plaque morphology. RESULTS The ratio of MMP-9/TIMP-1 showed significantly higher values in the ACS group compared with the control group (0.22+/-0.10 versus 0.11+/-0.03; P<0.001). Adiponectin was negatively correlated with the ratio of MMP-9/TIMP-1 (r=--0.332; P=0.008) and positively correlated with interleukin-10 (r=0.651; P=0.001). Multivariate logistic regression analysis showed that adiponectin (P=0.046) and MMP-9/TIMP-1 (P=0.044) are independent predictors for ACS, and MMP-9/TIMP-1 (P=0.013) is an independent predictor for complex lesion morphology plaques. CONCLUSION In the present study, it was found that adiponectin has a negative relationship with the ratio of MMP-9/TIMP-1 in patients with ACS, and that the ratio of MMP-9/TIMP-1 is an independent predictor of the stability of atherosclerotic plaque and the severity of coronary atherosclerosis.
Collapse
Affiliation(s)
- Min Cheng
- Department of Interventional Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | |
Collapse
|
43
|
Tian L, Luo N, Klein RL, Chung BH, Garvey WT, Fu Y. Adiponectin reduces lipid accumulation in macrophage foam cells. Atherosclerosis 2008; 202:152-61. [PMID: 18511057 DOI: 10.1016/j.atherosclerosis.2008.04.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Revised: 03/31/2008] [Accepted: 04/02/2008] [Indexed: 01/23/2023]
Abstract
Adiponectin is one of several, important metabolically active cytokines secreted from adipocytes. Low circulating levels of this adipokine have been associated epidemiologically with obesity, insulin resistance, type II diabetes, and cardiovascular disease. To determine if adiponectin can modulate lipid metabolism in macrophages, we expressed the adiponectin gene in human THP-1 macrophage foam cells using a lentiviral vector expression system and demonstrated that macrophages transduced with the adiponectin gene had decreased lipid accumulation compared with control macrophages transduced with the LacZ gene. Macrophages transduced with the adiponectin gene also exhibited decreased oxidized low-density lipoprotein (oxLDL) uptake and increased HDL-mediated cholesterol efflux. Additional studies suggest two potential mechanisms for the reduced lipid accumulation in these adiponectin-transduced macrophage foam cells. The first mechanism involves the PPARgamma and LXR signaling pathways which up-regulate the expression of ABCA1 and promote lipid efflux from these cells. The second mechanism involves decreased lipid uptake and increased lipid hydrolysis which may result from decreased SR-AI and increased SR-BI and HSL gene activities in the transformed macrophage foam cells. We also demonstrated that the expression of two proatherogenic cytokines, MCP-1 and TNFalpha, were decreased in the adiponectin-transduced macrophage foam cells. These results suggest that adiponectin may modulate multiple pathways of lipid metabolism in macrophages. Our studies provide new insights into potential mechanisms of adiponectin-mediated alterations in lipid metabolism and macrophage foam cell formation which may impact the development of atherosclerosis.
Collapse
Affiliation(s)
- Ling Tian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-3360, United States
| | | | | | | | | | | |
Collapse
|
44
|
Watanabe T, Nishio K, Kanome T, Matsuyama TA, Koba S, Sakai T, Sato K, Hongo S, Nose K, Ota H, Kobayashi Y, Katagiri T, Shichiri M, Miyazaki A. Impact of Salusin-α and -β on Human Macrophage Foam Cell Formation and Coronary Atherosclerosis. Circulation 2008; 117:638-48. [DOI: 10.1161/circulationaha.107.712539] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Takuya Watanabe
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Kae Nishio
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Tomoko Kanome
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Taka-aki Matsuyama
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Shinji Koba
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Tetsuo Sakai
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Kengo Sato
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Shigeki Hongo
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Kiyoshi Nose
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Hidekazu Ota
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Youichi Kobayashi
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Takashi Katagiri
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Masayoshi Shichiri
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| | - Akira Miyazaki
- From the Department of Biochemistry (T.W., K. Nishio, T. Kanome, S.H., A.M.), Second Department of Pathology (T.M., H.O.), and Third Department of Internal Medicine (S.K., T.S., Y.K., T. Katagiri), Showa University School of Medicine, Tokyo, Japan; Department of Microbiology, Showa University School of Pharmaceutical Sciences (K. Nose), Tokyo, Japan; and Tokyo Medical and Dental University (K.S., M.S.), Tokyo, Japan
| |
Collapse
|
45
|
Cai XJ, Li CJ, Chen L, Rong YY, Zhang Y, Zhang M. A hypothesis: adiponectin mediates anti-atherosclerosis via adventitia-AMPK-iNOS pathway. Med Hypotheses 2007; 70:1044-7. [PMID: 17910991 DOI: 10.1016/j.mehy.2007.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 08/14/2007] [Indexed: 11/23/2022]
Abstract
Adiponectin is an adipocyte-derived protein with insulin-sensitizing, anti-inflammatory, and anti-atherogenic properties and is abundantly found in plasma. Vascular adventitia is the outermost connective and supporting tissue of vessels. Recently, increasing evidence has shown that infection in the adventitia is one of the causes of atherosclerosis and restenosis. Our previous study indicated that local transferring adenovirus expressing adiponectin gene (Ad-APN) to intima and adventitia can suppress atherosclerosis, but the exact mechanism is still obscure. We speculate that with infection in the adventitia, adiponectin can activate AMP-activated protein kinase (AMPK) through adiponectin receptors in the membranes of adventitial fibroblasts and then inhibit the expression and activity of inducible nitric oxide synthase (iNOS); secretion of adventitial infective factors; division, proliferation and translation of adventitial fibroblasts; and change of adventitial fibroblasts to myofibroblasts, finally decreasing oxidative/nitrative stress to reduce atherosclerotic plaque area and stabilize atherosclerotic plaques. The proposition may provide clues into the development of a novel treatment for atherosclerosis.
Collapse
Affiliation(s)
- Xiao-jun Cai
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, Shandong 250012, China
| | | | | | | | | | | |
Collapse
|
46
|
Rae C, Robertson SA, Taylor JMW, Graham A. Resistin induces lipolysis and re-esterification of triacylglycerol stores, and increases cholesteryl ester deposition, in human macrophages. FEBS Lett 2007; 581:4877-83. [PMID: 17889853 DOI: 10.1016/j.febslet.2007.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Revised: 08/28/2007] [Accepted: 09/09/2007] [Indexed: 11/23/2022]
Abstract
Human resistin, found within atheroma, exerts inflammatory, angiogenic and proliferative effects in vascular cells and may predict coronary events. Here, we investigate mechanisms by which resistin contributes to macrophage 'foam cell' formation. Increases in macrophage (THP-1) cholesteryl ester mass, in the presence or absence of oxidized LDL, were not explained by altered cholesterol efflux. Instead, resistin enhanced fractional turnover of the endogenous triacylglycerol pool, increased uptake and decreased oxidation of exogenous fatty acids, and decreased phosphorylation of acetyl CoA carboxylase, all factors increasing the availability of fatty acyl CoA substrate for acyl CoA: cholesterol acyltransferase-1, thereby enhancing macrophage cholesteryl ester deposition.
Collapse
Affiliation(s)
- Colin Rae
- Vascular Biology Group, Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Cowcaddens Road, Glasgow G4 0BA, United Kingdom
| | | | | | | |
Collapse
|
47
|
Inoue T, Kotooka N, Morooka T, Komoda H, Uchida T, Aso Y, Inukai T, Okuno T, Node K. High molecular weight adiponectin as a predictor of long-term clinical outcome in patients with coronary artery disease. Am J Cardiol 2007; 100:569-74. [PMID: 17697807 DOI: 10.1016/j.amjcard.2007.03.062] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Revised: 03/12/2007] [Accepted: 03/12/2007] [Indexed: 11/19/2022]
Abstract
Adiponectin is an adipocyte-specific secretory protein that is highly and specifically expressed in adipose tissue, and low plasma levels of adiponectin are associated with coronary artery disease (CAD). It has been suggested that high molecular weight (HMW) adiponectin is more important for vascular protection than total amount of adiponectin. To establish the clinical relevance of HMW adiponectin, we measured its serum levels in 149 patients with CAD. The levels were lower in vasospastic angina pectoris (3.4 +/- 2.4 microg/ml, p <0.01), stable angina pectoris (3.3 +/- 2.6 microg/ml, p <0.001), and healed myocardial infarction (3.8 +/- 2.9 microg/ml, p <0.01) than chest pain syndrome (controls) (6.6 +/- 5.4 microg/ml). The levels were also lower in multivessel CAD (3.4 +/- 2.4 microg/dl) compared with single vessel CAD (4.2 +/- 2.7 microg/ml, p <0.05) or no organic stenosis (5.1 +/- 3.5 microg/ml, p <0.01). In univariate analysis, diabetes mellitus (p = 0.03), insulin resistance (p = 0.06), high-sensitivity C-reactive protein levels (p = 0.0012), and low HMW adiponectin levels (p = 0.0001) predicted cardiovascular events during 7 years of follow-up. However, multivariate analysis showed that only HMW adiponectin levels were an independent predictor of cardiovascular events (relative risk 2.79, 95% confidence interval 1.49 to 5.24, p = 0.0014). In conclusion, serum HMW adiponectin levels may serve as a predictor of future cardiovascular events in patients with CAD as well as a marker for severity of CAD.
Collapse
Affiliation(s)
- Teruo Inoue
- Department of Cardiovascular and Renal Medicine, Saga University Faculty of Medicine, Saga, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Katagiri H, Yamada T, Oka Y. Adiposity and cardiovascular disorders: disturbance of the regulatory system consisting of humoral and neuronal signals. Circ Res 2007; 101:27-39. [PMID: 17615379 DOI: 10.1161/circresaha.107.151621] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Obesity, a major healthcare issue, is associated with significant cardiovascular morbidities, including hypertension and atherosclerosis. Numerous intensive studies conducted this decade have revealed that adipose tissue is a major endocrine organ that secretes a variety of bioactive substances, termed adipocytokines. Adipocytokine secretion profiles are altered as obesity develops, which may increase the risk of obesity-related cardiovascular disorders. For instance, leptin is upregulated in obese subjects and plays important roles in the pathophysiology of obesity-related atherogenesis through multiple mechanisms, such as its proliferative, proinflammatory, prothrombotic, and prooxidant actions. In contrast, adiponectin, which is downregulated in obese subjects, has protective effects against cardiovascular disorders at various atherogenic stages. In addition to these factors secreted by adipose tissue, neuronal circuits involving autonomic nerves are now being recognized as an important metabolic regulatory system and have thus attracted considerable attentions. Alterations in fat accumulation in intraabdominal organs, such as visceral adipose tissue and the liver, send afferent neuronal signals to the brain, leading to modulation of sympathetic tonus and thereby affecting the vasculature. Moreover, these humoral and neuronal signaling pathways communicate with each other, resulting in cooperative metabolic regulation among tissues/organs throughout the body. Further elucidation of these regulatory systems is anticipated to lead to new approaches to devising therapeutic strategies for the metabolic syndrome.
Collapse
Affiliation(s)
- Hideki Katagiri
- Division of Advanced Therapeutics for Metabolic Diseases, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | |
Collapse
|
49
|
Lara-Castro C, Fu Y, Chung BH, Garvey WT. Adiponectin and the metabolic syndrome: mechanisms mediating risk for metabolic and cardiovascular disease. Curr Opin Lipidol 2007; 18:263-70. [PMID: 17495599 DOI: 10.1097/mol.0b013e32814a645f] [Citation(s) in RCA: 221] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Adiponectin is secreted exclusively by adipocytes, aggregates in multimeric forms, and circulates at high concentrations in blood. This review summarizes recent studies highlighting cellular effects of adiponectin and its role in human lipid metabolism and atherosclerosis. RECENT FINDINGS Adiponectin is an important autocrine/paracrine factor in adipose tissue that modulates differentiation of preadipocytes and favors formation of mature adipocytes. It also functions as an endocrine factor, influencing whole-body metabolism via effects on target organs. Adiponectin multimers exert differential biologic effects, with the high-molecular-weight multimer associated with favorable metabolic effects (i.e. greater insulin sensitivity, reduced visceral adipose mass, reduced plasma triglycerides, and increased HDL-cholesterol). Adiponectin influences plasma lipoprotein levels by altering the levels and activity of key enzymes (lipoprotein lipase and hepatic lipase) responsible for the catabolism of triglyceride-rich lipoproteins and HDL. It thus influences atherosclerosis by affecting the balance of atherogenic and antiatherogenic lipoproteins in plasma, and by modulating cellular processes involved in foam cell formation. SUMMARY Recent studies emphasize the role played by adiponectin in the homeostasis of adipose tissue and in the pathogenesis of the metabolic syndrome, type 2 diabetes, and atherosclerosis. These pleiotropic effects make it an attractive therapeutic target for obesity-related conditions.
Collapse
Affiliation(s)
- Cristina Lara-Castro
- Department of Nutrition Sciences, University of Alabama at Birmingham, 35294, USA
| | | | | | | |
Collapse
|
50
|
Park PH, McMullen MR, Huang H, Thakur V, Nagy LE. Short-term treatment of RAW264.7 macrophages with adiponectin increases tumor necrosis factor-alpha (TNF-alpha) expression via ERK1/2 activation and Egr-1 expression: role of TNF-alpha in adiponectin-stimulated interleukin-10 production. J Biol Chem 2007; 282:21695-703. [PMID: 17537727 PMCID: PMC1978175 DOI: 10.1074/jbc.m701419200] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Adiponectin is an adipokine with potent anti-inflammatory properties. However, the mechanisms by which adiponectin suppresses macrophage function are not well understood. Treatment of RAW264.7 macrophages with adiponectin for 18 h decreased lipopolysaccharide (LPS)-stimulated tumor necrosis factor-alpha (TNF-alpha) production. Here we demonstrate that globular adiponectin (gAcrp) initially increased TNF-alpha expression in RAW264.7 macrophages; this TNF-alpha then contributed to increased expression of interleukin-10, which in turn was required for the development of tolerance to subsequent LPS exposure. gAcrp-mediated increases in TNF-alpha mRNA accumulation were associated with increased TNF-alpha promoter activity. gAcrp increased the DNA binding activity of both Egr-1 and NFkappaB; mutation of either the Egr-1 or NFkappaB binding sites in the TNF-alpha promoter decreased gAcrp-stimulated promoter activity. Further, co-transfection with either dominant negative Egr-1 or the IkappaB super-repressor prevented gAcrp-stimulated TNF-alpha promoter activity. gAcrp also increased Egr-1 promoter activity, mRNA accumulation, and DNA binding activity. Inhibition of ERK1/2 with U0126 potently suppressed gAcrp-stimulated Egr-1 promoter activity, as well as TNF-alpha promoter activity. In summary, these data demonstrate that adiponectin initially increases TNF-alpha production by macrophages via ERK1/2-->Egr-1 and NFkappaB-dependent mechanisms; these increases in TNF-alpha in turn lead to increased expression of interleukin-10 and an eventual dampening of LPS-mediated cytokine production in macrophages.
Collapse
Affiliation(s)
- Pil-Hoon Park
- Department of Pathobiology, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|