1
|
Jiang P, Cheng B, Wang Z, Zheng Z, Duan Q. Distinct effects of physical and functional ablation of brown adipose tissue on T3-dependent pathological cardiac remodeling. Biochem Biophys Res Commun 2024; 735:150844. [PMID: 39432923 DOI: 10.1016/j.bbrc.2024.150844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
Heart failure tends to deteriorate in colder climates, heightening the risk of major adverse cardiovascular events. Brown adipose tissue (BAT) serves as both a thermogenic organ and an atypical site for triiodothyronine (T3) synthesis in response to cold. This study investigates the potential role of BAT in contributing to abdominal aortic constriction (AAC)-induced pathological cardiac remodeling during cold exposure. In this study, we developed a mouse model of pathological cardiac remodeling using AAC. Physical excision of interscapular BAT (iBATx) was performed during cold exposure, and T3 synthesis levels were measured. Additionally, the impact of uncoupling protein 1 (UCP1) knockout on thermogenic function and pathological cardiac remodeling was investigated. In vitro studies were conducted to assess the effect of T3 on cardiomyocyte hypertrophy induced by phenylephrine (PE). Physical removal of interscapular BAT during cold exposure decreased T3 synthesis and mitigated pathological cardiac remodeling. Conversely, UCP1 knockout eliminated thermogenic function during cold exposure, while preserving BAT integrity increased T3 synthesis and exacerbated pathological cardiac remodeling. In vitro, T3 further aggravated cardiomyocyte hypertrophy caused by PE. These findings underscore the distinct effects of physical and functional BAT ablation on pathological cardiac remodeling, primarily through altering T3 levels rather than thermogenesis in cold environments. This research provides new insights into the differential roles of BAT in cardiac health, particularly under cold exposure conditions.
Collapse
Affiliation(s)
- Ping Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China; Medical College of Nanchang University, Nanchang, 330006, China
| | - Banghong Cheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China; Department of Cardiology, Zhuzhou Central Hospital (Zhuzhou Hospital Affiliated to Xiangya School of Medicine), Zhuzhou, China
| | - Zhichao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China; Medical College of Nanchang University, Nanchang, 330006, China
| | - Zeqi Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China; Medical College of Nanchang University, Nanchang, 330006, China.
| | - Qiong Duan
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China; Medical College of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
2
|
Xu Z, Li H, Cao G, Li P, Zhou H, Sun Y. The protective role of brown adipose tissue in cardiac cell damage after myocardial infarction and heart failure. Lipids Health Dis 2024; 23:338. [PMID: 39415186 PMCID: PMC11481725 DOI: 10.1186/s12944-024-02326-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/06/2024] [Indexed: 10/18/2024] Open
Abstract
Acute myocardial infarction (AMI) and related cardiovascular disease complications are the leading causes of mortality worldwide. Brown adipose tissue (BAT) is thermogenic and characterized by the uncoupling protein expression. Recent studies have found that in cardiovascular diseases, activated BAT can effectively improve the prognosis of AMI and concurrent heart failure through intercellular communication. However, a clear and systematic understanding of the myocardial protective mechanism of BAT after AMI is lacking, especially in the endocrine function of BAT. This review describes the effects of BAT on various cells in the heart after AMI. BAT plays a protective role on cardiac cells and fibroblasts during ischemia/reperfusion (I/R), myocardial remodeling, and myocardial fibrosis. This review also discusses the changes caused by BAT activation in different stages of heart failure. Finally, this review summarizes the treatment methods that target BAT to improve AMI. Further in-depth researches are still needed to clarify the underlying mechanism of the connection between BAT and different cells in cardiac tissue in order to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Zhe Xu
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hong Li
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guojie Cao
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Panpan Li
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haitao Zhou
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Sun
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
3
|
Campos I, Richter B, Thomas SM, Czaya B, Yanucil C, Kentrup D, Fajol A, Li Q, Secor SM, Faul C. FGFR4 Is Required for Concentric Growth of Cardiac Myocytes during Physiologic Cardiac Hypertrophy. J Cardiovasc Dev Dis 2024; 11:320. [PMID: 39452290 PMCID: PMC11508992 DOI: 10.3390/jcdd11100320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Fibroblast growth factor (FGF) 23 is a bone-derived hormone that promotes renal phosphate excretion. Serum FGF23 is increased in chronic kidney disease (CKD) and contributes to pathologic cardiac hypertrophy by activating FGF receptor (FGFR) 4 on cardiac myocytes, which might lead to the high cardiovascular mortality in CKD patients. Increases in serum FGF23 levels have also been observed following endurance exercise and in pregnancy, which are scenarios of physiologic cardiac hypertrophy as an adaptive response of the heart to increased demand. To determine whether FGF23/FGFR4 contributes to physiologic cardiac hypertrophy, we studied FGFR4 knockout mice (FGFR4-/-) during late pregnancy. In comparison to virgin littermates, pregnant wild-type and FGFR4-/- mice showed increases in serum FGF23 levels and heart weight; however, the elevation in myocyte area observed in pregnant wild-type mice was abrogated in pregnant FGFR4-/- mice. This outcome was supported by treatments of cultured cardiac myocytes with serum from fed Burmese pythons, another model of physiologic hypertrophy, where the co-treatment with an FGFR4-specific inhibitor abrogated the serum-induced increase in cell area. Interestingly, we found that in pregnant mice, the heart, and not the bone, shows elevated FGF23 expression, and that increases in serum FGF23 are not accompanied by changes in phosphate metabolism. Our study suggests that in physiologic cardiac hypertrophy, the heart produces FGF23 that contributes to hypertrophic growth of cardiac myocytes in a paracrine and FGFR4-dependent manner, and that the kidney does not respond to heart-derived FGF23.
Collapse
Affiliation(s)
- Isaac Campos
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Beatrice Richter
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Sarah Madison Thomas
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Brian Czaya
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Christopher Yanucil
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Dominik Kentrup
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Abul Fajol
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Qing Li
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Stephen M. Secor
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA;
| | - Christian Faul
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| |
Collapse
|
4
|
Li Y, Zhou E, Yu Y, Wang B, Zhang L, Lei R, Xue B, Tian X, Niu J, Liu J, Zhang K, Luo B. Butyrate attenuates cold-induced hypertension via gut microbiota and activation of brown adipose tissue. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 943:173835. [PMID: 38851345 DOI: 10.1016/j.scitotenv.2024.173835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE Chronic exposure to cold temperature is known to elevate blood pressure, leading to a condition known as cold-induced hypertension (CIH). Our previous research suggested correlations between alterations in gut microbiota, decrease in butyrate level, and the onset and progression of CIH. However, the role of butyrate in CIH and the underlying mechanisms need further investigation. METHODS We exposed Specific Pathogen Free (SPF) rats to continuous cold temperature (4 ± 1 °C) for 6 weeks to establish a CIH rat model. Rats were divided into different groups by dose and duration, and the rats under cold were administered with butyrate (0.5 or 1 g/kg/day) daily. We assessed hypertension-associated phenotypes, pathological morphological changes, and endocrine-related phenotypes of brown adipose tissue (BAT). The effects of butyrate on gut microbiota and intestinal content metabolism were evaluated by 16s RNA sequencing and non-targeted metabolomics, respectively. RESULTS The systolic blood pressure (SBP) of rats exposed to cold after supplemented with butyrate were significantly lower than that of the Cold group. Butyrate may increase the species, abundance, and diversity of gut microbiota in rats. Specifically, butyrate intervention enriched beneficial bacterial genera, such as Lactobacillaceae, and decreased the levels of harmful bacteria genera, such as Actinobacteriota and Erysipeiotrichaceae. Cold exposure significantly increased BAT cells and the number of mitochondria. After butyrate supplementation, the levels of peroxisome proliferator-activated receptor gamma coactivator 1a and fibroblast growth factor 21 in BAT were significantly elevated (P < 0.05), and the volume and number of lipid droplets increased. The levels of ANG II and high-density lipoprotein were elevated in the Cold group but decreased after butyrate supplementation. CONCLUSION Butyrate may attenuate blood pressure in CIH by promoting the growth of beneficial bacteria and the secretion of beneficial derived factors produced by BAT, thus alleviating the elevation of blood pressure induced by cold. This study demonstrates the anti-hypertensive effects of butyrate and its potential therapeutic mechanisms, offering novel insights to the prevention and treatment of CIH in populations living or working in cold environments.
Collapse
Affiliation(s)
- Yanlin Li
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Erkai Zhou
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Yunhui Yu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Bo Wang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Ling Zhang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Ruoyi Lei
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Baode Xue
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Xiaoyu Tian
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Jingping Niu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Jiangtao Liu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China.
| | - Kai Zhang
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, One University Place, Rensselaer, NY 12144, USA.
| | - Bin Luo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China; Shanghai Key Laboratory of Meteorology and Health, Shanghai Meteorological Bureau, Shanghai 200030, People's Republic of China; Shanghai Typhoon Institute, China Meteorological Administration, Shanghai 200030, People's Republic of China.
| |
Collapse
|
5
|
Hu Y, Huang Y, Jiang Y, Weng L, Cai Z, He B. The Different Shades of Thermogenic Adipose Tissue. Curr Obes Rep 2024; 13:440-460. [PMID: 38607478 DOI: 10.1007/s13679-024-00559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE OF REVIEW By providing a concise overview of adipose tissue types, elucidating the regulation of adipose thermogenic capacity in both physiological contexts and chronic wasting diseases (a protracted hypermetabolic state that precipitates sustained catabolism and consequent progressive corporeal atrophy), and most importantly, delving into the ongoing discourse regarding the role of adipose tissue thermogenic activation in chronic wasting diseases, this review aims to provide researchers with a comprehensive understanding of the field. RECENT FINDINGS Adipose tissue, traditionally classified as white, brown, and beige (brite) based on its thermogenic activity and potential, is intricately regulated by complex mechanisms in response to exercise or cold exposure. This regulation is adipose depot-specific and dependent on the duration of exposure. Excessive thermogenic activation of adipose tissue has been observed in chronic wasting diseases and has been considered a pathological factor that accelerates disease progression. However, this conclusion may be confounded by the detrimental effects of excessive lipolysis. Recent research also suggests that such activation may play a beneficial role in the early stages of chronic wasting disease and provide potential therapeutic effects. A more comprehensive understanding of the changes in adipose tissue thermogenesis under physiological and pathological conditions, as well as the underlying regulatory mechanisms, is essential for the development of novel interventions to improve health and prevent disease.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yijie Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yangjing Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lvkan Weng
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
6
|
Tang H, Kong Q, Zhang Z, Wu W, Yuan L, Liu X. Regulation of transcription factor function by purinergic signalling in cardiovascular diseases. Purinergic Signal 2024:10.1007/s11302-024-10045-8. [PMID: 39215950 DOI: 10.1007/s11302-024-10045-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular diseases (CVDs), including hypertension, atherosclerosis, myocardial ischemia, and myocardial infarction, constitute the primary cause of mortality worldwide. Transcription factors play critical roles in the development of CVDs and contribute to the pathophysiology of these diseases by coordinating the transcription of many genes involved in inflammation, oxidative stress, angiogenesis, and glycolytic metabolism. One important regulator of hemostasis in both healthy and pathological settings has been identified as a purinergic signalling pathway. Research has demonstrated that several signalling networks implicated in the pathophysiology of CVDs are formed by transcription factors that are regulated by purinergic substances. Here, we briefly summarize the roles and mechanisms of the transcription factors regulated by purinergic pathways in various types of CVD. This information will be essential for discovering novel approaches for CVD treatment and prevention.
Collapse
Affiliation(s)
- Hao Tang
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qihang Kong
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhewei Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenchao Wu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Lixing Yuan
- Public Laboratory of West China Second University Hospital and Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, 610041, China.
| | - Xiaojing Liu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Wang Y, Li Q, Zhou S, Tan P. Contents of exosomes derived from adipose tissue and their regulation on inflammation, tumors, and diabetes. Front Endocrinol (Lausanne) 2024; 15:1374715. [PMID: 39220365 PMCID: PMC11361949 DOI: 10.3389/fendo.2024.1374715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Adipose tissue (AT) serves as an energy-capacitive organ and performs functions involving paracrine- and endocrine-mediated regulation via extracellular vesicles (EVs) secretion. Exosomes, a subtype of EVs, contain various bioactive molecules with regulatory effects, such as nucleic acids, proteins, and lipids. AT-derived exosomes (AT-exos) include exosomes derived from various cells in AT, including adipocytes, adipose-derived stem cells (ADSCs), macrophages, and endothelial cells. This review aimed to comprehensively evaluate the impacts of different AT-exos on the regulation of physiological and pathological processes. The contents and functions of adipocyte-derived exosomes and ADSC-derived exosomes are compared simultaneously, highlighting their similarities and differences. The contents of AT-exos have been shown to exert complex regulatory effects on local inflammation, tumor dynamics, and insulin resistance. Significantly, differences in the cargoes of AT-exos have been observed among diabetes patients, obese individuals, and healthy individuals. These differences could be used to predict the development of diabetes mellitus and as therapeutic targets for improving insulin sensitivity and glucose tolerance. However, further research is needed to elucidate the underlying mechanisms and potential applications of AT-exos.
Collapse
Affiliation(s)
- Yanwen Wang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangbai Zhou
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pohching Tan
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Jang MH, Song J. Adenosine and adenosine receptors in metabolic imbalance-related neurological issues. Biomed Pharmacother 2024; 177:116996. [PMID: 38897158 DOI: 10.1016/j.biopha.2024.116996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/08/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024] Open
Abstract
Metabolic syndromes (e.g., obesity) are characterized by insulin resistance, chronic inflammation, impaired glucose metabolism, and dyslipidemia. Recently, patients with metabolic syndromes have experienced not only metabolic problems but also neuropathological issues, including cognitive impairment. Several studies have reported blood-brain barrier (BBB) disruption and insulin resistance in the brain of patients with obesity and diabetes. Adenosine, a purine nucleoside, is known to regulate various cellular responses (e.g., the neuroinflammatory response) by binding with adenosine receptors in the central nervous system (CNS). Adenosine has four known receptors: A1R, A2AR, A2BR, and A3R. These receptors play distinct roles in various physiological and pathological processes in the brain, including endothelial cell homeostasis, insulin sensitivity, microglial activation, lipid metabolism, immune cell infiltration, and synaptic plasticity. Here, we review the recent findings on the role of adenosine receptor-mediated signaling in neuropathological issues related to metabolic imbalance. We highlight the importance of adenosine signaling in the development of therapeutic solutions for neuropathological issues in patients with metabolic syndromes.
Collapse
Affiliation(s)
- Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea.
| |
Collapse
|
9
|
Challa AA, Vidal P, Maurya SK, Maurya CK, Baer LA, Wang Y, James NM, Pardeshi PJ, Fasano M, Carley AN, Stanford KI, Lewandowski ED. UCP1-dependent brown adipose activation accelerates cardiac metabolic remodeling and reduces initial hypertrophic and fibrotic responses to pathological stress. FASEB J 2024; 38:e23709. [PMID: 38809700 PMCID: PMC11163965 DOI: 10.1096/fj.202400922r] [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: 04/22/2024] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
Brown adipose tissue (BAT) is correlated to cardiovascular health in rodents and humans, but the physiological role of BAT in the initial cardiac remodeling at the onset of stress is unknown. Activation of BAT via 48 h cold (16°C) in mice following transverse aortic constriction (TAC) reduced cardiac gene expression for LCFA uptake and oxidation in male mice and accelerated the onset of cardiac metabolic remodeling, with an early isoform shift of carnitine palmitoyltransferase 1 (CPT1) toward increased CPT1a, reduced entry of long chain fatty acid (LCFA) into oxidative metabolism (0.59 ± 0.02 vs. 0.72 ± 0.02 in RT TAC hearts, p < .05) and increased carbohydrate oxidation with altered glucose transporter content. BAT activation with TAC reduced early hypertrophic expression of β-MHC by 61% versus RT-TAC and reduced pro-fibrotic TGF-β1 and COL3α1 expression. While cardiac natriuretic peptide expression was yet to increase at only 3 days TAC, Nppa and Nppb expression were elevated in Cold TAC versus RT TAC hearts 2.7- and 2.4-fold, respectively. Eliminating BAT thermogenic activation with UCP1 KO mice eliminated differences between Cold TAC and RT TAC hearts, confirming effects of BAT activation rather than autonomous cardiac responses to cold. Female responses to BAT activation were blunted, with limited UCP1 changes with cold, partly due to already activated BAT in females at RT compared to thermoneutrality. These data reveal a previously unknown physiological mechanism of UCP1-dependent BAT activation in attenuating early cardiac hypertrophic and profibrotic signaling and accelerating remodeled metabolic activity in the heart at the onset of cardiac stress.
Collapse
Affiliation(s)
- Azariyas A. Challa
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Pablo Vidal
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Santosh K. Maurya
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Chandan K. Maurya
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Lisa A. Baer
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Yang Wang
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Natasha Maria James
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Parth J. Pardeshi
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Matthew Fasano
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Andrew N. Carley
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Kristin I. Stanford
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - E. Douglas Lewandowski
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| |
Collapse
|
10
|
Ghesmati Z, Rashid M, Fayezi S, Gieseler F, Alizadeh E, Darabi M. An update on the secretory functions of brown, white, and beige adipose tissue: Towards therapeutic applications. Rev Endocr Metab Disord 2024; 25:279-308. [PMID: 38051471 PMCID: PMC10942928 DOI: 10.1007/s11154-023-09850-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 12/07/2023]
Abstract
Adipose tissue, including white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue, is vital in modulating whole-body energy metabolism. While WAT primarily stores energy, BAT dissipates energy as heat for thermoregulation. Beige adipose tissue is a hybrid form of adipose tissue that shares characteristics with WAT and BAT. Dysregulation of adipose tissue metabolism is linked to various disorders, including obesity, type 2 diabetes, cardiovascular diseases, cancer, and infertility. Both brown and beige adipocytes secrete multiple molecules, such as batokines, packaged in extracellular vesicles or as soluble signaling molecules that play autocrine, paracrine, and endocrine roles. A greater understanding of the adipocyte secretome is essential for identifying novel molecular targets in treating metabolic disorders. Additionally, microRNAs show crucial roles in regulating adipose tissue differentiation and function, highlighting their potential as biomarkers for metabolic disorders. The browning of WAT has emerged as a promising therapeutic approach in treating obesity and associated metabolic disorders. Many browning agents have been identified, and nanotechnology-based drug delivery systems have been developed to enhance their efficacy. This review scrutinizes the characteristics of and differences between white, brown, and beige adipose tissues, the molecular mechanisms involved in the development of the adipocytes, the significant roles of batokines, and regulatory microRNAs active in different adipose tissues. Finally, the potential of WAT browning in treating obesity and atherosclerosis, the relationship of BAT with cancer and fertility disorders, and the crosstalk between adipose tissue with circadian system and circadian disorders are also investigated.
Collapse
Affiliation(s)
- Zeinab Ghesmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Fayezi
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Frank Gieseler
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany.
| |
Collapse
|
11
|
Romero-Becera R, Santamans AM, Arcones AC, Sabio G. From Beats to Metabolism: the Heart at the Core of Interorgan Metabolic Cross Talk. Physiology (Bethesda) 2024; 39:98-125. [PMID: 38051123 DOI: 10.1152/physiol.00018.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/26/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023] Open
Abstract
The heart, once considered a mere blood pump, is now recognized as a multifunctional metabolic and endocrine organ. Its function is tightly regulated by various metabolic processes, at the same time it serves as an endocrine organ, secreting bioactive molecules that impact systemic metabolism. In recent years, research has shed light on the intricate interplay between the heart and other metabolic organs, such as adipose tissue, liver, and skeletal muscle. The metabolic flexibility of the heart and its ability to switch between different energy substrates play a crucial role in maintaining cardiac function and overall metabolic homeostasis. Gaining a comprehensive understanding of how metabolic disorders disrupt cardiac metabolism is crucial, as it plays a pivotal role in the development and progression of cardiac diseases. The emerging understanding of the heart as a metabolic and endocrine organ highlights its essential contribution to whole body metabolic regulation and offers new insights into the pathogenesis of metabolic diseases, such as obesity, diabetes, and cardiovascular disorders. In this review, we provide an in-depth exploration of the heart's metabolic and endocrine functions, emphasizing its role in systemic metabolism and the interplay between the heart and other metabolic organs. Furthermore, emerging evidence suggests a correlation between heart disease and other conditions such as aging and cancer, indicating that the metabolic dysfunction observed in these conditions may share common underlying mechanisms. By unraveling the complex mechanisms underlying cardiac metabolism, we aim to contribute to the development of novel therapeutic strategies for metabolic diseases and improve overall cardiovascular health.
Collapse
Affiliation(s)
| | | | - Alba C Arcones
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| |
Collapse
|
12
|
Mendez-Gutierrez A, Aguilera CM, Cereijo R, Osuna-Prieto FJ, Martinez-Tellez B, Rico MC, Sanchez-Infantes D, Villarroya F, Ruiz JR, Sanchez-Delgado G. Cold exposure modulates potential brown adipokines in humans, but only FGF21 is associated with brown adipose tissue volume. Obesity (Silver Spring) 2024; 32:560-570. [PMID: 38247441 DOI: 10.1002/oby.23970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 01/23/2024]
Abstract
OBJECTIVE The study objective was to investigate the effect of cold exposure on the plasma levels of five potential human brown adipokines (chemokine ligand 14 [CXCL14], growth differentiation factor 15 [GDF15], fibroblast growth factor 21 [FGF21], interleukin 6 [IL6], and bone morphogenic protein 8b [BMP8b]) and to study whether such cold-induced effects are related to brown adipose tissue (BAT) volume, activity, or radiodensity in young humans. METHODS Plasma levels of brown adipokines were measured before and 1 h and 2 h after starting an individualized cold exposure in 30 young adults (60% women, 21.9 ± 2.3 y; 24.9 ± 5.1 kg/m2 ). BAT volume, 18 F-fluorodeoxyglucose uptake, and radiodensity were assessed by a static positron emission tomography-computerized tomography scan after cold exposure. RESULTS Cold exposure increased the concentration of CXCL14 (Δ2h = 0.58 ± 0.98 ng/mL; p = 0.007), GDF15 (Δ2h = 19.63 ± 46.2 pg/mL; p = 0.013), FGF21 (Δ2h = 33.72 ± 55.13 pg/mL; p = 0.003), and IL6 (Δ1h = 1.98 ± 3.56 pg/mL; p = 0.048) and reduced BMP8b (Δ2h = -37.12 ± 83.53 pg/mL; p = 0.022). The cold-induced increase in plasma FGF21 was positively associated with BAT volume (Δ2h: β = 0.456; R2 = 0.307; p = 0.001), but not with 18 F-fluorodeoxyglucose uptake or radiodensity. None of the changes in the other studied brown adipokines was related to BAT volume, activity, or radiodensity. CONCLUSIONS Cold exposure modulates plasma levels of several potential brown adipokines in humans, whereas only cold-induced changes in FGF21 levels are associated with BAT volume. These findings suggest that human BAT might contribute to the circulatory pool of FGF21.
Collapse
Affiliation(s)
- Andrea Mendez-Gutierrez
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Center of Biomedical Research, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Granada, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Concepcion M Aguilera
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Center of Biomedical Research, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Granada, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Rubén Cereijo
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, Sant Joan de Déu Research Institute, University of Barcelona, Barcelona, Spain
| | - Francisco J Osuna-Prieto
- Hospital Universitari Joan XXIII de Tarragona, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Department of Physical Education and Sports, Faculty of Sports Science, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Borja Martinez-Tellez
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Physical Education and Sports, Faculty of Sports Science, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Education, Faculty of Education Sciences and SPORT Research Group (CTS-1024), CERNEP Research Center, University of Almería, Almería, Spain
| | - Maria C Rico
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Center of Biomedical Research, University of Granada, Granada, Spain
| | - David Sanchez-Infantes
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), Madrid, Spain
| | - Francesc Villarroya
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, Sant Joan de Déu Research Institute, University of Barcelona, Barcelona, Spain
| | - Jonatan R Ruiz
- Instituto de Investigación Biosanitaria, Granada, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Physical Education and Sports, Faculty of Sports Science, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Guillermo Sanchez-Delgado
- "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Center of Biomedical Research, University of Granada, Granada, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Physical Education and Sports, Faculty of Sports Science, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
- Department of Medicine, Division of Endocrinology, Centre de recherche du Centre Hospitalier de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
13
|
Wang T, Zhao C, Zhang J, Li S, Zhang Y, Gong Y, Zhou Y, Yan L, Zhang S, Zhang Z, Hu H, Liu A, Bai X, Zou Z. Whitening of brown adipose tissue inhibits osteogenic differentiation via secretion of S100A8/A9. iScience 2024; 27:108857. [PMID: 38303710 PMCID: PMC10830855 DOI: 10.1016/j.isci.2024.108857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/20/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
The mechanism by which brown adipose tissue (BAT) regulates bone metabolism is unclear. Here, we reveal that BAT secretes S100A8/A9, a previously unidentified BAT adipokine (batokine), to impair bone formation. Brown adipocytes-specific knockout of Rheb (RhebBAD KO), the upstream activator of mTOR, causes BAT malfunction to inhibit osteogenesis. Rheb depletion induces NF-κB dependent S100A8/A9 secretion from brown adipocytes, but not from macrophages. In wild-type mice, age-related Rheb downregulation in BAT is associated with enhanced S100A8/A9 secretion. Either batokines from RhebBAD KO mice, or recombinant S100A8/A9, inhibits osteoblast differentiation of mesenchymal stem cells in vitro by targeting toll-like receptor 4 on their surfaces. Conversely, S100A8/A9 neutralization not only rescues the osteogenesis repressed in the RhebBAD KO mice, but also alleviates age-related osteoporosis in wild-type mice. Collectively, our data revealed an unexpected BAT-bone crosstalk driven by Rheb-S100A8/A9, uncovering S100A8/A9 as a promising target for the treatment, and potentially, prevention of osteoporosis.
Collapse
Affiliation(s)
- Ting Wang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chaoran Zhao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiahuan Zhang
- Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shengfa Li
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Youming Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Gong
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yingyue Zhou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lei Yan
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sheng Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopadics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongling Hu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, China
| | - Anling Liu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhipeng Zou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
14
|
Zhu J, Hou B, Rong H, Xu K, Jiang L, Yang S, Zhu H, Yang H, Jiao Y, Liu Y, Ni K, Ma Z. Blocking brown adipocyte β 3-adrenoceptor attenuates blood-spinal cord barrier impairment and chronic postsurgical pain in a rat model of preoperative stress. Int Immunopharmacol 2024; 128:111530. [PMID: 38278068 DOI: 10.1016/j.intimp.2024.111530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/28/2024]
Abstract
Preoperative stress has been recognized as an independent risk factor for chronic postsurgical pain (CPSP). However, the underlying mechanisms of CPSP influenced by preoperative stress remain elusive. Previous studies indicated that excessive stress could induce disruption of the blood-spinal cord barrier (BSCB). We wondered whether and how BSCB involves in CPSP by using a single prolonged stress (SPS) combining plantar incision model in male rats to mimic preoperative stress-related postsurgical pain. Here, we observed that preoperative SPS-exposed rats exhibited relentless incisional pain, which was accompanied by impairment of BSCB and persistent elevation of serum IL-6. Intraperitoneal injections of Tocilizumab (an IL-6 receptor monoclonal antibody) not only mitigated BSCB breakdown but also alleviated pain behaviors. In addition, intervening β3-adrenoceptor (ADRB3) signaling in brown adipocytes by SR59230a (a specific ADRB3 antagonist) treatment or removal of brown adipose tissues could effectively decrease serum IL-6 levels, ameliorate BSCB disruption, and alleviate incisional pain. Further results displayed that SI-exposed rats also showed markedly spinal microglia activation. And exogenous His-tagged IL-6 could pass through the disrupted BSCB, which might contribute to microglia activation. Injection of SR59230a or ablation of brown adipose tissues could effectively reduce the activation of spinal microglia. Thus, our findings suggest that serum IL-6 induced by brown adipocyte ADRB3 signaling contributed to BSCB disruption and spinal microglia activation, which might be involved in preoperative stress mediated CPSP. This work indicates a promising treatment strategy for preoperative stress induced CPSP by blocking ADRB3.
Collapse
Affiliation(s)
- Jixiang Zhu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224006, China
| | - Bailing Hou
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Hui Rong
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Ke Xu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Li Jiang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing Medical University, Nanjing 210008, China
| | - Shuai Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Huijie Zhu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Haikou Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Yang Jiao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yue Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Kun Ni
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing Medical University, Nanjing 210008, China.
| |
Collapse
|
15
|
Cai M, Ye H, Zhu X, Li X, Cai L, Jin J, Chen Q, Shi Y, Yang L, Wang L, Huang X. Fibroblast Growth Factor 21 Relieves Lipopolysaccharide-Induced Acute Lung Injury by Suppressing JAK2/STAT3 Signaling Pathway. Inflammation 2024; 47:209-226. [PMID: 37864659 PMCID: PMC10799097 DOI: 10.1007/s10753-023-01905-3] [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: 02/19/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 10/23/2023]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a life-threatening disease without an effective drug at present. Fibroblast growth factor 21 (FGF21) was reported to be protective against inflammation in metabolic disease in recent studies. However, the role of FGF21 in ALI has been rarely investigated. In this study, it was found that the expression of FGF21 was markedly increased in lung tissue under lipopolysaccharide (LPS) stimulation in vivo, whereas it was decreased in lung epithelial cells under LPS stimulation in vitro. Therefore, our research aimed to elucidate the potential role of FGF21 in LPS-induced ALI and to detect possible underlying mechanisms. The results revealed that the deficiency of FGF21 aggravated pathological damage, inflammatory infiltration, and pulmonary function in LPS-induced ALI, while exogenous administration of FGF21 improved these manifestations. Moreover, through RNA sequencing and enrichment analysis, it was unveiled that FGF21 might play a protective role in LPS-induced ALI via JAK2/STAT3 signaling pathway. The therapeutic effect of FGF21 was weakened after additional usage of JAK2 activator in vivo. Further investigation revealed that FGF21 significantly inhibited STAT3 phosphorylation and impaired the nuclear translocation of STAT3 in vitro. In addition, the aggravation of inflammation caused by silencing FGF21 can be alleviated by JAK2 inhibitor in vitro. Collectively, these findings unveil a potent protective effect of FGF21 against LPS-induced ALI by inhibiting the JAK2/STAT3 pathway, implying that FGF21 might be a novel and effective therapy for ALI.
Collapse
Affiliation(s)
- Mengsi Cai
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Huihui Ye
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Xiayan Zhu
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Xiuchun Li
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Luqiong Cai
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Jiajia Jin
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Qiwen Chen
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Yuzhe Shi
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Lehe Yang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Liangxing Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China.
| | - Xiaoying Huang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou Medical University, Xuefu North Street, Wenzhou, Zhejiang, 325000, People's Republic of China.
| |
Collapse
|
16
|
Pfeifer A, Mikhael M, Niemann B. Inosine: novel activator of brown adipose tissue and energy homeostasis. Trends Cell Biol 2024; 34:72-82. [PMID: 37188562 DOI: 10.1016/j.tcb.2023.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/17/2023]
Abstract
Extracellular purinergic molecules act as signaling molecules that bind to cellular receptors and regulate signaling pathways. Growing evidence suggests that purines regulate adipocyte function and whole-body metabolism. Here, we focus on one specific purine: inosine. Brown adipocytes, which are important regulators of whole-body energy expenditure (EE), release inosine when they are stressed or become apoptotic. Unexpectedly, inosine activates EE in neighboring brown adipocytes and enhances differentiation of brown preadipocytes. Increasing extracellular inosine, either directly by increasing inosine intake or indirectly via pharmacological inhibition of cellular inosine transporters, increases whole-body EE and counteracts obesity. Thus, inosine and other closely related purines might be a novel approach to tackle obesity and associated metabolic disorders by enhancing EE.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany.
| | - Mickel Mikhael
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Birte Niemann
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| |
Collapse
|
17
|
Ziqubu K, Dludla PV, Mabhida SE, Jack BU, Keipert S, Jastroch M, Mazibuko-Mbeje SE. Brown adipose tissue-derived metabolites and their role in regulating metabolism. Metabolism 2024; 150:155709. [PMID: 37866810 DOI: 10.1016/j.metabol.2023.155709] [Citation(s) in RCA: 1] [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: 04/04/2023] [Revised: 09/28/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
The discovery and rejuvenation of metabolically active brown adipose tissue (BAT) in adult humans have offered a new approach to treat obesity and metabolic diseases. Beyond its accomplished role in adaptive thermogenesis, BAT secretes signaling molecules known as "batokines", which are instrumental in regulating whole-body metabolism via autocrine, paracrine, and endocrine action. In addition to the intrinsic BAT metabolite-oxidizing activity, the endocrine functions of these molecules may help to explain the association between BAT activity and a healthy systemic metabolic profile. Herein, we review the evidence that underscores the significance of BAT-derived metabolites, especially highlighting their role in controlling physiological and metabolic processes involving thermogenesis, substrate metabolism, and other essential biological processes. The conversation extends to their capacity to enhance energy expenditure and mitigate features of obesity and its related metabolic complications. Thus, metabolites derived from BAT may provide new avenues for the discovery of metabolic health-promoting drugs with far-reaching impacts. This review aims to dissect the complexities of the secretory role of BAT in modulating local and systemic metabolism in metabolic health and disease.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | - Phiwayinkosi V Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Sihle E Mabhida
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Babalwa U Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Susanne Keipert
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | | |
Collapse
|
18
|
Alvarez-Gallego F, González-Blázquez R, Gil-Ortega M, Somoza B, Calderón-Dominguez M, Moratinos J, Garcia-Garcia V, Fernández P, González-Moreno D, Viana M, Alcalá M. Angiotensin II type 2 receptor as a novel activator of brown adipose tissue in obesity. Biofactors 2023; 49:1106-1120. [PMID: 37286331 DOI: 10.1002/biof.1981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/20/2023] [Indexed: 06/09/2023]
Abstract
The angiotensin II type 2 receptor (AT2R) exerts vasorelaxant, anti-inflammatory, and antioxidant properties. In obesity, its activation counterbalances the adverse cardiovascular effects of angiotensin II mediated by the AT1R. Preliminary results indicate that it also promotes brown adipocyte differentiation in vitro. Our hypothesis is that AT2R activation could increase BAT mass and activity in obesity. Five-week-old male C57BL/6J mice were fed a standard or a high-fat (HF) diet for 6 weeks. Half of the animals were treated with compound 21 (C21), a selective AT2R agonist, (1 mg/kg/day) in the drinking water. Electron transport chain (ETC), oxidative phosphorylation, and UCP1 proteins were measured in the interscapular BAT (iBAT) and thoracic perivascular adipose tissue (tPVAT) as well as inflammatory and oxidative parameters. Differentiation and oxygen consumption rate (OCR) in the presence of C21 was tested in brown preadipocytes. In vitro, C21-differentiated brown adipocytes showed an AT2R-dependent increase of differentiation markers (Ucp1, Cidea, Pparg) and increased basal and H+ leak-linked OCR. In vivo, HF-C21 mice showed increased iBAT mass compared to HF animals. Both their iBAT and tPVAT showed higher protein levels of the ETC protein complexes and UCP1, together with a reduction of inflammatory and oxidative markers. The activation of the AT2R increases BAT mass, mitochondrial activity, and reduces markers of tissue inflammation and oxidative stress in obesity. Therefore, insulin reduction and better vascular responses are achieved. Thus, the activation of the protective arm of the renin-angiotensin system arises as a promising tool in the treatment of obesity.
Collapse
Affiliation(s)
- Fabiola Alvarez-Gallego
- Departamento de Química y Bioquímica, Facultad de Farmacia., Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Raquel González-Blázquez
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - María Calderón-Dominguez
- Biomedical Research and Innovation Institute of Cadiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cádiz, Spain
| | - Javier Moratinos
- Instituto de Medicina Molecular Aplicada Nemesio Díez, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Virginia Garcia-Garcia
- Instituto de Medicina Molecular Aplicada Nemesio Díez, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Paloma Fernández
- Instituto de Medicina Molecular Aplicada Nemesio Díez, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Daniel González-Moreno
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Marta Viana
- Departamento de Química y Bioquímica, Facultad de Farmacia., Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Martín Alcalá
- Departamento de Química y Bioquímica, Facultad de Farmacia., Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, Madrid, Spain
| |
Collapse
|
19
|
Vincenzi F, Pasquini S, Contri C, Cappello M, Nigro M, Travagli A, Merighi S, Gessi S, Borea PA, Varani K. Pharmacology of Adenosine Receptors: Recent Advancements. Biomolecules 2023; 13:1387. [PMID: 37759787 PMCID: PMC10527030 DOI: 10.3390/biom13091387] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Adenosine receptors (ARs) are widely acknowledged pharmacological targets yet are still underutilized in clinical practice. Their ubiquitous distribution in almost all cells and tissues of the body makes them, on the one hand, excellent candidates for numerous diseases, and on the other hand, intrinsically challenging to exploit selectively and in a site-specific manner. This review endeavors to comprehensively depict the substantial advancements witnessed in recent years concerning the development of drugs that modulate ARs. Through preclinical and clinical research, it has become evident that the modulation of ARs holds promise for the treatment of numerous diseases, including central nervous system disorders, cardiovascular and metabolic conditions, inflammatory and autoimmune diseases, and cancer. The latest studies discussed herein shed light on novel mechanisms through which ARs exert control over pathophysiological states. They also introduce new ligands and innovative strategies for receptor activation, presenting compelling evidence of efficacy along with the implicated signaling pathways. Collectively, these emerging insights underscore a promising trajectory toward harnessing the therapeutic potential of these multifaceted targets.
Collapse
Affiliation(s)
- Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Silvia Pasquini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Martina Cappello
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Manuela Nigro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Alessia Travagli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Stefania Merighi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Stefania Gessi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | | | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| |
Collapse
|
20
|
Pestel J, Blangero F, Watson J, Pirola L, Eljaafari A. Adipokines in obesity and metabolic-related-diseases. Biochimie 2023; 212:48-59. [PMID: 37068579 DOI: 10.1016/j.biochi.2023.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
The discovery of leptin in the 1990s led to a reconsideration of adipose tissue (AT) as not only a fatty acid storage organ, but also a proper endocrine tissue. AT is indeed capable of secreting bioactive molecules called adipokines for white AT or batokines for brown/beige AT, which allow communication with numerous organs, especially brain, heart, liver, pancreas, and/or the vascular system. Adipokines exert pro or anti-inflammatory activities. An equilibrated balance between these two sets ensures homeostasis of numerous tissues and organs. During the development of obesity, AT remodelling leads to an alteration of its endocrine activity, with increased secretion of pro-inflammatory adipokines relative to the anti-inflammatory ones, as shown in the graphical abstract. Pro-inflammatory adipokines take part in the initiation of local and systemic inflammation during obesity and contribute to comorbidities associated to obesity, as detailed in the present review.
Collapse
Affiliation(s)
- Julien Pestel
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Ferdinand Blangero
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Julia Watson
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Luciano Pirola
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Assia Eljaafari
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France; Hospices Civils de Lyon: 2 quai des Célestins, 69001 Lyon, France.
| |
Collapse
|
21
|
Martí-Pàmies Í, Thoonen R, Morley M, Graves L, Tamez J, Caplan A, McDaid K, Yao V, Hindle A, Gerszten RE, Laurie A. Farrell, Li L, Tseng YH, Profeta G, Buys ES, Bloch DB, Scherrer-Crosbie M. Brown Adipose Tissue and BMP3b Decrease Injury in Cardiac Ischemia-Reperfusion. Circ Res 2023; 133:353-365. [PMID: 37462036 PMCID: PMC10528340 DOI: 10.1161/circresaha.122.322337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 07/07/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Despite advances in treatment, myocardial infarction (MI) is a leading cause of heart failure and death worldwide, with both ischemia and reperfusion (I/R) causing cardiac injury. A previous study using a mouse model of nonreperfused MI showed activation of brown adipose tissue (BAT). Recent studies showed that molecules secreted by BAT target the heart. We investigated whether BAT attenuates cardiac injury in I/R and sought to identify potential cardioprotective proteins secreted by BAT. METHODS Myocardial I/R surgery with or without BAT transplantation was performed in wild-type (WT) mice and in mice with impaired BAT function (uncoupling protein 1 [Ucp1]-deficient mice). To identify potential cardioprotective factors produced by BAT, RNA-seq (RNA sequencing) was performed in BAT from WT and Ucp1-/- mice. Subsequently, myocardial I/R surgery with or without BAT transplantation was performed in Bmp3b (bone morphogenetic protein 3b)-deficient mice, and WT mice subjected to myocardial I/R were treated using BMP3b. RESULTS Dysfunction of BAT in mice was associated with larger MI size after I/R; conversely, augmenting BAT by transplantation decreased MI size. We identified Bmp3b as a protein secreted by BAT after I/R. Compared with WT mice, Bmp3b-deficient mice developed larger MIs. Increasing functional BAT by transplanting BAT from WT mice to Bmp3b-deficient mice reduced I/R injury whereas transplanting BAT from Bmp3b-deficient mice did not. Treatment of WT mice with BMP3b before reperfusion decreased MI size. The cardioprotective effect of BMP3b was mediated through SMAD1/5/8. In humans, the plasma level of BMP3b increased after MI and was positively correlated with the extent of cardiac injury. CONCLUSIONS The results of this study suggest a cardioprotective role of BAT and BMP3b, a protein secreted by BAT, in a model of I/R injury. Interventions increasing BMP3b levels or targeting Smad 1/5 may represent novel therapeutic approaches to decrease myocardial damage in I/R injury.
Collapse
Affiliation(s)
- Íngrid Martí-Pàmies
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Robrecht Thoonen
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - Michael Morley
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Lauren Graves
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Jesus Tamez
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Alex Caplan
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Kendra McDaid
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Vincent Yao
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - Allyson Hindle
- Anesthesia Center for Critical Care Research, Massachusetts General Hospital, Boston, MA, United States
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Laurie A. Farrell
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Li Li
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Gerson Profeta
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Emmanuel S Buys
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, United States
| | - Donald B Bloch
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, United States
- The Center for Immunology and Inflammatory Diseases and the Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, United States
| | - Marielle Scherrer-Crosbie
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
22
|
Zhuang T, Lei Y, Chang JJ, Zhou YP, Li Y, Li YX, Yang YF, Chen MH, Meng T, Fu SM, Huang LH, Cheang WS, Cooke JP, Dong ZH, Bai YN, Ruan CC. A2AR-mediated lymphangiogenesis via VEGFR2 signaling prevents salt-sensitive hypertension. Eur Heart J 2023; 44:2730-2742. [PMID: 37377160 PMCID: PMC10393074 DOI: 10.1093/eurheartj/ehad377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 04/17/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
AIMS Excess dietary sodium intake and retention lead to hypertension. Impaired dermal lymphangiogenesis and lymphatic dysfunction-mediated sodium and fluid imbalance are pathological mechanisms. The adenosine A2A receptor (A2AR) is expressed in lymphatic endothelial cells (LECs), while the roles and mechanisms of LEC-A2AR in skin lymphangiogenesis during salt-induced hypertension are not clear. METHODS AND RESULTS The expression of LEC-A2AR correlated with lymphatic vessel density in both high-salt diet (HSD)-induced hypertensive mice and hypertensive patients. Lymphatic endothelial cell-specific A2AR knockout mice fed HSD exhibited 17 ± 2% increase in blood pressure and 17 ± 3% increase in Na+ content associated with decreased lymphatic density (-19 ± 2%) compared with HSD-WT mice. A2AR activation by agonist CGS21680 increased lymphatic capillary density and decreased blood pressure in HSD-WT mice. Furthermore, this A2AR agonist activated MSK1 directly to promote VEGFR2 activation and endocytosis independently of VEGF as assessed by phosphoprotein profiling and immunoprecipitation assays in LECs. VEGFR2 kinase activity inhibitor fruquintinib or VEGFR2 knockout in LECs but not VEGF-neutralizing antibody bevacizumab suppressed A2AR activation-mediated decrease in blood pressure. Immunostaining revealed phosphorylated VEGFR2 and MSK1 expression in the LECs were positively correlated with skin lymphatic vessel density and A2AR level in hypertensive patients. CONCLUSION The study highlights a novel A2AR-mediated VEGF-independent activation of VEGFR2 signaling in dermal lymphangiogenesis and sodium balance, which might be a potential therapeutic target in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Tao Zhuang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Yu Lei
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Jin-Jia Chang
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Yan-Ping Zhou
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pu-Jian Road, Shanghai 200032, China
| | - Yan Li
- Department of Cardiology, RuiJin Hospital/LuWan Branch, Shanghai Jiao Tong University School of Medicine, 149 Chong-Qing-Nan Road, Shanghai 200032, China
| | - Yan-Xiu Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guang-Zhou Road, Nanjing 210000, China
| | - Yong-Feng Yang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Mei-Hua Chen
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Ting Meng
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Shi-Man Fu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Li-Hao Huang
- Department of Chemistry and Institute of Metabolism and Integrative Biology, Shanghai Key Laboratory of Metabolic Remodeling and Health, Fudan University, 38 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Wai-San Cheang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Long-Ma Road, Macau 999078, China
| | - John P Cooke
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Zhi-Hui Dong
- Department of Vascular Surgery, Zhongshan Hospital, and Center for Vascular Surgery and Wound Care, Jinshan Hospital, Fudan University, 180 Feng-Lin Road, Shanghai 200032, China
| | - Ying-Nan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Feng-Lin Road, Shanghai 200032, China
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| |
Collapse
|
23
|
Puengel T, Tacke F. Efruxifermin, an investigational treatment for fibrotic or cirrhotic non-alcoholic steatohepatitis (NASH). Expert Opin Investig Drugs 2023. [PMID: 37376813 DOI: 10.1080/13543784.2023.2230115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/14/2023] [Accepted: 06/23/2023] [Indexed: 06/29/2023]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease and strongly associated with metabolic disorders: obesity, type 2 diabetes (T2D), cardiovascular disease. Persistent metabolic injury results in inflammatory processes leading to nonalcoholic steatohepatitis (NASH), liver fibrosis and ultimately cirrhosis. To date, no pharmacologic agent is approved for the treatment of NASH. Fibroblast growth factor 21 (FGF21) agonism has been linked to beneficial metabolic effects ameliorating obesity, steatosis and insulin resistance, supporting its potential as a therapeutic target in NAFLD. AREAS COVERED Efruxifermin (EFX, also AKR-001 or AMG876) is an engineered Fc-FGF21 fusion protein with an optimized pharmacokinetic and pharmacodynamic profile, which is currently tested in several phase 2 clinical trials for the treatment of NASH, fibrosis and compensated liver cirrhosis. EFX improved metabolic disturbances including glycemic control, showed favorable safety and tolerability, and demonstrated antifibrotic efficacy according to FDA requirements for phase 3 trials. EXPERT OPINION While some other FGF-21 agonists (e.g. pegbelfermin) are currently not further investigated, available evidence supports the development of EFX as a promising anti-NASH drug in fibrotic and cirrhotic populations. However, antifibrotic efficacy, long-term safety and benefits (i.e. cardiovascular risk, decompensation events, disease progression, liver transplantation, mortality) remain to be determined.
Collapse
Affiliation(s)
- Tobias Puengel
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
24
|
Zhao Z, Cui X, Liao Z. Mechanism of fibroblast growth factor 21 in cardiac remodeling. Front Cardiovasc Med 2023; 10:1202730. [PMID: 37416922 PMCID: PMC10322220 DOI: 10.3389/fcvm.2023.1202730] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/07/2023] [Indexed: 07/08/2023] Open
Abstract
Cardiac remodeling is a basic pathological process that enables the progression of multiple cardiac diseases to heart failure. Fibroblast growth factor 21 is considered a regulator in maintaining energy homeostasis and shows a positive role in preventing damage caused by cardiac diseases. This review mainly summarizes the effects and related mechanisms of fibroblast growth factor 21 on pathological processes associated with cardiac remodeling, based on a variety of cells of myocardial tissue. The possibility of Fibroblast growth factor 21 as a promising treatment for the cardiac remodeling process will also be discussed.
Collapse
Affiliation(s)
- Zeyu Zhao
- Queen Mary College, Nanchang University, Nanchang, China
| | - Xuemei Cui
- Fourth Clinical Medical College, Nanchang University, Nanchang, China
| | - Zhangping Liao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology School of Pharmaceutical Science, Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Wang S, Liu Y, Chen J, He Y, Ma W, Liu X, Sun X. Effects of multi-organ crosstalk on the physiology and pathology of adipose tissue. Front Endocrinol (Lausanne) 2023; 14:1198984. [PMID: 37383400 PMCID: PMC10293893 DOI: 10.3389/fendo.2023.1198984] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
In previous studies, adipocytes were found to play an important role in regulating whole-body nutrition and energy balance, and are also important in energy metabolism, hormone secretion, and immune regulation. Different adipocytes have different contributions to the body, with white adipocytes primarily storing energy and brown adipocytes producing heat. Recently discovered beige adipocytes, which have characteristics in between white and brown adipocytes, also have the potential to produce heat. Adipocytes interact with other cells in the microenvironment to promote blood vessel growth and immune and neural network interactions. Adipose tissue plays an important role in obesity, metabolic syndrome, and type 2 diabetes. Dysfunction in adipose tissue endocrine and immune regulation can cause and promote the occurrence and development of related diseases. Adipose tissue can also secrete multiple cytokines, which can interact with organs; however, previous studies have not comprehensively summarized the interaction between adipose tissue and other organs. This article reviews the effect of multi-organ crosstalk on the physiology and pathology of adipose tissue, including interactions between the central nervous system, heart, liver, skeletal muscle, and intestines, as well as the mechanisms of adipose tissue in the development of various diseases and its role in disease treatment. It emphasizes the importance of a deeper understanding of these mechanisms for the prevention and treatment of related diseases. Determining these mechanisms has enormous potential for identifying new targets for treating diabetes, metabolic disorders, and cardiovascular diseases.
Collapse
Affiliation(s)
- Sufen Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yifan Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiaqi Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yuejing He
- Clinical Laboratory, Dongguan Eighth People’s Hospital, Dongguan, China
| | - Wanrui Ma
- Department of General Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xuerong Sun
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
26
|
Yan B, Mei Z, Tang Y, Song H, Wu H, Jing Q, Zhang X, Yan C, Han Y. FGF21-FGFR1 controls mitochondrial homeostasis in cardiomyocytes by modulating the degradation of OPA1. Cell Death Dis 2023; 14:311. [PMID: 37156793 PMCID: PMC10167257 DOI: 10.1038/s41419-023-05842-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/10/2023]
Abstract
Fibroblast growth factor 21 (FGF21) is a pleiotropic hormone secreted primarily by the liver and is considered a major regulator of energy homeostasis. Recent research has revealed that FGF21 could play an important role in cardiac pathological remodeling effects and prevention of cardiomyopathy; however, the underlying mechanism remains largely unknown. This study aimed to determine the mechanism underlying the cardioprotective effects of FGF21. We engineered FGF21 knock out mice and subsequently elucidated the effects of FGF21 and its downstream mediators using western blotting, qRT-PCR, and mitochondrial morphological and functional analyses. FGF21 knockout mice showed cardiac dysfunction, accompanied by a decline in global longitudinal strain (GLS) and ejection fraction (EF), independent of metabolic disorders. Mitochondrial quality, quantity, and function were abnormal, accompanied by decreased levels of optic atrophy-1 (OPA1) in FGF21 KO mice. In contrast to FGF21 knockout, cardiac-specific overexpression of FGF21 alleviated the cardiac dysfunction caused by FGF21 deficiency. In an in vitro study, FGF21 siRNA deteriorated mitochondrial dynamics and impaired function induced by cobalt chloride (CoCl2). Both recombinant FGF21 and adenovirus-mediated FGF21 overexpression could alleviate CoCl2-induced mitochondrial impairment by restoring mitochondrial dynamics. FGF21 was essential for maintaining mitochondrial dynamics and function of the cardiomyocytes. As a regulator of cardiomyocyte mitochondrial homeostasis under oxidative stress, FGF21 could be an important new target for therapeutic options for patients with heart failure.
Collapse
Affiliation(s)
- Bing Yan
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China
| | - Zhu Mei
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Yaohan Tang
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Haixu Song
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Hanlin Wu
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Quanmin Jing
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Xiaolin Zhang
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Chenghui Yan
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China.
| | - Yaling Han
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016, China.
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
27
|
Purinergic receptor: a crucial regulator of adipose tissue functions. Purinergic Signal 2023; 19:273-281. [PMID: 36515790 PMCID: PMC9984650 DOI: 10.1007/s11302-022-09907-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/14/2022] [Indexed: 12/15/2022] Open
Abstract
Obesity is a public-health challenge resulting from an imbalance between energy expenditure and calorie intake. This health problem exacerbates a variety of metabolic complications worldwide. Adipose tissue is an essential regulator of energy homeostasis, and the functions within it are regulated by purinergic receptors. A1R, P2X7R, and P2YR mainly mediate energy homeostasis primarily through regulating energy storage and adipokines secretion in white adipose tissue (WAT). P2X5R is a novel-specific cell surface marker in brown/beige adipocytes. A2R is a promising therapeutic target for stimulating energy expenditure in brown adipose tissue (BAT) and also mediating WAT browning. Based on these features, purinergic receptors may be an appropriate target in treating obesity. In this review, the role of purinergic receptors in different types of adipose tissue is summarized. An improved understanding of purinergic receptor functions in adipose tissue may lead to more effective treatment interventions for obesity and its related metabolic disorders.
Collapse
|
28
|
Gu X, Wang L, Liu S, Shan T. Adipose tissue adipokines and lipokines: Functions and regulatory mechanism in skeletal muscle development and homeostasis. Metabolism 2023; 139:155379. [PMID: 36538987 DOI: 10.1016/j.metabol.2022.155379] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/29/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Skeletal muscle plays important roles in normal biological activities and whole-body energy homeostasis in humans. The growth and development of skeletal muscle also directly influence meat production and meat quality in animal production. Therefore, regulating the development and homeostasis of skeletal muscle is crucial for human health and animal production. Adipose tissue, which includes white adipose tissue (WAT) and brown adipose tissue (BAT), not only functions as an energy reserve but also has attracted substantial attention because of its role as an endocrine organ. The novel signalling molecules known as "adipokines" and "lipokines" that are secreted by adipose tissue were identified through the secretomic technique, which broadened our understanding of the previously unknown crosstalk between adipose tissue and skeletal muscle. In this review, we summarize and discuss the secretory role of adipose tissues, both WAT and BAT, as well as the regulatory roles of various adipokines and lipokines in skeletal muscle development and homeostasis. We suggest that adipokines and lipokines have potential as drug candidates for the treatment of skeletal muscle dysfunction and related metabolic diseases and as promising nutrients for improving animal production.
Collapse
Affiliation(s)
- Xin Gu
- College of Animal Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Shiqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
29
|
Saito M, Okamatsu-Ogura Y. Thermogenic Brown Fat in Humans: Implications in Energy Homeostasis, Obesity and Metabolic Disorders. World J Mens Health 2023:41.e26. [PMID: 36792089 DOI: 10.5534/wjmh.220224] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/08/2022] [Indexed: 01/27/2023] Open
Abstract
In mammals including humans, there are two types of adipose tissue, white and brown adipose tissues (BATs). White adipose tissue is the primary site of energy storage, while BAT is a specialized tissue for non-shivering thermogenesis to dissipate energy as heat. Although BAT research has long been limited mostly in small rodents, the rediscovery of metabolically active BAT in adult humans has dramatically promoted the translational studies on BAT in health and diseases. It is now established that BAT, through its thermogenic and energy dissipating activities, plays a role in the regulation of body temperature, whole-body energy expenditure, and body fatness. Moreover, increasing evidence has demonstrated that BAT secretes various paracrine and endocrine factors, which influence other peripheral tissues and control systemic metabolic homeostasis, suggesting BAT as a metabolic regulator, other than for thermogenesis. In fact, clinical studies have revealed an association of BAT not only with metabolic disorders such as insulin resistance, diabetes, dyslipidemia, and fatty liver, but also with cardiovascular diseases including hypertension and atherosclerosis. Thus, BAT is an intriguing tissue combating obesity and related metabolic diseases. In this review, we summarize current knowledge on human BAT, focusing its patho-physiological roles in energy homeostasis, obesity and related metabolic disorders. The effects of aging and sex on BAT are also discussed.
Collapse
Affiliation(s)
- Masayuki Saito
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
30
|
Kim K, Im H, Son Y, Kim M, Tripathi SK, Jeong LS, Lee YH. Anti-obesity effects of the dual-active adenosine A 2A/A 3 receptor-ligand LJ-4378. Int J Obes (Lond) 2022; 46:2128-2136. [PMID: 36167764 DOI: 10.1038/s41366-022-01224-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND OBJECTIVES A2A adenosine receptor (A2AAR)-mediated signaling in adipose tissues has been investigated as a potential target for obesity-related metabolic diseases. LJ-4378 has been developed as a dual-acting ligand with A2AAR agonist and A3 adenosine receptor (A3AR) antagonist activity. The current study aimed to investigate the anti-obesity effects of LJ-4378 and its underlying molecular mechanisms. METHODS Immortalized brown adipocytes were used for in vitro analysis. A high-fat diet (HFD)-induced obesity and cell death-inducing DFFA-like effector A reporter mouse models were used for in vivo experiments. The effects of LJ-4378 on lipolysis and mitochondrial metabolism were evaluated using immunoblotting, mitochondrial staining, and oxygen consumption rate analyses. The in vivo anti-obesity effects of LJ-4378 were evaluated using indirect calorimetry, body composition analyses, glucose tolerance tests, and histochemical analyses. RESULTS In vitro LJ-4378 treatment increased the levels of brown adipocyte markers and mitochondrial proteins, including uncoupling protein 1. The effects of LJ-4378 on lipolysis of adipocytes were more potent than those of the A2AAR agonist or A3AR antagonist. In vivo, LJ-4378 treatment increased energy expenditure by 17.0% (P value < 0.0001) compared to vehicle controls. LJ-4378 (1 mg/kg, i.p.) treatment for 10 days reduced body weight and fat content by 8.24% (P value < 0.0001) and 24.2% (P value = 0.0044), respectively, and improved glucose tolerance in the HFD-fed mice. LJ-4378 increased the expression levels of brown adipocyte markers and mitochondrial proteins in interscapular brown and inguinal white adipose tissue. CONCLUSION These findings support the in vivo anti-obesity effects of LJ-4378, and suggest a novel therapeutic approach to combat obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Kyungmin Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Hyeonyeong Im
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Yeonho Son
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Minjae Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Sushil Kumar Tripathi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Lak Shin Jeong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
31
|
Abstract
It is important to understand how different human organs coordinate and interact with each other. Since obesity and cardiac disease frequently coincide, the crosstalk between adipose tissues and heart has drawn attention. We appreciate that specific peptides/proteins, lipids, nucleic acids, and even organelles shuttle between the adipose tissues and heart. These bioactive components can profoundly affect the metabolism of cells in distal organs, including heart. Importantly, this process can be dysregulated under pathophysiological conditions. This also opens the door to efforts targeting these mediators as potential therapeutic strategies to treat patients who manifest diabetes and cardiovascular disease. Here, we summarize the recent progress toward a better understanding of how the adipose tissues and heart interact with each other.
Collapse
|
32
|
Yin X, Chen Y, Ruze R, Xu R, Song J, Wang C, Xu Q. The evolving view of thermogenic fat and its implications in cancer and metabolic diseases. Signal Transduct Target Ther 2022; 7:324. [PMID: 36114195 PMCID: PMC9481605 DOI: 10.1038/s41392-022-01178-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 02/07/2023] Open
Abstract
AbstractThe incidence of metabolism-related diseases like obesity and type 2 diabetes mellitus has reached pandemic levels worldwide and increased gradually. Most of them are listed on the table of high-risk factors for malignancy, and metabolic disorders systematically or locally contribute to cancer progression and poor prognosis of patients. Importantly, adipose tissue is fundamental to the occurrence and development of these metabolic disorders. White adipose tissue stores excessive energy, while thermogenic fat including brown and beige adipose tissue dissipates energy to generate heat. In addition to thermogenesis, beige and brown adipocytes also function as dynamic secretory cells and a metabolic sink of nutrients, like glucose, fatty acids, and amino acids. Accordingly, strategies that activate and expand thermogenic adipose tissue offer therapeutic promise to combat overweight, diabetes, and other metabolic disorders through increasing energy expenditure and enhancing glucose tolerance. With a better understanding of its origins and biological functions and the advances in imaging techniques detecting thermogenesis, the roles of thermogenic adipose tissue in tumors have been revealed gradually. On the one hand, enhanced browning of subcutaneous fatty tissue results in weight loss and cancer-associated cachexia. On the other hand, locally activated thermogenic adipocytes in the tumor microenvironment accelerate cancer progression by offering fuel sources and is likely to develop resistance to chemotherapy. Here, we enumerate current knowledge about the significant advances made in the origin and physiological functions of thermogenic fat. In addition, we discuss the multiple roles of thermogenic adipocytes in different tumors. Ultimately, we summarize imaging technologies for identifying thermogenic adipose tissue and pharmacologic agents via modulating thermogenesis in preclinical experiments and clinical trials.
Collapse
|
33
|
Niemann B, Haufs-Brusberg S, Puetz L, Feickert M, Jaeckstein MY, Hoffmann A, Zurkovic J, Heine M, Trautmann EM, Müller CE, Tönjes A, Schlein C, Jafari A, Eltzschig HK, Gnad T, Blüher M, Krahmer N, Kovacs P, Heeren J, Pfeifer A. Apoptotic brown adipocytes enhance energy expenditure via extracellular inosine. Nature 2022; 609:361-368. [PMID: 35790189 PMCID: PMC9452294 DOI: 10.1038/s41586-022-05041-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 06/29/2022] [Indexed: 11/09/2022]
Abstract
Brown adipose tissue (BAT) dissipates energy1,2 and promotes cardiometabolic health3. Loss of BAT during obesity and ageing is a principal hurdle for BAT-centred obesity therapies, but not much is known about BAT apoptosis. Here, untargeted metabolomics demonstrated that apoptotic brown adipocytes release a specific pattern of metabolites with purine metabolites being highly enriched. This apoptotic secretome enhances expression of the thermogenic programme in healthy adipocytes. This effect is mediated by the purine inosine that stimulates energy expenditure in brown adipocytes by the cyclic adenosine monophosphate-protein kinase A signalling pathway. Treatment of mice with inosine increased BAT-dependent energy expenditure and induced 'browning' of white adipose tissue. Mechanistically, the equilibrative nucleoside transporter 1 (ENT1, SLC29A1) regulates inosine levels in BAT: ENT1-deficiency increases extracellular inosine levels and consequently enhances thermogenic adipocyte differentiation. In mice, pharmacological inhibition of ENT1 as well as global and adipose-specific ablation enhanced BAT activity and counteracted diet-induced obesity, respectively. In human brown adipocytes, knockdown or blockade of ENT1 increased extracellular inosine, which enhanced thermogenic capacity. Conversely, high ENT1 levels correlated with lower expression of the thermogenic marker UCP1 in human adipose tissues. Finally, the Ile216Thr loss of function mutation in human ENT1 was associated with significantly lower body mass index and 59% lower odds of obesity for individuals carrying the Thr variant. Our data identify inosine as a metabolite released during apoptosis with a 'replace me' signalling function that regulates thermogenic fat and counteracts obesity.
Collapse
Affiliation(s)
- Birte Niemann
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany.
| | - Saskia Haufs-Brusberg
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Laura Puetz
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Martin Feickert
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Michelle Y Jaeckstein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Jelena Zurkovic
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva-Maria Trautmann
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
| | - Christa E Müller
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Christian Schlein
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Azin Jafari
- Clinic and Polyclinic for General, Visceral, Thoracic and Vascular Surgery, University Hospital, University of Bonn, Bonn, Germany
| | - Holger K Eltzschig
- Department of Anesthesiology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany.
- PharmaCenter Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
34
|
Pinckard KM, Stanford KI. The Heartwarming Effect of Brown Adipose Tissue. Mol Pharmacol 2022; 102:460-471. [PMID: 34933905 PMCID: PMC9341250 DOI: 10.1124/molpharm.121.000328] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/27/2021] [Indexed: 11/22/2022] Open
Abstract
Brown adipose tissue (BAT) is a metabolically active tissue that improves glucose metabolism and protects against the development of type 2 diabetes and obesity. However, the role of BAT to improve cardiovascular health has only recently been investigated. In this review, we discuss multiple mechanisms through which both the thermogenic and endocrine functions of BAT mediate cardiac health. β-adrenergic stimulation activates the thermogenic function of BAT, resulting in reduced circulating lipids and glucose, and enhanced clearance of hepatic cholesterol-enriched remnants leading to reduced atherosclerotic region size. Additionally, the thermogenic role of BAT has been implicated in activation of the protein kinase B-extracellular-signal-regulated kinase (ERK) 1/2 pathway after myocardial infarction (MI), contributing to reduced injury size. The endocrine function of BAT has also been implicated to improve both systemic metabolic health and cardiac health. Specifically, the batokines fibroblast growth factor 21 (FGF21) and 12,13-diHOME improve cardiovascular health via reduced hypertension, hypertrophy and MI injury size (FGF21) or by directly improving cardiac function via calcium cycling (12,13-diHOME). Finally, we discuss relevant pharmacological treatment methods currently aiming to activate BAT, typically through sympathetic activation. SIGNIFICANCE STATEMENT: This mini-review discusses the role of BAT to improve cardiac health via thermogenic and endocrine effects in both rodents and humans and highlights the need for therapeutic methods which activate or mimic BAT activity.
Collapse
Affiliation(s)
- Kelsey M Pinckard
- Department of Physiology and Cell Biology (K.M.P., K.I.S.), Center for Diabetes and Metabolism Research Center, Dorothy M. Davis Heart and Lung Research Institute (K.M.P., K.I.S.), and Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (K.I.S.)
| | - Kristin I Stanford
- Department of Physiology and Cell Biology (K.M.P., K.I.S.), Center for Diabetes and Metabolism Research Center, Dorothy M. Davis Heart and Lung Research Institute (K.M.P., K.I.S.), and Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (K.I.S.)
| |
Collapse
|
35
|
Yanucil C, Kentrup D, Li X, Grabner A, Schramm K, Martinez EC, Li J, Campos I, Czaya B, Heitman K, Westbrook D, Wende AR, Sloan A, Roche JM, Fornoni A, Kapiloff MS, Faul C. FGF21-FGFR4 signaling in cardiac myocytes promotes concentric cardiac hypertrophy in mouse models of diabetes. Sci Rep 2022; 12:7326. [PMID: 35513431 PMCID: PMC9072546 DOI: 10.1038/s41598-022-11033-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 04/18/2022] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor (FGF) 21, a hormone that increases insulin sensitivity, has shown promise as a therapeutic agent to improve metabolic dysregulation. Here we report that FGF21 directly targets cardiac myocytes by binding β-klotho and FGF receptor (FGFR) 4. In combination with high glucose, FGF21 induces cardiac myocyte growth in width mediated by extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. While short-term FGF21 elevation can be cardio-protective, we find that in type 2 diabetes (T2D) in mice, where serum FGF21 levels are elevated, FGFR4 activation induces concentric cardiac hypertrophy. As T2D patients are at risk for heart failure with preserved ejection fraction (HFpEF), we propose that induction of concentric hypertrophy by elevated FGF21-FGFR4 signaling may constitute a novel mechanism promoting T2D-associated HFpEF such that FGFR4 blockade might serve as a cardio-protective therapy in T2D. In addition, potential adverse cardiac effects of FGF21 mimetics currently in clinical trials should be investigated.
Collapse
Affiliation(s)
- Christopher Yanucil
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Dominik Kentrup
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, USA
| | - Xueyi Li
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, 1651 Page Mill Road, Mail Code 5356, Palo Alto, CA, USA
| | - Alexander Grabner
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Karla Schramm
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Eliana C Martinez
- Department of Pediatrics and Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, FL, Miami, USA
| | - Jinliang Li
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, 1651 Page Mill Road, Mail Code 5356, Palo Alto, CA, USA
- Department of Pediatrics and Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, FL, Miami, USA
| | - Isaac Campos
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Brian Czaya
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Kylie Heitman
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - David Westbrook
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Adam R Wende
- Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexis Sloan
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Johanna M Roche
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alessia Fornoni
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Michael S Kapiloff
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, 1651 Page Mill Road, Mail Code 5356, Palo Alto, CA, USA.
- Department of Pediatrics and Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, FL, Miami, USA.
| | - Christian Faul
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA.
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
36
|
Li X, Zhang Y, Su L, Cai L, Zhang C, Zhang J, Sun J, Chai M, Cai M, Wu Q, Zhang C, Yan X, Wang L, Huang X. FGF21 alleviates pulmonary hypertension by inhibiting mTORC1/EIF4EBP1 pathway via H19. J Cell Mol Med 2022; 26:3005-3021. [PMID: 35437883 PMCID: PMC9097832 DOI: 10.1111/jcmm.17318] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 11/30/2022] Open
Abstract
Long non‐coding RNAs (lncRNAs) play a significant role in pulmonary hypertension (PH). Our preliminary data showed that hypoxia‐induced PH is attenuated by fibroblast growth factor 21 (FGF21) administration. Therefore, we further investigated the regulatory role of long non‐coding RNAs in PH treated with FGF21. RNA sequencing analysis and real‐time PCR identified a significantly up‐regulation of the H19 after FGF21 administration. Moreover, gain‐ and loss‐of‐function assays demonstrated that FGF21 suppressed hypoxia‐induced proliferation of pulmonary artery smooth muscle cells partially through upregulation of H19. In addition, FGF21 deficiency markedly exacerbated hypoxia‐induced increases of pulmonary artery pressure and pulmonary vascular remodelling. In addition, AAV‐mediated H19 overexpression reversed the malignant phenotype of FGF21 knockout mice under hypoxia expose. Further investigation uncovered that H19 also acted as an orchestra conductor that inhibited the function of mechanistic target of rapamycin complex 1 (mTORC1) by disrupting the interaction of mTORC1 with eukaryotic translation initiation factor 4E–binding protein 1 (EIF4EBP1). Our work highlights the important role of H19 in PH treated with FGF21 and suggests a mechanism involving mTORC1/EIF4EBP1 inhibition, which may provide a fundamental for clinical application of FGF21 in PH.
Collapse
Affiliation(s)
- Xiuchun Li
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Yaxin Zhang
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Lihuang Su
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Luqiong Cai
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Chi Zhang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, P.R. China
| | - Jianhao Zhang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, P.R. China
| | - Junwei Sun
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Mengyu Chai
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Mengsi Cai
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Qian Wu
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Chi Zhang
- Chinese-American Research Institute for Diabetic Complications at Department of Pharmacy, Wenzhou Medical University, Wenzhou, P.R. China
| | - Xiaoqing Yan
- Chinese-American Research Institute for Diabetic Complications at Department of Pharmacy, Wenzhou Medical University, Wenzhou, P.R. China
| | - Liangxing Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, P.R. China
| |
Collapse
|
37
|
Zhao H, Chen X, Hu G, Li C, Guo L, Zhang L, Sun F, Xia Y, Yan W, Cui Z, Guo Y, Guo X, Huang C, Fan M, Wang S, Zhang F, Tao L. Small Extracellular Vesicles From Brown Adipose Tissue Mediate Exercise Cardioprotection. Circ Res 2022; 130:1490-1506. [PMID: 35387487 DOI: 10.1161/circresaha.121.320458] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Long-term exercise provides reliable cardioprotection via mechanisms still incompletely understood. Although traditionally considered a thermogenic tissue, brown adipose tissue (BAT) communicates with remote organs (eg, the heart) through its endocrine function. BAT expands in response to exercise, but its involvement in exercise cardioprotection remains undefined. OBJECTIVE This study investigated whether small extracellular vesicles (sEVs) secreted by BAT and their contained microRNAs (miRNAs) regulate cardiomyocyte survival and participate in exercise cardioprotection in the context of myocardial ischemia/reperfusion (MI/R) injury. METHODS AND RESULTS Four weeks of exercise resulted in a significant BAT expansion in mice. Surgical BAT ablation before MI/R weakened the salutary effects of exercise. Adeno-associated virus 9 vectors carrying short hairpin RNA targeting Rab27a (a GTPase required for sEV secretion) or control viruses were injected in situ into the interscapular BAT. Exercise-mediated protection against MI/R injury was greatly attenuated in mice whose BAT sEV secretion was suppressed by Rab27a silencing. Intramyocardial injection of the BAT sEVs ameliorated MI/R injury, revealing the cardioprotective potential of BAT sEVs. Discovery-driven experiments identified miR-125b-5p, miR-128-3p, and miR-30d-5p (referred to as the BAT miRNAs) as essential BAT sEV components for mediating cardioprotection. BAT-specific inhibition of the BAT miRNAs prevented their upregulation in plasma sEVs and hearts of exercised mice and attenuated exercise cardioprotection. Mechanistically, the BAT miRNAs cooperatively suppressed the proapoptotic MAPK (mitogen-associated protein kinase) pathway by targeting a series of molecules (eg, Map3k5, Map2k7, and Map2k4) in the signaling cascade. Delivery of BAT sEVs into hearts or cardiomyocytes suppressed MI/R-related MAPK pathway activation, an effect that disappeared with the combined use of the BAT miRNA inhibitors. CONCLUSIONS The sEVs secreted by BAT participate in exercise cardioprotection via delivering the cardioprotective miRNAs into the heart. These results provide novel insights into the mechanisms underlying the BAT-cardiomyocyte interaction and highlight BAT sEVs and their contained miRNAs as alternative candidates for exercise cardioprotection.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Department of Pharmacy, the 960th Hospital of the Logistics Support Force, Jinan, China (H.Z.)
| | - Xiyao Chen
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Department of Geriatrics, The Fourth Military Medical University, Xi'an, China. (X.C.)
| | - Guangyu Hu
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Congye Li
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Lanyan Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ling Zhang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Fangfang Sun
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Yunlong Xia
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Wenjun Yan
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ze Cui
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Yongzhen Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Xijing Hospital and Department of Toxicology, School of Public Health, The Fourth Military Medical University, Xi'an, China. (Y.G.)
| | - Xiong Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Chong Huang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Miaomiao Fan
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Shan Wang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Fuyang Zhang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ling Tao
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW This review highlights aspects of brown adipose tissue (BAT) communication with other organ systems and how BAT-to-tissue cross-talk could help elucidate future obesity treatments. RECENT FINDINGS Until recently, research on BAT has focused mainly on its thermogenic activity. New research has identified an endocrine/paracrine function of BAT and determined that many BAT-derived molecules, termed "batokines," affect the physiology of a variety of organ systems and cell types. Batokines encompass a variety of signaling molecules including peptides, metabolites, lipids, or microRNAs. Recent studies have noted significant effects of batokines on physiology as it relates whole-body metabolism and cardiac function. This review will discuss batokines and other BAT processes that affect the liver, cardiovascular system, skeletal muscle, immune cells, and brown and white adipose tissue. Brown adipose tissue has a crucial secretory function that plays a key role in systemic physiology.
Collapse
Affiliation(s)
- Felix T Yang
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, 460 W. 12th Ave, Columbus, OH, 43210, USA
- Diabetes and Metabolism Research Center, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, 460 W. 12th Ave, Columbus, OH, 43210, USA.
- Diabetes and Metabolism Research Center, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
39
|
Sommakia S, Almaw NH, Lee SH, Ramadurai DKA, Taleb I, Kyriakopoulos CP, Stubben CJ, Ling J, Campbell RA, Alharethi RA, Caine WT, Navankasattusas S, Hoareau GL, Abraham AE, Fang JC, Selzman CH, Drakos SG, Chaudhuri D. FGF21 (Fibroblast Growth Factor 21) Defines a Potential Cardiohepatic Signaling Circuit in End-Stage Heart Failure. Circ Heart Fail 2022; 15:e008910. [PMID: 34865514 PMCID: PMC8930477 DOI: 10.1161/circheartfailure.121.008910] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Extrinsic control of cardiomyocyte metabolism is poorly understood in heart failure (HF). FGF21 (Fibroblast growth factor 21), a hormonal regulator of metabolism produced mainly in the liver and adipose tissue, is a prime candidate for such signaling. METHODS To investigate this further, we examined blood and tissue obtained from human subjects with end-stage HF with reduced ejection fraction at the time of left ventricular assist device implantation and correlated serum FGF21 levels with cardiac gene expression, immunohistochemistry, and clinical parameters. RESULTS Circulating FGF21 levels were substantially elevated in HF with reduced ejection fraction, compared with healthy subjects (HF with reduced ejection fraction: 834.4 [95% CI, 628.4-1040.3] pg/mL, n=40; controls: 146.0 [86.3-205.7] pg/mL, n=20, P=1.9×10-5). There was clear FGF21 staining in diseased cardiomyocytes, and circulating FGF21 levels negatively correlated with the expression of cardiac genes involved in ketone metabolism, consistent with cardiac FGF21 signaling. FGF21 gene expression was very low in failing and nonfailing hearts, suggesting extracardiac production of the circulating hormone. Circulating FGF21 levels were correlated with BNP (B-type natriuretic peptide) and total bilirubin, markers of chronic cardiac and hepatic congestion. CONCLUSIONS Circulating FGF21 levels are elevated in HF with reduced ejection fraction and appear to bind to the heart. The liver is likely the main extracardiac source. This supports a model of hepatic FGF21 communication to diseased cardiomyocytes, defining a potential cardiohepatic signaling circuit in human HF.
Collapse
Affiliation(s)
- Salah Sommakia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Naredos H. Almaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Sandra H. Lee
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Dinesh K. A. Ramadurai
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Iosif Taleb
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Christos P. Kyriakopoulos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Chris J. Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Jing Ling
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Robert A. Campbell
- Department of Internal Medicine, Division of General Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Rami A. Alharethi
- U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake Veterans Affairs Health Care System, Salt Lake City, UT
| | - William T. Caine
- U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake Veterans Affairs Health Care System, Salt Lake City, UT
| | - Sutip Navankasattusas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Guillaume L. Hoareau
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Department of Surgery, Division of Emergency Medicine, University of Utah, Salt Lake City, UT, USA
| | - Anu E. Abraham
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT
| | - James C. Fang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT
| | - Craig H. Selzman
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake Veterans Affairs Health Care System, Salt Lake City, UT
- Department of Surgery, Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, UT
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
40
|
Docosahexaenoic Acid-Enhanced Autophagic Flux Improves Cardiac Dysfunction after Myocardial Infarction by Targeting the AMPK/mTOR Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1509421. [PMID: 35265261 PMCID: PMC8898772 DOI: 10.1155/2022/1509421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/28/2022] [Accepted: 02/06/2022] [Indexed: 11/17/2022]
Abstract
Background and Purpose. Docosahexaenoic acid (DHA) is a type of polyunsaturated fatty acid enriched in cod liver oil and seaweed. It is necessary for the human body and has important functions, such as antioxidation and antiatherosclerosis activities. Long-term oral administration of DHA or the use of DHA at the initial stage of ischemia can increase the level of autophagy and exert a protective effect on neurological functions related to cerebral infarction. However, the effect of DHA on myocardial injury and cardiac insufficiency after myocardial infarction (MI) is unknown. This study was aimed at exploring whether DHA plays a protective role in AMI and its specific molecular mechanism. Experimental Method. In vitro cardiomyocyte hypoxia and in vivo MI injury models were used to determine the role of DHA in MI. Hypoxic injury induced damage in cultured neonatal mouse cardiomyocytes (NMCs). The C57BL/6J mouse MI model was established by permanent ligation of the left anterior descending branch. Main Results. DHA improved the cardiomyocyte viability of NMCs induced by hypoxia injury and reduced cell necrosis. DHA reduced infarct size, improved heart function, and reduced the degree of myocardial fibrosis in mice after MI. In addition, DHA enhanced autophagy flux and reduced apoptosis in vitro and in vivo. In addition, we found that chloroquine, an autophagy inhibitor, blocked the protective effect of DHA on cardiomyocyte apoptosis and cardiac dysfunction, indicating that DHA exerts cardioprotective effects in part by promoting autophagy flux. We also observed that DHA enhanced autophagy flux by activating the AMPK/mTOR signaling pathway. Conclusions and Significance. In conclusion, our findings indicate for the first time that DHA improves MI-induced cardiac dysfunction by promoting AMPK/mTOR-mediated autophagic flux.
Collapse
|
41
|
Gavaldà-Navarro A, Villarroya J, Cereijo R, Giralt M, Villarroya F. The endocrine role of brown adipose tissue: An update on actors and actions. Rev Endocr Metab Disord 2022; 23:31-41. [PMID: 33712997 DOI: 10.1007/s11154-021-09640-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
In recent years, brown adipose tissue (BAT) has been recognized not only as a main site of non-shivering thermogenesis in mammals, but also as an endocrine organ. BAT secretes a myriad of regulatory factors. These so-called batokines exert local autocrine and paracrine effects, as well as endocrine actions targeting tissues and organs at a distance. The endocrine batokines include peptide factors, such as fibroblast growth factor-21 (FGF21), neuregulin-4 (NRG4), phospholipid transfer protein (PLTP), interleukin-6, adiponectin and myostatin, and also lipids (lipokines; e.g., 12,13-dihydroxy-9Z-octadecenoic acid [12,13-diHOME]) and miRNAs (e.g., miR-99b). The liver, heart, and skeletal muscle are the most commonly reported targets of batokines. In response to BAT thermogenic activation, batokines such as NRG4 and PLTP are released and act to reduce hepatic steatosis and improve insulin sensitivity. Stress-induced interleukin-6-mediated signaling from BAT to liver favors hepatic glucose production through enhanced gluconeogenesis. Batokines may act on liver to induce the secretion of regulatory hepatokines (e.g. FGF21 and bile acids in response to miR-99b and PLTP, respectively), thereby resulting in a systemic expansion of BAT-originating signals. Batokines also target extrahepatic tissues: FGF21 and 12,13-diHOME are cardioprotective, whereas BAT-secreted myostatin and 12,13-diHOME influence skeletal muscle development and performance. Further research is needed to ascertain in humans the role of batokines, which have been identified mostly in experimental models. The endocrine role of BAT may explain the association between active BAT and a healthy metabolism in the human system, which is characterized by small amounts of BAT and a likely moderate BAT-mediated energy expenditure.
Collapse
Affiliation(s)
- Aleix Gavaldà-Navarro
- Departament de Bioquimica I Biomedicina Molecular, and Institut de Biomedicina de La Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues, Catalonia, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición, Madrid, Spain
| | - Joan Villarroya
- Departament de Bioquimica I Biomedicina Molecular, and Institut de Biomedicina de La Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues, Catalonia, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición, Madrid, Spain
| | - Rubén Cereijo
- Departament de Bioquimica I Biomedicina Molecular, and Institut de Biomedicina de La Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición, Madrid, Spain
- Institut de Recerca Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Marta Giralt
- Departament de Bioquimica I Biomedicina Molecular, and Institut de Biomedicina de La Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues, Catalonia, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición, Madrid, Spain
| | - Francesc Villarroya
- Departament de Bioquimica I Biomedicina Molecular, and Institut de Biomedicina de La Universitat de Barcelona, Barcelona, Catalonia, Spain.
- Institut de Recerca Sant Joan de Déu, Esplugues, Catalonia, Spain.
- CIBER Fisiopatología de La Obesidad Y Nutrición, Madrid, Spain.
| |
Collapse
|
42
|
Attenuation of Tumor Development in Mammary Carcinoma Rats by Theacrine, an Antagonist of Adenosine 2A Receptor. Molecules 2021; 26:molecules26247455. [PMID: 34946538 PMCID: PMC8706909 DOI: 10.3390/molecules26247455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Caffeine has been reported to induce anti-tumor immunity for attenuating breast cancer by blocking the adenosine 2A receptor. Molecular modeling showed that theacrine, a purine alkaloid structurally similar to caffeine, might be an antagonist of the adenosine 2A receptor equivalent to or more effective than caffeine. Theacrine was further demonstrated to be an effective antagonist of the adenosine 2A receptor as its concurrent supplementation significantly reduced the elevation of AMPK phosphorylation level in MCF-7 human breast cells induced by CGS21680, an agonist of adenosine 2A receptors. In an animal model, the development of mammary carcinoma induced by 7,12-Dimethylbenz[a]anthracene in Sprague–Dawley rats could be attenuated by daily supplement of theacrine of 50 or 100 mg/kg body weight. Both expression levels of cleaved-caspase-3/pro-caspase-3 and granzyme B in tumor tissues were significantly elevated when theacrine was supplemented, indicating the induction of programmed cell death in tumor cells might be involved in the attenuation of mammary carcinoma. Similar to the caffeine, significant elevation of interferon-γ and tumor necrosis factor-α was observed in the serum and tumor tissues of rats after the theacrine supplement of 50 mg/kg body weight. Taken together, theacrine is an effective antagonist of adenosine 2A receptors and possesses great potential to be used to attenuate breast cancer.
Collapse
|
43
|
Zhang Y, Liu D, Long XX, Fang QC, Jia WP, Li HT. The role of FGF21 in the pathogenesis of cardiovascular disease. Chin Med J (Engl) 2021; 134:2931-2943. [PMID: 34939977 PMCID: PMC8710326 DOI: 10.1097/cm9.0000000000001890] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Indexed: 12/16/2022] Open
Abstract
ABSTRACT The morbidity and mortality of cardiovascular diseases (CVDs) are increasing worldwide and seriously threaten human life and health. Fibroblast growth factor 21 (FGF21), a metabolic regulator, regulates glucose and lipid metabolism and may exert beneficial effects on the cardiovascular system. In recent years, FGF21 has been found to act directly on the cardiovascular system and may be used as an early biomarker of CVDs. The present review highlights the recent progress in understanding the relationship between FGF21 and CVDs including coronary heart disease, myocardial ischemia, cardiomyopathy, and heart failure and also explores the related mechanism of the cardioprotective effect of FGF21. FGF21 plays an important role in the prediction, treatment, and improvement of prognosis in CVDs. This cardioprotective effect of FGF21 may be achieved by preventing endothelial dysfunction and lipid accumulating, inhibiting cardiomyocyte apoptosis and regulating the associated oxidative stress, inflammation and autophagy. In conclusion, FGF21 is a promising target for the treatment of CVDs, however, its clinical application requires further clarification of the precise role of FGF21 in CVDs.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
- Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Dan Liu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
- Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiao-Xue Long
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
- Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qi-Chen Fang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Wei-Ping Jia
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Hua-Ting Li
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
44
|
Shamsi F, Wang CH, Tseng YH. The evolving view of thermogenic adipocytes - ontogeny, niche and function. Nat Rev Endocrinol 2021; 17:726-744. [PMID: 34625737 PMCID: PMC8814904 DOI: 10.1038/s41574-021-00562-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/12/2022]
Abstract
The worldwide incidence of obesity and its sequelae, such as type 2 diabetes mellitus, have reached pandemic levels. Central to the development of these metabolic disorders is adipose tissue. White adipose tissue stores excess energy, whereas brown adipose tissue (BAT) and beige (also known as brite) adipose tissue dissipate energy to generate heat in a process known as thermogenesis. Strategies that activate and expand BAT and beige adipose tissue increase energy expenditure in animal models and offer therapeutic promise to treat obesity. A better understanding of the molecular mechanisms underlying the development of BAT and beige adipose tissue and the activation of thermogenic function is the key to creating practical therapeutic interventions for obesity and metabolic disorders. In this Review, we discuss the regulation of the tissue microenvironment (the adipose niche) and inter-organ communication between BAT and other tissues. We also cover the activation of BAT and beige adipose tissue in response to physiological cues (such as cold exposure, exercise and diet). We highlight advances in harnessing the therapeutic potential of BAT and beige adipose tissue by genetic, pharmacological and cell-based approaches in obesity and metabolic disorders.
Collapse
Affiliation(s)
- Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Chih-Hao Wang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
45
|
Ma Y, Bhuiyan MS, Kim I, Tang X. Editorial: Metabolic Regulation in the Development of Cardiovascular Diseases. Front Cell Dev Biol 2021; 9:768689. [PMID: 34722552 PMCID: PMC8548466 DOI: 10.3389/fcell.2021.768689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/21/2021] [Indexed: 01/17/2023] Open
Affiliation(s)
- Yimei Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - InKyeom Kim
- Department of Pharmacology, Cardiovascular Research Institute, BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Yuko OO, Saito M. Brown Fat as a Regulator of Systemic Metabolism beyond Thermogenesis. Diabetes Metab J 2021; 45:840-852. [PMID: 34176254 PMCID: PMC8640153 DOI: 10.4093/dmj.2020.0291] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/26/2021] [Indexed: 12/01/2022] Open
Abstract
Brown adipose tissue (BAT) is a specialized tissue for nonshivering thermogenesis to dissipate energy as heat. Although BAT research has long been limited mostly in small rodents, the rediscovery of metabolically active BAT in adult humans has dramatically promoted the translational studies on BAT in health and diseases. Moreover, several remarkable advancements have been made in brown fat biology over the past decade: The molecular and functional analyses of inducible thermogenic adipocytes (socalled beige adipocytes) arising from a developmentally different lineage from classical brown adipocytes have been accelerated. In addition to a well-established thermogenic activity of uncoupling protein 1 (UCP1), several alternative thermogenic mechanisms have been discovered, particularly in beige adipocytes. It has become clear that BAT influences other peripheral tissues and controls their functions and systemic homeostasis of energy and metabolic substrates, suggesting BAT as a metabolic regulator, other than for thermogenesis. This notion is supported by discovering that various paracrine and endocrine factors are secreted from BAT. We review the current understanding of BAT pathophysiology, particularly focusing on its role as a metabolic regulator in small rodents and also in humans.
Collapse
Affiliation(s)
| | - Masayuki Saito
- Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Nutrition, Tenshi College, Sapporo, Japan
- Corresponding author: Masayuki Saito https://orcid.org/0000-0002-3058-3003 Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan E-mail:
| |
Collapse
|
47
|
Shaw A, Tóth BB, Király R, Arianti R, Csomós I, Póliska S, Vámos A, Korponay-Szabó IR, Bacso Z, Győry F, Fésüs L, Kristóf E. Irisin Stimulates the Release of CXCL1 From Differentiating Human Subcutaneous and Deep-Neck Derived Adipocytes via Upregulation of NFκB Pathway. Front Cell Dev Biol 2021; 9:737872. [PMID: 34708041 PMCID: PMC8542801 DOI: 10.3389/fcell.2021.737872] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Thermogenic brown and beige adipocytes might open up new strategies in combating obesity. Recent studies in rodents and humans have indicated that these adipocytes release cytokines, termed "batokines". Irisin was discovered as a polypeptide regulator of beige adipocytes released by myocytes, primarily during exercise. We performed global RNA sequencing on adipocytes derived from human subcutaneous and deep-neck precursors, which were differentiated in the presence or absence of irisin. Irisin did not exert an effect on the expression of characteristic thermogenic genes, while upregulated genes belonging to various cytokine signaling pathways. Out of the several upregulated cytokines, CXCL1, the highest upregulated, was released throughout the entire differentiation period, and predominantly by differentiated adipocytes. Deep-neck area tissue biopsies also showed a significant release of CXCL1 during 24 h irisin treatment. Gene expression data indicated upregulation of the NFκB pathway upon irisin treatment, which was validated by an increase of p50 and decrease of IκBα protein level, respectively. Continuous blocking of the NFκB pathway, using a cell permeable inhibitor of NFκB nuclear translocation, significantly reduced CXCL1 release. The released CXCL1 exerted a positive effect on the adhesion of endothelial cells. Together, our findings demonstrate that irisin stimulates the release of a novel adipokine, CXCL1, via upregulation of NFκB pathway in neck area derived adipocytes, which might play an important role in improving tissue vascularization.
Collapse
Affiliation(s)
- Abhirup Shaw
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Beáta B Tóth
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Róbert Király
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Rini Arianti
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - István Csomós
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Vámos
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Ilma R Korponay-Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Bacso
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Ferenc Győry
- Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Fésüs
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Endre Kristóf
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
48
|
MiR-21-3p Inhibits Adipose Browning by Targeting FGFR1 and Aggravates Atrial Fibrosis in Diabetes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9987219. [PMID: 34484568 PMCID: PMC8413063 DOI: 10.1155/2021/9987219] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/22/2021] [Accepted: 08/07/2021] [Indexed: 12/29/2022]
Abstract
A relationship between excess epicardial adipose tissue (EAT) and the risk of atrial fibrillation (AF) has been reported. Browning of EAT may be a novel approach for the prevention or treatment of AF by attenuating atrial fibrosis. Previous studies have identified microRNA-21 (miR-21) as a regulatory factor in atrial fibrosis. The present study examined the role of different subtypes of miR-21 in adipose browning and atrial fibrosis under hyperglycemic conditions. Wild type and miR-21 knockout C57BL/6 mice were used to establish a diabetic model via intraperitoneal injection of streptozotocin. A coculture model of atrial fibroblasts and adipocytes was also established. We identified miR-21-3p as a key regulator that controls adipocyte browning and participates in atrial fibrosis under hyperglycemic conditions. Moreover, fibroblast growth factor receptor (FGFR) 1, a direct target of miR-21-3p, decreased in this setting and controlled adipose browning. Gain and loss-of-function experiments identified a regulatory pathway in adipocytes involving miR-21a-3p, FGFR1, FGF21, and PPARγ that regulated adipocyte browning and participated in hyperglycemia-induced atrial fibrosis. Modulation of this signaling pathway may provide a therapeutic option for the prevention and treatment of atrial fibrosis or AF in DM.
Collapse
|
49
|
Cheng YW, Zhang ZB, Lan BD, Lin JR, Chen XH, Kong LR, Xu L, Ruan CC, Gao PJ. PDGF-D activation by macrophage-derived uPA promotes AngII-induced cardiac remodeling in obese mice. J Exp Med 2021; 218:e20210252. [PMID: 34236404 PMCID: PMC8273546 DOI: 10.1084/jem.20210252] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/03/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity-induced secretory disorder of adipose tissue-derived factors is important for cardiac damage. However, whether platelet-derived growth factor-D (PDGF-D), a newly identified adipokine, regulates cardiac remodeling in angiotensin II (AngII)-infused obese mice is unclear. Here, we found obesity induced PDGF-D expression in adipose tissue as well as more severe cardiac remodeling compared with control lean mice after AngII infusion. Adipocyte-specific PDGF-D knockout attenuated hypertensive cardiac remodeling in obese mice. Consistently, adipocyte-specific PDGF-D overexpression transgenic mice (PA-Tg) showed exacerbated cardiac remodeling after AngII infusion without high-fat diet treatment. Mechanistic studies indicated that AngII-stimulated macrophages produce urokinase plasminogen activator (uPA) that activates PDGF-D by splicing full-length PDGF-D into the active PDGF-DD. Moreover, bone marrow-specific uPA knockdown decreased active PDGF-DD levels in the heart and improved cardiac remodeling in HFD hypertensive mice. Together, our data provide for the first time a new interaction pattern between macrophage and adipocyte: that macrophage-derived uPA activates adipocyte-secreted PDGF-D, which finally accelerates AngII-induced cardiac remodeling in obese mice.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Bei Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei-Di Lan
- Department of Cardiology, First Affiliated Hospital, Xi’an Jiao Tong University, Xi’an, Shanxi, China
| | - Jing-Rong Lin
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Hui Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling-Ran Kong
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Xu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-Chao Ruan
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Zhang H, Liu M, Kim HT, Feeley BT, Liu X. Preconditioning improves muscle regeneration after ischemia-reperfusion injury. J Orthop Res 2021; 39:1889-1897. [PMID: 33232533 PMCID: PMC9257970 DOI: 10.1002/jor.24909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/19/2020] [Accepted: 11/08/2020] [Indexed: 02/04/2023]
Abstract
Ischemia-reperfusion injury (IRI) is a critical condition associated with serious clinical manifestations. Extensive research has focused on the strategies increasing organ tolerance to IRI. Preconditioning (PC) has been shown to provide protection to various organs toward IRI. However, the underlying mechanisms remain unknown. This study aimed to evaluate the role of PC on muscle regeneration after IRI and the potential underlying mechanisms. Three-month-old male UCP-1 reporter mice underwent unilateral hindlimb IRI with or without PC, the tissue viability and injury index were measured at 24 h after IRI. Hindlimb gait, muscle contractility, muscle histology were analyzed at 2 weeks after IRI. In another group of animals, β3 adrenergic receptor (β3AR) agonist amibegron and β3AR antagonist SR-59230A were administrated before PC/IRI, the hindlimb function and muscle regeneration were evaluated at 2 weeks after IRI. Our results showed that PC has little effect on improving the tissue viability at the acute phase of IRI, but it showed a long-term beneficial role of improving hindlimb function and muscle regeneration as evidenced by increased central nuclei regenerating myofibers. The effects of PC are related to inducing muscle fibro-adipogenic progenitor (FAP) brown/beige-like adipocyte (BAT) differentiation. Amibegron treatment displayed a similar role of PC while SR-59230A abolished the effect of PC. This study suggests PC has a beneficial role in promoting muscle regeneration after IRI through β3AR signaling pathway-stimulated FAP-BAT differentiation.
Collapse
Affiliation(s)
- He Zhang
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA,Department of Orthopedic Surgery, University of California-San Francisco, San Francisco, California, USA,Department of Exercise Physiology, Beijing Sports University, Beijing, China
| | - Mengyao Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA,Department of Orthopedic Surgery, University of California-San Francisco, San Francisco, California, USA
| | - Hubert T. Kim
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA,Department of Orthopedic Surgery, University of California-San Francisco, San Francisco, California, USA
| | - Brian T. Feeley
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA,Department of Orthopedic Surgery, University of California-San Francisco, San Francisco, California, USA
| | - Xuhui Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA,Department of Orthopedic Surgery, University of California-San Francisco, San Francisco, California, USA
| |
Collapse
|