1
|
McKenzie B, Peloquin M, Graves JL, Chen F, Tovar A, Carttar TA, Tucker K, Vo K, Nelson M, Super KM, Austriaco J, Weber SY, Naka A, McCandless EE, Greenwood K, Juarez-Salinas D, Halioua-Haubold CL, Ratcliff ER. Changes in insulin, adiponectin and lipid concentrations with age are associated with frailty and reduced quality of life in dogs. Sci Rep 2025; 15:5380. [PMID: 39948141 PMCID: PMC11825863 DOI: 10.1038/s41598-025-89923-z] [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: 11/21/2024] [Accepted: 02/10/2025] [Indexed: 02/16/2025] Open
Abstract
Declining metabolic function with aging is a conserved phenotype across many species. While aging-associated changes in metabolic status have been investigated rigorously in humans, less is known about metabolic aging in dogs. In this cross-sectional study, we aimed to examine changes in metabolic health with age, and any associations with frailty and quality of life, in a diverse population of companion dogs. This cross-sectional study enrolled 451 mature, adult companion dogs. Serum adiponectin, ALP, ALT, AST, cholesterol, insulin, IGF-1 and glucose levels were quantified. Additionally, plasma FFA, SFA, PA, OA and LA were quantified in a 61 dog subpopulation. All analytes were significantly associated with age, with the exception of AST. Elevated ALP, ALT, cholesterol, insulin, FFA, PA and OA were correlated with increased frailty scores, while higher levels of glucose and adiponectin were correlated with reduced frailty scores. The strength of these associations increased with age. Higher ALP, ALT and insulin were associated with lower HRQL scores after adjusting for covariates. Our findings establish novel associations between deleterious aging-associated metabolic changes and validated measures of clinical well-being in companion dogs. Future research should investigate the causality of these associations to inform therapeutic strategies targeting age-associated changes to frailty and quality of life.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kenny Vo
- Cellular Longevity Inc, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Tan W, Wang Y, Cheng S, Liu Z, Xie M, Song L, Qiu Q, Wang X, Li Z, Liu T, Guo F, Wang J, Zhou X. AdipoRon ameliorates the progression of heart failure with preserved ejection fraction via mitigating lipid accumulation and fibrosis. J Adv Res 2025; 68:299-315. [PMID: 38382593 PMCID: PMC11785573 DOI: 10.1016/j.jare.2024.02.015] [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: 12/12/2023] [Revised: 02/17/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION Obesity and imbalance in lipid homeostasis contribute greatly to heart failure with preserved ejection fraction (HFpEF), the dominant form of heart failure. Few effective therapies exist to control metabolic alterations and lipid homeostasis. OBJECTIVES We aimed to investigate the cardioprotective roles of AdipoRon, the adiponectin receptor agonist, in regulating lipid accumulation in the two-hit HFpEF model. METHODS HFpEF mouse model was induced using 60 % high-fat diet plus L-NAME drinking water. Then, AdipoRon (50 mg/kg) or vehicle were administered by gavage to the two-hit HFpEF mouse model once daily for 4 weeks. Cardiac function was evaluated using echocardiography, and Postmortem analysis included RNA-sequencing, untargeted metabolomics, transmission electron microscopy and molecular biology methods. RESULTS Our study presents the pioneering evidence that AdipoR was downregulated and impaired fatty acid oxidation in the myocardia of HFpEF mice, which was associated with lipid metabolism as indicated by untargeted metabolomics. AdipoRon, orally active synthetic adiponectin receptor agonist, could upregulate AdipoR1/2 (independently of adiponectin) and reduce lipid droplet accumulation, and alleviate fibrosis to restore HFpEF phenotypes. Finally, AdipoRon primarily exerted its effects through restoring the balance of myocardial fatty acid intake, transport, and oxidation via the downstream AMPKα or PPARα signaling pathways. The protective effects of AdipoRon in HFpEF mice were reversed by compound C and GW6471, inhibitors of AMPKα and PPARα, respectively. CONCLUSIONS AdipoRon ameliorated the HFpEF phenotype by promoting myocardial fatty acid oxidation, decreasing fatty acid transport, and inhibiting fibrosis via the upregulation of AdipoR and the activation of AdipoR1/AMPKα and AdipoR2/PPARα-related downstream pathways. These findings underscore the therapeutic potential of AdipoRon in HFpEF. Importantly, all these parameters get restored in the context of continued mechanical and metabolic stressors associated with HFpEF.
Collapse
Affiliation(s)
- Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Yijun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Siyi Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Mengjie Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Lingpeng Song
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Qinfang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Xiaofei Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Zeyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Tianyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Fuding Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China.
| | - Jun Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China.
| |
Collapse
|
3
|
Nakamura S, Asaba S, Tanaka M, Matsui T. Oral Administration of the Adiponectin Receptor 1 Agonistic Dipeptide Tyr-Pro Prevents Hyperglycemia in Spontaneously Diabetic Torii Rats. ACS OMEGA 2025; 10:1411-1418. [PMID: 39829448 PMCID: PMC11740145 DOI: 10.1021/acsomega.4c09030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025]
Abstract
The dipeptide Tyr-Pro, a novel natural agonist of adiponectin receptor 1 (AdipoR1), promotes glucose uptake in skeletal muscle cells. This study investigated the antidiabetic effect of orally administered Tyr-Pro in spontaneously diabetic Torii (SDT) rats. Oral administration of Tyr-Pro (1 mg/kg/day) improved glucose intolerance in SDT rats at 22 weeks of prediabetic age. By 29 weeks of age, fasting blood glucose levels (BGLs) increased to 142 ± 14 mg/dL in the control group, whereas those in the Tyr-Pro group remained within the normal range (80-99 mg/dL), demonstrating a novel antidiabetic effect in vivo. Substantially increased levels of AdipoR1 and p-AMPK/AMPK were observed in the skeletal muscle of Tyr-Pro-administrated SDT rats. The intake of Tyr-Pro also enhanced insulin secretion and inhibited p-IRS-1(Ser) in skeletal muscle. These findings demonstrate that Tyr-Pro prevented the onset of diabetes and improved impaired insulin signaling pathways in SDT rats by inducing AdipoR1-mediated AMPK activation.
Collapse
Affiliation(s)
- Saya Nakamura
- Department of Bioscience
and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Sumire Asaba
- Department of Bioscience
and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Mitsuru Tanaka
- Department of Bioscience
and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Toshiro Matsui
- Department of Bioscience
and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
4
|
Qian Y, Qi Y, Lin J, Zhang T, Mo L, Xue Q, Zheng N, Niu Y, Dong X, Shi Y, Jiang Y. AdipoRon ameliorates chronic ethanol induced cardiac necroptosis by reducing ceramide mediated mtROS. Free Radic Biol Med 2025; 229:237-250. [PMID: 39805512 DOI: 10.1016/j.freeradbiomed.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/30/2024] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
Chronic ethanol (EtOH) consumption has been widely recognized as a significant contributor to cardiotoxicity. However, no specific treatment is currently available to ameliorate chronic ethanol induced cardiotoxicity. Adiponectin receptor agonist AdipoRon exerts protective effects in multiple organs through alleviating lipotoxicity. Our previous study showed that chronic ethanol consumption increased de novo ceramide synthesis and necroptosis in myocardium. In this study, we investigated the role of AdipoRon on ceramide metabolism and necroptosis in chronic ethanol-treated myocardium. Eight-week-old C57/BL6J mice were fed with a Lieber-Decarli diet containing vehicle or AdipoRon for 12 weeks. Cardiac function, histology and oxidative stress were assessed. We found that chronic ethanol treatment decreased expression of AdipoR2 in myocardium and H9c2 cells, whereas AdipoRon improved cardiac function, reduced myocardium ceramide levels and suppressed necroptosis. By pharmacological interventions, RNA interference and point mutations in AdipoR2, we demonstrated that AdipoRon reduced ceramide levels through PPARα mediated lipid metabolism rather than AdipoR2's ceramidase activity. Using transmission electron microscope and reactive oxygen species (ROS) staining, we showed that chronic ethanol induced myocardium mitochondria damage and mitochondrial reactive oxygen species (mtROS) accumulation. Meanwhile, we found that AdipoRon ameliorated chronic ethanol induced cardiac necroptosis via the SIRT3-SOD2-mtROS pathway. Moreover, C6 ceramide treatment recapitulated chronic ethanol in inducing mtROS and necroptosis, whereas the ceramide synthesis inhibitors myriocin (MYR) and fumonisin B1 (FB1) attenuated chronic ethanol induced mtROS and necroptosis. Collectively, AdipoRon ameliorates chronic ethanol induced cardiac necroptosis by reducing ceramide de novo synthesis and mtROS, which highlights the therapeutic potential of targeting ceramide metabolism and oxidative stress pathways in treating ethanol induced cardiotoxicity.
Collapse
Affiliation(s)
- Yile Qian
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yanyu Qi
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Junyi Lin
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Tianyi Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lingjie Mo
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qiupeng Xue
- Forensic Science and Information Technology Research Centre of Supreme People's Procuratorate, Beijing, 100726, China
| | - Nianchang Zheng
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yaqin Niu
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiaoru Dong
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yan Shi
- Academy of Forensic Science Shanghai Key Laboratory of Forensic Medicine, Shanghai, 200063, China.
| | - Yan Jiang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Gianopoulos I, Mantzoros CS, Daskalopoulou SS. Adiponectin and Adiponectin Receptors in Atherosclerosis. Endocr Rev 2025; 46:1-25. [PMID: 39106421 PMCID: PMC11720176 DOI: 10.1210/endrev/bnae021] [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: 12/20/2023] [Revised: 05/14/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
Adiponectin is an abundantly secreted hormone that communicates information between the adipose tissue, and the immune and cardiovascular systems. In metabolically healthy individuals, adiponectin is usually found at high levels and helps improve insulin responsiveness of peripheral tissues, glucose tolerance, and fatty acid oxidation. Beyond its metabolic functions in insulin-sensitive tissues, adiponectin plays a prominent role in attenuating the development of atherosclerotic plaques, partially through regulating macrophage-mediated responses. In this context, adiponectin binds to its receptors, adiponectin receptor 1 (AdipoR1) and AdipoR2 on the cell surface of macrophages to activate a downstream signaling cascade and induce specific atheroprotective functions. Notably, macrophages modulate the stability of the plaque through their ability to switch between proinflammatory responders, and anti-inflammatory proresolving mediators. Traditionally, the extremes of the macrophage polarization spectrum span from M1 proinflammatory and M2 anti-inflammatory phenotypes. Previous evidence has demonstrated that the adiponectin-AdipoR pathway influences M1-M2 macrophage polarization; adiponectin promotes a shift toward an M2-like state, whereas AdipoR1- and AdipoR2-specific contributions are more nuanced. To explore these concepts in depth, we discuss in this review the effect of adiponectin and AdipoR1/R2 on 1) metabolic and immune responses, and 2) M1-M2 macrophage polarization, including their ability to attenuate atherosclerotic plaque inflammation, and their potential as therapeutic targets for clinical applications.
Collapse
Affiliation(s)
- Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA 02130, USA
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
6
|
Laurindo LF, Laurindo LF, Rodrigues VD, Catharin VMCS, Simili OAG, Barboza GO, Catharin VCS, Sloan KP, Barbalho SM. Unraveling the rationale and conducting a comprehensive assessment of AdipoRon (adiponectin receptor agonist) as a candidate drug for diabetic nephropathy and cardiomyopathy prevention and intervention-a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:165-177. [PMID: 39136737 DOI: 10.1007/s00210-024-03362-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 02/02/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a widespread chronic disease characterized by persistent hyperglycemia, leading to severe complications such as diabetic cardiomyopathy and nephropathy, significantly affecting patient health and quality of life. The complex mechanisms underlying these complications include chronic inflammation, oxidative stress, and metabolic dysregulation. Diabetic cardiomyopathy, marked by structural and functional heart abnormalities, and diabetic nephropathy, characterized by progressive kidney damage, are major contributors to the increased morbidity and mortality associated with T2DM. AdipoRon, a synthetic adiponectin receptor agonist, has shown potential in preclinical studies for mimicking the beneficial effects of endogenous adiponectin, reducing inflammation and oxidative stress, and improving lipid metabolism and mitochondrial function. This systematic review evaluates the therapeutic potential of AdipoRon, focusing on its impact on diabetic cardiomyopathy and nephropathy. Through a comprehensive literature search and analysis, we highlight AdipoRon's role in ameliorating cardiovascular and renal complications in various animal models and cellular systems. The findings underscore the urgent need for translational clinical studies to validate AdipoRon's efficacy and safety in human populations, aiming to advance this promising therapeutic approach from experimental models to clinical application, potentially offering new hope for improved management of diabetic complications.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil.
- Department of Administration, Associate Degree in Hospital Management, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil.
| | - Lívia Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de São José Do Rio Preto (FAMERP), São José Do Rio Preto 15090-000, São Paulo, Brazil
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil
| | - Virgínia Maria Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- School of Medicine, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Otávio Augusto Garcia Simili
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Gabriella Oliveira Barboza
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Vitor Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- School of Medicine, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | | | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- School of Medicine, Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
- UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| |
Collapse
|
7
|
Sun S, Guo H, Chen G, Zhang H, Zhang Z, Wang X, Li D, Li X, Zhao G, Lin F. Peroxisome proliferator‑activated receptor γ coactivator‑1α in heart disease (Review). Mol Med Rep 2025; 31:17. [PMID: 39513608 PMCID: PMC11551696 DOI: 10.3892/mmr.2024.13382] [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: 07/29/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Heart disease (HD) is a general term for various diseases affecting the heart. An increasing body of evidence suggests that the pathogenesis of HD is closely related to mitochondrial dysfunction. Peroxisome proliferator‑activated receptor γ coactivator‑1α (PGC‑1α) is a transcriptional coactivator that plays an important role in mitochondrial function by regulating mitochondrial biogenesis, energy metabolism and oxidative stress. The present review shows that PGC‑1α expression and activity in the heart are controlled by multiple signaling pathways, including adenosine monophosphate‑activated protein kinase, sirtuin 1/3 and nuclear factor κB. These can mediate the activation or inhibition of transcription and post‑translational modifications (such as phosphorylation and acetylation) of PGC‑1α. Furthermore, it highlighted the recent progress of PGC‑1α in HD, including heart failure, coronary heart disease, diabetic cardiomyopathy, drug‑induced cardiotoxicity and arrhythmia. Understanding the mechanisms underlying PGC‑1α in response to pathological stimulation may prove to be beneficial in developing new ideas and strategies for preventing and treating HDs. Meanwhile, the present review explored why the opposite results occurred when PGC‑1α was used as a target therapy.
Collapse
Affiliation(s)
- Siyu Sun
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Huige Guo
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang, Henan 453000, P.R. China
| | - Guohui Chen
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Hui Zhang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Zhanrui Zhang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Xiulong Wang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Dongxu Li
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Xuefang Li
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Guoan Zhao
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Fei Lin
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| |
Collapse
|
8
|
Abou-Samra M, Dubuisson N, Marino A, Selvais CM, Romain V, Davis-López de Carrizosa MA, Noel L, Beauloye C, Brichard SM, Horman S. Striking Cardioprotective Effects of an Adiponectin Receptor Agonist in an Aged Mouse Model of Duchenne Muscular Dystrophy. Antioxidants (Basel) 2024; 13:1551. [PMID: 39765879 PMCID: PMC11674016 DOI: 10.3390/antiox13121551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Adiponectin (ApN) is a hormone with potent effects on various tissues. We previously demonstrated its ability to counteract Duchenne muscular dystrophy (DMD), a severe muscle disorder. However, its therapeutic use is limited. AdipoRon, an orally active ApN mimic, offers a promising alternative. While cardiomyopathy is the primary cause of mortality in DMD, the effects of ApN or AdipoRon on dystrophic hearts have not been investigated. Our recent findings demonstrated the significant protective effects of AdipoRon on dystrophic skeletal muscle. In this study, we investigated whether AdipoRon effects could be extended to dystrophic hearts. As cardiomyopathy develops late in mdx mice (DMD mouse model), 14-month-old mdx mice were orally treated for two months with AdipoRon at a dose of 50 mg/kg/day and then compared with untreated mdx and wild-type (WT) controls. Echocardiography revealed cardiac dysfunction and ventricular hypertrophy in mdx mice, which were fully reversed in AdipoRon-treated mice. AdipoRon also reduced markers of cardiac inflammation, oxidative stress, hypertrophy, and fibrosis while enhancing mitochondrial biogenesis via ApN receptor-1 and CAMKK2/AMPK pathways. Remarkably, treated mice also showed improved skeletal muscle strength and endurance. By offering protection to both cardiac and skeletal muscles, AdipoRon holds potential as a comprehensive therapeutic strategy for better managing DMD.
Collapse
Affiliation(s)
- Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (C.M.S.); (V.R.); (M.A.D.-L.d.C.); (L.N.); (S.M.B.)
| | - Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (C.M.S.); (V.R.); (M.A.D.-L.d.C.); (L.N.); (S.M.B.)
- Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Alice Marino
- Pole of Cardiovascular Research, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (A.M.); (C.B.); (S.H.)
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (C.M.S.); (V.R.); (M.A.D.-L.d.C.); (L.N.); (S.M.B.)
| | - Versele Romain
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (C.M.S.); (V.R.); (M.A.D.-L.d.C.); (L.N.); (S.M.B.)
| | - Maria A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (C.M.S.); (V.R.); (M.A.D.-L.d.C.); (L.N.); (S.M.B.)
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (C.M.S.); (V.R.); (M.A.D.-L.d.C.); (L.N.); (S.M.B.)
| | - Christophe Beauloye
- Pole of Cardiovascular Research, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (A.M.); (C.B.); (S.H.)
- Department of Cardiovascular Intensive Care, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Sonia M. Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (C.M.S.); (V.R.); (M.A.D.-L.d.C.); (L.N.); (S.M.B.)
| | - Sandrine Horman
- Pole of Cardiovascular Research, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (A.M.); (C.B.); (S.H.)
| |
Collapse
|
9
|
Sun J, Du B, Chen M, Jia J, Wang X, Hong J. FBXO28 reduces high-fat diet-induced hyperlipidemia in mice by alleviating abnormal lipid metabolism and inflammatory responses. J Endocrinol Invest 2024; 47:2757-2774. [PMID: 38696123 DOI: 10.1007/s40618-024-02376-5] [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: 11/24/2023] [Accepted: 04/12/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Hyperlipidemia is a lipid metabolism disorder with increasing incidence and prevalence worldwide. Abnormal lipid metabolism and inflammation are two significant characteristics of hyperlipidemia. The purpose of this study was to explore the role and mechanism of F-box only protein 28 (FBXO28) in hyperlipidemia. METHODS Mice were fed with high-fat diet (HFD) to elicit obesity, and 3T3-L1 preadipocytes were stimulated with MDI cocktail (IBMX, DEX and insulin) to evoke differentiation. In vivo and in vitro role of FBXO28 in hyperlipidemia was investigated by hematoxylin-eosin and oil Red O staining, the lipid biochemistry measurement, enzyme-linked immunosorbent assay, reverse transcription quantitative polymerase chain reaction and western blotting assays. The mechanism of FBXO28 explored by co-immunoprecipitation, immunofluorescence, ubiquitination and cycloheximide assays. RESULTS Low expression of FBXO28 was found in hyperlipidemia in silico, in vivo and in vitro. Upregulation of FBXO28 declined the body weight, fat accumulation, and serum lipid content in HFD-fed mice. Abnormal lipid accumulation, and the level of liposynthetic genes and beta-oxidation related genes were improved by overexpression of FBXO28 both in HFD-elicited mice and MDI-treated 3T3-L1 preadipocytes. Besides, overexpression of FBXO28 declined HFD-induced the level of proinflammatory factors and F4/80. Mechanically, FBXO28 directly bound RAB27A and promoted its ubiquitinated degradation. Thus, upregulation of RAB27A inverted the improved role of FBXO28 in abnormal lipid metabolism and inflammation in vivo and in vitro. CONCLUSION FBXO28 ameliorated abnormal lipid metabolism and inflammation through the ubiquitinated degradation of RAB27A, thereby attenuating HFD-induced hyperlipidemia. The results could promote the treatment of hyperlipidemia, and the relevant diseases.
Collapse
Affiliation(s)
- J Sun
- Cadre's Ward, The Fourth Clinical College of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - B Du
- Department of Neurology, Urumqi Midong District Hospital of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - M Chen
- Cadre's Ward, Traditional Chinese Medicine Hospital of Xinjiang Medical University, No.116, Huanghe Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - J Jia
- Cadre's Ward, Traditional Chinese Medicine Hospital of Xinjiang Medical University, No.116, Huanghe Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - X Wang
- Cadre's Ward, Traditional Chinese Medicine Hospital of Xinjiang Medical University, No.116, Huanghe Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - J Hong
- Cadre's Ward, Traditional Chinese Medicine Hospital of Xinjiang Medical University, No.116, Huanghe Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
10
|
Xu JW, Chen FF, Qv YH, Sun CC, Zhang D, Guo Z, Wang YJ, Wang JF, Liu T, Dong L, Qi Q. Unleashing AdipoRon's Potential: A Fresh Approach to Tackle Pseudomonas aeruginosa Infections in Bronchiectasis via Sphingosine Metabolism Modulation. J Inflamm Res 2024; 17:7653-7674. [PMID: 39469062 PMCID: PMC11514707 DOI: 10.2147/jir.s483689] [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: 08/02/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Purpose Bronchiectasis patients are prone to Pseudomonas aeruginosa infection due to decreased level of sphingosine in airway. Adiponectin receptor agonist AdipoRon activates the intrinsic ceramidase activity of adiponectin receptor 1 (AdipoR1) and positively regulates sphingosine metabolism. This study aimed to investigate the potential therapeutic benefit of AdipoRon against Pseudomonas aeruginosa infection. Methods A mouse model of Pseudomonas aeruginosa lung infection and a co-culture model of human bronchial epithelial cells with Pseudomonas aeruginosa were established to explore the protective effect of AdipoRon. Liquid chromatography-mass spectrometry was used to detect the effect of AdipoRon on sphingosine level in lung of Pseudomonas aeruginosa-infected mouse models. Results The down-regulation of adiponectin and AdipoR1 in airway of bronchiectasis patients was linked to Pseudomonas aeruginosa infection. By activating AdipoR1, AdipoRon reduced Pseudomonas aeruginosa adherence on bronchial epithelial cells and protected cilia from damage in vitro. With the treatment of AdipoRon, the load of Pseudomonas aeruginosa in lung significantly decreased, and peribronchial inflammatory cell infiltration was lessened in vivo. The reduced level of sphingosine in the airway of Pseudomonas aeruginosa infected mice was replenished by AdipoRon, thus playing a protective role in the airway. Moreover, AdipoRon activated P-AMPKα/PGC1α, inhibited TLR4/P-NF-κB p65, and reduced expression of pro-apoptotic bax. However, the protective effect of AdipoRon on resisting Pseudomonas aeruginosa infection was weakened when AdipoR1 was knocked down. Conclusion AdipoRon protects bronchial epithelial cells and lung by enhancing their resistance to Pseudomonas aeruginosa infection. The mechanism might be modulating sphingosine metabolism and activating P-AMPKα/PGC1α while inhibiting TLR4/P-NF-κB p65.
Collapse
Affiliation(s)
- Jia-wei Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Characteristic Laboratory of Clinical Transformation of Respiratory Biological Immunity and Regenerative Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Fang-fang Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Characteristic Laboratory of Clinical Transformation of Respiratory Biological Immunity and Regenerative Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Ying-hui Qv
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Characteristic Laboratory of Clinical Transformation of Respiratory Biological Immunity and Regenerative Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Cong-cong Sun
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Characteristic Laboratory of Clinical Transformation of Respiratory Biological Immunity and Regenerative Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Dong Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Characteristic Laboratory of Clinical Transformation of Respiratory Biological Immunity and Regenerative Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Zhi Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Characteristic Laboratory of Clinical Transformation of Respiratory Biological Immunity and Regenerative Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Yu-jiao Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Jun-fei Wang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, People’s Republic of China
| | - Tian Liu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, People’s Republic of China
| | - Liang Dong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Characteristic Laboratory of Clinical Transformation of Respiratory Biological Immunity and Regenerative Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Qian Qi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Characteristic Laboratory of Clinical Transformation of Respiratory Biological Immunity and Regenerative Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, 250014, People’s Republic of China
| |
Collapse
|
11
|
Hemat Jouy S, Mohan S, Scichilone G, Mostafa A, Mahmoud AM. Adipokines in the Crosstalk between Adipose Tissues and Other Organs: Implications in Cardiometabolic Diseases. Biomedicines 2024; 12:2129. [PMID: 39335642 PMCID: PMC11428859 DOI: 10.3390/biomedicines12092129] [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: 08/25/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Adipose tissue was previously regarded as a dormant organ for lipid storage until the identification of adiponectin and leptin in the early 1990s. This revelation unveiled the dynamic endocrine function of adipose tissue, which has expanded further. Adipose tissue has emerged in recent decades as a multifunctional organ that plays a significant role in energy metabolism and homeostasis. Currently, it is evident that adipose tissue primarily performs its function by secreting a diverse array of signaling molecules known as adipokines. Apart from their pivotal function in energy expenditure and metabolism regulation, these adipokines exert significant influence over a multitude of biological processes, including but not limited to inflammation, thermoregulation, immune response, vascular function, and insulin sensitivity. Adipokines are pivotal in regulating numerous biological processes within adipose tissue and facilitating communication between adipose tissue and various organs, including the brain, gut, pancreas, endothelial cells, liver, muscle, and more. Dysregulated adipokines have been implicated in several metabolic diseases, like obesity and diabetes, as well as cardiovascular diseases. In this article, we attempted to describe the significance of adipokines in developing metabolic and cardiovascular diseases and highlight their role in the crosstalk between adipose tissues and other tissues and organs.
Collapse
Affiliation(s)
- Shaghayegh Hemat Jouy
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Central Tehran Branch, Islamic Azad University, Tehran 14778-93855, Iran;
| | - Sukrutha Mohan
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
| | - Giorgia Scichilone
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
| | - Amro Mostafa
- Department of Pharmacology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Abeer M. Mahmoud
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
12
|
Liu YF, Tian Y, Chen XF, Zhang C, Huang L. Role of osteokines in atherosclerosis. Cell Biochem Funct 2024; 42:e4107. [PMID: 39154288 DOI: 10.1002/cbf.4107] [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: 03/21/2024] [Revised: 06/03/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024]
Abstract
Despite their diverse physiologies and roles, the heart, skeletal muscles, and smooth muscles all derive from a common embryonic source as bones. Moreover, bone tissue, skeletal and smooth muscles, and the heart share conserved signaling pathways. The maintenance of skeletal health is precisely regulated by osteocytes, osteoblasts, and osteoclasts through coordinated secretion of bone-derived factors known as osteokines. Increasing evidence suggests the involvement of osteokines in regulating atherosclerotic vascular disease. Therefore, this review aims to examine the evidence for the role of osteokines in atherosclerosis development and progression comprehensively. Specifically discussed are extensively studied osteokines in atherosclerosis such as osteocalcin, osteopontin, osteoprotegerin, and fibroblast growth factor 23. Additionally, we highlighted the effects of exercise on modulating these key regulators derived from bone tissue metabolism. We believe that gaining an enhanced understanding of how osteocalcin contributes to the process of atherosclerosis will enable us to develop targeted and comprehensive therapeutic strategies against diseases associated with its progression.
Collapse
Affiliation(s)
- Yi-Fan Liu
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, People's Republic of China
| | - Xiao-Fang Chen
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, People's Republic of China
| | - Liang Huang
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
13
|
Elkanawati RY, Sumiwi SA, Levita J. Impact of Lipids on Insulin Resistance: Insights from Human and Animal Studies. Drug Des Devel Ther 2024; 18:3337-3360. [PMID: 39100221 PMCID: PMC11298177 DOI: 10.2147/dddt.s468147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024] Open
Abstract
Insulin resistance (IR) is a complex pathological condition central to metabolic diseases such as type 2 diabetes mellitus (T2DM), cardiovascular disease, non-alcoholic fatty liver disease, and polycystic ovary syndrome (PCOS). This review evaluates the impact of lipids on insulin resistance (IR) by analyzing findings from human and animal studies. The articles were searched on the PubMed database using two keywords: (1) "Role of Lipids AND Insulin Resistance AND Humans" and (2) "Role of Lipids AND Insulin Resistance AND Animal Models". Studies in humans revealed that elevated levels of free fatty acids (FFAs) and triglycerides (TGs) are closely associated with reduced insulin sensitivity, and interventions like metformin and omega-3 fatty acids show potential benefits. In animal models, high-fat diets disrupt insulin signaling and increase inflammation, with lipid mediators such as diacylglycerol (DAG) and ceramides playing significant roles. DAG activates protein kinase C, which eventually impairs insulin signaling, while ceramides inhibit Akt/PKB, further contributing to IR. Understanding these mechanisms is crucial for developing effective prevention and treatment strategies for IR-related diseases.
Collapse
Affiliation(s)
- Rani Yulifah Elkanawati
- Master Program in Pharmacy, Faculty of Pharmacy, Padjadjaran University, Jawa Barat, West Java, 45363, Indonesia
| | - Sri Adi Sumiwi
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, West Java, 45363, Indonesia
| | - Jutti Levita
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, West Java, 45363, Indonesia
| |
Collapse
|
14
|
Pan X, Hao E, Zhang F, Wei W, Du Z, Yan G, Wang X, Deng J, Hou X. Diabetes cardiomyopathy: targeted regulation of mitochondrial dysfunction and therapeutic potential of plant secondary metabolites. Front Pharmacol 2024; 15:1401961. [PMID: 39045049 PMCID: PMC11263127 DOI: 10.3389/fphar.2024.1401961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a specific heart condition in diabetic patients, which is a major cause of heart failure and significantly affects quality of life. DCM is manifested as abnormal cardiac structure and function in the absence of ischaemic or hypertensive heart disease in individuals with diabetes. Although the development of DCM involves multiple pathological mechanisms, mitochondrial dysfunction is considered to play a crucial role. The regulatory mechanisms of mitochondrial dysfunction mainly include mitochondrial dynamics, oxidative stress, calcium handling, uncoupling, biogenesis, mitophagy, and insulin signaling. Targeting mitochondrial function in the treatment of DCM has attracted increasing attention. Studies have shown that plant secondary metabolites contribute to improving mitochondrial function and alleviating the development of DCM. This review outlines the role of mitochondrial dysfunction in the pathogenesis of DCM and discusses the regulatory mechanism for mitochondrial dysfunction. In addition, it also summarizes treatment strategies based on plant secondary metabolites. These strategies targeting the treatment of mitochondrial dysfunction may help prevent and treat DCM.
Collapse
Affiliation(s)
- Xianglong Pan
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Fan Zhang
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wei Wei
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zhengcai Du
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Guangli Yan
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xijun Wang
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xiaotao Hou
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
15
|
Norris MK, Tippetts TS, Wilkerson JL, Nicholson RJ, Maschek JA, Levade T, Medin JA, Summers SA, Holland WL. Adiponectin overexpression improves metabolic abnormalities caused by acid ceramidase deficiency but does not prolong lifespan in a mouse model of Farber Disease. Mol Genet Metab Rep 2024; 39:101077. [PMID: 38595987 PMCID: PMC11002753 DOI: 10.1016/j.ymgmr.2024.101077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/23/2024] [Indexed: 04/11/2024] Open
Abstract
Farber Disease is a debilitating and lethal childhood disease of ceramide accumulation caused by acid ceramidase deficiency. The potent induction of a ligand-gated neutral ceramidase activity promoted by adiponectin may provide sufficient lowering of ceramides to allow for the treatment of Farber Disease. In vitro, adiponectin or adiponectin receptor agonist treatments lowered total ceramide concentrations in human fibroblasts from a patient with Farber Disease. However, adiponectin overexpression in a Farber Disease mouse model did not improve lifespan or immune infiltration. Intriguingly, mice heterozygous for the Farber Disease mutation were more prone to glucose intolerance and insulin resistance when fed a high-fat diet, and adiponectin overexpression protected from these metabolic perturbations. These studies suggest that adiponectin evokes a ceramidase activity that is not reliant on the functional expression of acid ceramidase, but indicates that additional strategies are required to ameliorate outcomes of Farber Disease.
Collapse
Affiliation(s)
- Marie K. Norris
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| | - Trevor S. Tippetts
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph L. Wilkerson
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| | - Rebekah J. Nicholson
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| | - J. Alan Maschek
- Metabolomics Core Facility, University of Utah, Salt Lake City, UT, USA
| | - Thierry Levade
- Laboratoire de Biochimie Métabolique, CHU Toulouse and INSERM U1037, Centre de Recherches en Cancérologie de Toulouse, Université Paul Sabatier, 31037 Toulouse, France
| | - Jeffrey A. Medin
- Departments of Pediatrics and Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott A. Summers
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| | - William L. Holland
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
- Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
16
|
Zhang J, Zhou H, Lei F, Jiang K, Liao Y, Huang F, Chen M. Cholesterol 25-hydroxylase prevents type 2 diabetes mellitus induced cardiomyopathy by alleviating cardiac lipotoxicity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167158. [PMID: 38588780 DOI: 10.1016/j.bbadis.2024.167158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/28/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
OBJECTIVES Diabetic cardiomyopathy (DCM) is the leading cause of mortality in type 2 diabetes mellitus (T2DM) patients, with its underlying mechanisms still elusive. This study aims to investigate the role of cholesterol-25-monooxygenase (CH25H) in T2DM induced cardiomyopathy. METHODS High fat diet combined with streptozotocin (HFD/STZ) were used to establish a T2DM model. CH25H and its product 25-hydroxycholesterol (25HC) were detected in the hearts of T2DM model. Gain- or loss-of-function of CH25H were performed by receiving AAV9-cTNT-CH25H or CH25H knockout (CH25H-/-) mice with HFD/STZ treatment. Cardiac function was evaluated using echocardiography, and cardiac tissues were collected for immunoblot analysis, histological assessment and quantitative polymerase chain reaction (qPCR). Mitochondrial morphology and function were evaluated using transmission electron microscopy (TEM) and Seahorse XF Cell Mito Stress Test Kit. RNA-sequence analysis was performed to determine the molecular changes associated with CH25H deletion. RESULTS CH25H and 25HC were significantly decreased in the hearts of T2DM mice. CH25H-/- mice treated with HFD/STZ exhibited impaired mitochondrial function and structure, increased lipid accumulation, and aggregated cardiac dysfunction. Conversely, T2DM mice receiving AAV9-CH25H displayed cardioprotective effects. Mechanistically, RNA sequencing and qPCR analysis revealed that CH25H deficiency decreased peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) and its target gene expression. Additionally, administration of ZLN005, a potent PGC-1α activator, partially protected against high glucose and palmitic acid induced mitochondria dysfunction and lipid accumulation in vitro. CONCLUSION Our study provides compelling evidence supporting the protective role of CH25H in T2DM-induced cardiomyopathy. Furthermore, the regulation of PGC-1α may be intricately involved in this cardioprotective process.
Collapse
MESH Headings
- Animals
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/prevention & control
- Diabetic Cardiomyopathies/etiology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Mice
- Mice, Knockout
- Male
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Steroid Hydroxylases/metabolism
- Steroid Hydroxylases/genetics
- Diet, High-Fat/adverse effects
- Mice, Inbred C57BL
- Hydroxycholesterols/metabolism
- Myocardium/metabolism
- Myocardium/pathology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
Collapse
Affiliation(s)
- Jialiang Zhang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Zhou
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Lei
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kexin Jiang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanbiao Liao
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fangyang Huang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Mao Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
17
|
Han Y, Sun Q, Chen W, Gao Y, Ye J, Chen Y, Wang T, Gao L, Liu Y, Yang Y. New advances of adiponectin in regulating obesity and related metabolic syndromes. J Pharm Anal 2024; 14:100913. [PMID: 38799237 PMCID: PMC11127227 DOI: 10.1016/j.jpha.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/18/2023] [Accepted: 12/07/2023] [Indexed: 05/29/2024] Open
Abstract
Obesity and related metabolic syndromes have been recognized as important disease risks, in which the role of adipokines cannot be ignored. Adiponectin (ADP) is one of the key adipokines with various beneficial effects, including improving glucose and lipid metabolism, enhancing insulin sensitivity, reducing oxidative stress and inflammation, promoting ceramides degradation, and stimulating adipose tissue vascularity. Based on those, it can serve as a positive regulator in many metabolic syndromes, such as type 2 diabetes (T2D), cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), sarcopenia, neurodegenerative diseases, and certain cancers. Therefore, a promising therapeutic approach for treating various metabolic diseases may involve elevating ADP levels or activating ADP receptors. The modulation of ADP genes, multimerization, and secretion covers the main processes of ADP generation, providing a comprehensive orientation for the development of more appropriate therapeutic strategies. In order to have a deeper understanding of ADP, this paper will provide an all-encompassing review of ADP.
Collapse
Affiliation(s)
- Yanqi Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Qianwen Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Wei Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yue Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanmin Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Tingting Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lili Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
18
|
Fleishman JS, Kumar S. Bile acid metabolism and signaling in health and disease: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:97. [PMID: 38664391 PMCID: PMC11045871 DOI: 10.1038/s41392-024-01811-6] [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: 11/28/2023] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
Bile acids, once considered mere dietary surfactants, now emerge as critical modulators of macronutrient (lipid, carbohydrate, protein) metabolism and the systemic pro-inflammatory/anti-inflammatory balance. Bile acid metabolism and signaling pathways play a crucial role in protecting against, or if aberrant, inducing cardiometabolic, inflammatory, and neoplastic conditions, strongly influencing health and disease. No curative treatment exists for any bile acid influenced disease, while the most promising and well-developed bile acid therapeutic was recently rejected by the FDA. Here, we provide a bottom-up approach on bile acids, mechanistically explaining their biochemistry, physiology, and pharmacology at canonical and non-canonical receptors. Using this mechanistic model of bile acids, we explain how abnormal bile acid physiology drives disease pathogenesis, emphasizing how ceramide synthesis may serve as a unifying pathogenic feature for cardiometabolic diseases. We provide an in-depth summary on pre-existing bile acid receptor modulators, explain their shortcomings, and propose solutions for how they may be remedied. Lastly, we rationalize novel targets for further translational drug discovery and provide future perspectives. Rather than dismissing bile acid therapeutics due to recent setbacks, we believe that there is immense clinical potential and a high likelihood for the future success of bile acid therapeutics.
Collapse
Affiliation(s)
- Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| |
Collapse
|
19
|
Ye W, Han K, Xie M, Li S, Chen G, Wang Y, Li T. Mitochondrial energy metabolism in diabetic cardiomyopathy: Physiological adaption, pathogenesis, and therapeutic targets. Chin Med J (Engl) 2024; 137:936-948. [PMID: 38527931 PMCID: PMC11046025 DOI: 10.1097/cm9.0000000000003075] [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: 07/08/2023] [Indexed: 03/27/2024] Open
Abstract
ABSTRACT Diabetic cardiomyopathy is defined as abnormal structure and function of the heart in the setting of diabetes, which could eventually develop heart failure and leads to the death of the patients. Although blood glucose control and medications to heart failure show beneficial effects on this disease, there is currently no specific treatment for diabetic cardiomyopathy. Over the past few decades, the pathophysiology of diabetic cardiomyopathy has been extensively studied, and an increasing number of studies pinpoint that impaired mitochondrial energy metabolism is a key mediator as well as a therapeutic target. In this review, we summarize the latest research in the field of diabetic cardiomyopathy, focusing on mitochondrial damage and adaptation, altered energy substrates, and potential therapeutic targets. A better understanding of the mitochondrial energy metabolism in diabetic cardiomyopathy may help to gain more mechanistic insights and generate more precise mitochondria-oriented therapies to treat this disease.
Collapse
Affiliation(s)
- Wanlin Ye
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kun Han
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
| | - Maodi Xie
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Sheyu Li
- Department of Endocrinology and Metabolism, Division of Guideline and Rapid Recommendation, Cochrane China Center, MAGIC China Center, Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Guo Chen
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanyan Wang
- Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
20
|
Cho S, Dadson K, Sung HK, Ayansola O, Mirzaesmaeili A, Noskovicova N, Zhao Y, Cheung K, Radisic M, Hinz B, Sater AAA, Hsu HH, Lopaschuk GD, Sweeney G. Cardioprotection by the adiponectin receptor agonist ALY688 in a preclinical mouse model of heart failure with reduced ejection fraction (HFrEF). Biomed Pharmacother 2024; 171:116119. [PMID: 38181714 DOI: 10.1016/j.biopha.2023.116119] [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: 10/29/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
AIMS Adiponectin has been shown to mediate cardioprotective effects and levels are typically reduced in patients with cardiometabolic disease. Hence, there has been intense interest in developing adiponectin-based therapeutics. The aim of this translational research study was to examine the functional significance of targeting adiponectin signaling with the adiponectin receptor agonist ALY688 in a mouse model of heart failure with reduced ejection fraction (HFrEF), and the mechanisms of cardiac remodeling leading to cardioprotection. METHODS AND RESULTS Wild-type mice were subjected to transverse aortic constriction (TAC) to induce left ventricular pressure overload (PO), or sham surgery, with or without daily subcutaneous ALY688-SR administration. Temporal analysis of cardiac function was conducted via weekly echocardiography for 5 weeks and we observed that ALY688 attenuated the PO-induced dysfunction. ALY688 also reduced cardiac hypertrophic remodeling, assessed via LV mass, heart weight to body weight ratio, cardiomyocyte cross sectional area, ANP and BNP levels. ALY688 also attenuated PO-induced changes in myosin light and heavy chain expression. Collagen content and myofibroblast profile indicated that fibrosis was attenuated by ALY688 with TIMP1 and scleraxis/periostin identified as potential mechanistic contributors. ALY688 reduced PO-induced elevation in circulating cytokines including IL-5, IL-13 and IL-17, and the chemoattractants MCP-1, MIP-1β, MIP-1alpha and MIP-3α. Assessment of myocardial transcript levels indicated that ALY688 suppressed PO-induced elevations in IL-6, TLR-4 and IL-1β, collectively indicating anti-inflammatory effects. Targeted metabolomic profiling indicated that ALY688 increased fatty acid mobilization and oxidation, increased betaine and putrescine plus decreased sphingomyelin and lysophospholipids, a profile indicative of improved insulin sensitivity. CONCLUSION These results indicate that the adiponectin mimetic peptide ALY688 reduced PO-induced fibrosis, hypertrophy, inflammation and metabolic dysfunction and represents a promising therapeutic approach for treating HFrEF in a clinical setting.
Collapse
Affiliation(s)
- Sungji Cho
- Department of Biology, York University, Toronto, ON, Canada
| | - Keith Dadson
- Department of Biology, York University, Toronto, ON, Canada
| | | | | | - Ali Mirzaesmaeili
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Nina Noskovicova
- Faculty of Dentistry, University of Toronto, Toronto, ON M5S3E2, Canada
| | - Yimu Zhao
- Toronto General Hospital Research Institute, Toronto, ON M5G 2C4, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Krisco Cheung
- Department of Chemical Engineering and Applied Chemistry; University of Toronto, Toronto, ON M5S 3E5, Canada
| | - Milica Radisic
- Toronto General Hospital Research Institute, Toronto, ON M5G 2C4, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry; University of Toronto, Toronto, ON M5S 3E5, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5S3E2, Canada; Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5B 1T8, Canada
| | - Ali A Abdul Sater
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Henry H Hsu
- Allysta Pharmaceuticals Inc. Bellevue, WA, USA
| | - Gary D Lopaschuk
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada.
| |
Collapse
|
21
|
Hafiane A. Adiponectin-mediated regulation of the adiponectin cascade in cardiovascular disease: Updates. Biochem Biophys Res Commun 2024; 694:149406. [PMID: 38134479 DOI: 10.1016/j.bbrc.2023.149406] [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: 08/08/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
The endocrine function of white adipose tissue is characterized by the synthesis of one its main hormones: adiponectin. Although the biological role of adiponectin has not been fully defined, clinical and experimental observations have shown that low plasma concentrations of adiponectin participate in the prevalence of insulin resistance and cardiovascular diseases, mainly in obese patients. Adiponectin also exerts its effects on the heart and blood vessels, thereby influencing their physiology. Studying the effects of adiponectin presents some complexities, primarily due to potential cross-interactions and interference with other pathways, such as the AdipoR1/R2 pathways. Under optimal conditions, the activation of the adiponectin cascade may involve signals such as AMPK and PPARα. Interestingly, these pathways may trigger similar responses, such as fatty acid oxidation. Understanding the downstream effectors of these pathways is crucial to comprehend the extent to which adiponectin signaling impacts metabolism. In this review, the aim is to explore the current mechanisms that regulate the adiponectin pathways. Additionally, updates on the major downstream factors involved in adiponectin signaling are provided, specifically in relation to metabolic syndrome and atherosclerosis.
Collapse
Affiliation(s)
- Anouar Hafiane
- Research Institute, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
22
|
Zhou J, Li M, Yu Z, Li C, Zhou L, Zhou X. Protective effect of Qingluotongbi formula against Tripterygium wilfordii induced liver injury in mice by improving fatty acid β-oxidation and mitochondrial biosynthesis. PHARMACEUTICAL BIOLOGY 2023; 61:80-88. [PMID: 36541729 PMCID: PMC9788700 DOI: 10.1080/13880209.2022.2157842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/21/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
CONTEXT Qingluotongbi formula (QLT) is a Chinese medicine compound consisting of Tripterygium wilfordii Hook. f. (Celastraceae, TW), Panax notoginseng (Burkill) F.H.Chen (Araliaceae, PN), Rehmannia glutinosa (Gaertn.) DC. (Orobanchaceae, RG), Sinomenium acutum (Thunb.) Rehder & E.H. Wilson (Menispermaceae, SA), and Bombyx mori L. (Bombycidae, BM). OBJECTIVE This study investigated the protective effect and possible mechanism of QLT against TW-induced liver injury in mice. MATERIALS AND METHODS To establish the model of TW-induced liver injury in mice, C57BL/6J mice were randomly divided into 4 groups: control group, low-dose TW group, middle-dose TW group, and high-dose TW group. To observe the effects of QLT and its individual ingredients against TW-induced liver injury, C57BL/6J mice were randomly divided into 7 groups: control group, TW group, QLT group, PN group, RG group, SA group, BM group.After administration for 7 days, C57BL/6J mice were tested for biochemical indicators and liver pathological changes. Then, we evaluated the mitochondrial function and analysed the gene and protein expression related to the peroxisome proliferator-activated receptor alpha (PPARα)/peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) pathway by quantitative real-time PCR (qRT-PCR) and Western blotting. RESULTS Compared with the control group (0.30 ± 0.35), TW significantly increased mice liver histological score (L, 0.95 ± 1.14; M, 1.25 ± 1.16; H, 4.00 ± 1.13). QLT and its ingredients significantly improved the pathology scores (CON, 0.63 ± 0.74; TW, 4.19 ± 1.53; QLT, 1.56 ± 0.62; PN, 1.94 ± 0.68; RG, 2.75 ± 1.39; SA, 4.13 ± 0.99; BM, 4.13 ± 0.99). Western blot and qRT-PCR analysis revealed that QLT and its ingredients reversed TW-induced suppression of PPARα/PGC1-α pathway.Discussion and conclusions: These findings provide valuable information for compound compatibility studies and TW clinical applications.
Collapse
Affiliation(s)
- Jie Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Zhichao Yu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Changqing Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingling Zhou
- Jiangsu Provincial Key Laboratory of Pharmacology and Safety Evaluation of Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xueping Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
23
|
Begum M, Choubey M, Tirumalasetty MB, Arbee S, Mohib MM, Wahiduzzaman M, Mamun MA, Uddin MB, Mohiuddin MS. Adiponectin: A Promising Target for the Treatment of Diabetes and Its Complications. Life (Basel) 2023; 13:2213. [PMID: 38004353 PMCID: PMC10672343 DOI: 10.3390/life13112213] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Diabetes mellitus, a chronic metabolic disorder characterized by hyperglycemia, presents a formidable global health challenge with its associated complications. Adiponectin, an adipocyte-derived hormone, has emerged as a significant player in glucose metabolism and insulin sensitivity. Beyond its metabolic effects, adiponectin exerts anti-inflammatory, anti-oxidative, and vasoprotective properties, making it an appealing therapeutic target for mitigating diabetic complications. The molecular mechanisms by which adiponectin impacts critical pathways implicated in diabetic nephropathy, retinopathy, neuropathy, and cardiovascular problems are thoroughly examined in this study. In addition, we explore possible treatment options for increasing adiponectin levels or improving its downstream signaling. The multifaceted protective roles of adiponectin in diabetic complications suggest its potential as a novel therapeutic avenue. However, further translational studies and clinical trials are warranted to fully harness the therapeutic potential of adiponectin in the management of diabetic complications. This review highlights adiponectin as a promising target for the treatment of diverse diabetic complications and encourages continued research in this pivotal area of diabetes therapeutics.
Collapse
Affiliation(s)
- Mahmuda Begum
- Department of Internal Medicine, HCA-St David’s Medical Center, 919 E 32nd St, Austin, TX 78705, USA;
| | - Mayank Choubey
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA; (M.C.); (M.B.T.); (M.W.)
| | - Munichandra Babu Tirumalasetty
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA; (M.C.); (M.B.T.); (M.W.)
| | - Shahida Arbee
- Institute for Molecular Medicine, Aichi Medical University, 1-Yazako, Karimata, Aichi, Nagakute 480-1103, Japan;
| | - Mohammad Mohabbulla Mohib
- Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Magdeburger Straße 6, 06112 Halle, Germany;
| | - Md Wahiduzzaman
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA; (M.C.); (M.B.T.); (M.W.)
| | - Mohammed A. Mamun
- CHINTA Research Bangladesh, Savar 1342, Bangladesh;
- Department of Public Health and Informatics, Jahangirnagar University, Savar 1342, Bangladesh
| | - Mohammad Borhan Uddin
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh;
| | - Mohammad Sarif Mohiuddin
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA; (M.C.); (M.B.T.); (M.W.)
| |
Collapse
|
24
|
Li AL, Lian L, Chen XN, Cai WH, Fan XB, Fan YJ, Li TT, Xie YY, Zhang JP. The role of mitochondria in myocardial damage caused by energy metabolism disorders: From mechanisms to therapeutics. Free Radic Biol Med 2023; 208:236-251. [PMID: 37567516 DOI: 10.1016/j.freeradbiomed.2023.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
Myocardial damage is the most serious pathological consequence of cardiovascular diseases and an important reason for their high mortality. In recent years, because of the high prevalence of systemic energy metabolism disorders (e.g., obesity, diabetes mellitus, and metabolic syndrome), complications of myocardial damage caused by these disorders have attracted widespread attention. Energy metabolism disorders are independent of traditional injury-related risk factors, such as ischemia, hypoxia, trauma, and infection. An imbalance of myocardial metabolic flexibility and myocardial energy depletion are usually the initial changes of myocardial injury caused by energy metabolism disorders, and abnormal morphology and functional destruction of the mitochondria are their important features. Specifically, mitochondria are the centers of energy metabolism, and recent evidence has shown that decreased mitochondrial function, caused by an imbalance in mitochondrial quality control, may play a key role in myocardial injury caused by energy metabolism disorders. Under chronic energy stress, mitochondria undergo pathological fission, while mitophagy, mitochondrial fusion, and biogenesis are inhibited, and mitochondrial protein balance and transfer are disturbed, resulting in the accumulation of nonfunctional and damaged mitochondria. Consequently, damaged mitochondria lead to myocardial energy depletion and the accumulation of large amounts of reactive oxygen species, further aggravating the imbalance in mitochondrial quality control and forming a vicious cycle. In addition, impaired mitochondria coordinate calcium homeostasis imbalance, and epigenetic alterations participate in the pathogenesis of myocardial damage. These pathological changes induce rapid progression of myocardial damage, eventually leading to heart failure or sudden cardiac death. To intervene more specifically in the myocardial damage caused by metabolic disorders, we need to understand the specific role of mitochondria in this context in detail. Accordingly, promising therapeutic strategies have been proposed. We also summarize the existing therapeutic strategies to provide a reference for clinical treatment and developing new therapies.
Collapse
Affiliation(s)
- Ao-Lin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lu Lian
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xin-Nong Chen
- Department of Traditional Chinese Medicine, Tianjin First Central Hospital, Tianjin, 300190, China
| | - Wen-Hui Cai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xin-Biao Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ya-Jie Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ting-Ting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ying-Yu Xie
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Jun-Ping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China.
| |
Collapse
|
25
|
de Assis GG, Murawska-Ciałowicz E. Exercise and Weight Management: The Role of Leptin-A Systematic Review and Update of Clinical Data from 2000-2022. J Clin Med 2023; 12:4490. [PMID: 37445524 DOI: 10.3390/jcm12134490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
A well-balanced metabolism means a lower risk for metabolism-related neuropsychiatric disorders. Leptin is a secretory adipokine involved in the central control of appetite that appears to play a role in the etiology of feeding-related disorders. Additionally, the influence of exercise on feeding behaviors potentially modulates the circulation of metabolites that signal through the central nervous system. In this systematic review, we collected the recent clinical evidence on the effect of exercise on leptin concentrations in health individuals published from 2000 to 20 September 2022, according to the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols (PRISMA 2020 statement). Six hundred and thirty-eight papers were retrieved and forty-eight papers were included in the qualitative synthesis. Data supports that exercise positively influences appetite via enhancing peripheral and central leptin signaling (reuptake), especially during weight loss. Exercise modulation of leptin signaling through leptin receptors helps to stabilize increases in food intake during periods of negative energy balance, prior to a decrease in the body fat tissue content. At a high intensity, exercise appears to counteract leptin resistance.
Collapse
Affiliation(s)
- Gilmara Gomes de Assis
- Department of Physiology and Pathology, School of Dentistry, UNESP, São Paulo State University, Araraquara 14801-903, Brazil
| | - Eugenia Murawska-Ciałowicz
- Department of Physiology and Biochemistry, Wroclaw University of Health and Sport Sciences, 51-612 Wroclaw, Poland
| |
Collapse
|
26
|
Demirel O, Berezin AE, Mirna M, Boxhammer E, Gharibeh SX, Hoppe UC, Lichtenauer M. Biomarkers of Atrial Fibrillation Recurrence in Patients with Paroxysmal or Persistent Atrial Fibrillation Following External Direct Current Electrical Cardioversion. Biomedicines 2023; 11:1452. [PMID: 37239123 PMCID: PMC10216298 DOI: 10.3390/biomedicines11051452] [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: 03/31/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Atrial fibrillation (AF) is associated with atrial remodeling, cardiac dysfunction, and poor clinical outcomes. External direct current electrical cardioversion is a well-developed urgent treatment strategy for patients presenting with recent-onset AF. However, there is a lack of accurate predictive serum biomarkers to identify the risks of AF relapse after electrical cardioversion. We reviewed the currently available data and interpreted the findings of several studies revealing biomarkers for crucial elements in the pathogenesis of AF and affecting cardiac remodeling, fibrosis, inflammation, endothelial dysfunction, oxidative stress, adipose tissue dysfunction, myopathy, and mitochondrial dysfunction. Although there is ample strong evidence that elevated levels of numerous biomarkers (such as natriuretic peptides, C-reactive protein, galectin-3, soluble suppressor tumorigenicity-2, fibroblast growth factor-23, turn-over collagen biomarkers, growth differential factor-15) are associated with AF occurrence, the data obtained in clinical studies seem to be controversial in terms of their predictive ability for post-cardioversion outcomes. Novel circulating biomarkers are needed to elucidate the modality of this approach compared with conventional predictive tools. Conclusions: Biomarker-based strategies for predicting events after AF treatment require extensive investigation in the future, especially in the presence of different gender and variable comorbidity profiles. Perhaps, a multiple biomarker approach exerts more utilization for patients with different forms of AF than single biomarker use.
Collapse
Affiliation(s)
- Ozan Demirel
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Alexander E. Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
- Internal Medicine Department, Zaporozhye State Medical University, 69035 Zaporozhye, Ukraine
| | - Moritz Mirna
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Elke Boxhammer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Sarah X. Gharibeh
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Uta C. Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.D.); (M.M.); (E.B.); (S.X.G.); (U.C.H.); (M.L.)
| |
Collapse
|
27
|
Peng J, Chen Q, Wu C. The role of adiponectin in cardiovascular disease. Cardiovasc Pathol 2023; 64:107514. [PMID: 36634790 DOI: 10.1016/j.carpath.2022.107514] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease (CVD) is a common disease that seriously threatens the health of human beings, especially middle-aged and elderly people over 50 years old. It has the characteristics of high prevalence, high disability rate and high mortality rate. Previous studies have shown that adiponectin has therapeutic effects on a variety of CVDs. As a key adipokine, adiponectin, is an abundant peptide-regulated hormone that is mainly released by adipocytes and cardiomyocytes, as well as endothelial and skeletal cells. Adiponectin can protect against CVD by improving lipid metabolism, protecting vascular endothelial cells and inhibiting foam cell formation and vascular smooth muscle cell proliferation. Further investigation of the molecular and cellular mechanisms underlying the adiponectin system may provide new ideas for the treatment of CVD. Herein, this review aims to describe the structure and function of adiponectin and adiponectin receptors, introduce the function of adiponectin in the protection of cardiovascular disease and analyze the potential use and clinical significance of this hormone in the protection and treatment of cardiovascular disease, which shows that adiponectin can be expected to become a new therapeutic target and biomarker for the diagnosis and treatment of CVD.
Collapse
Affiliation(s)
- Jin Peng
- Clinical Medical Research Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qian Chen
- Clinical Medical Research Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Chuncao Wu
- Insititution of Chinese Materia Medica Preparation, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China.
| |
Collapse
|
28
|
Salmons HI, Gow C, Limberg AK, Bettencourt JW, Carstens MF, Payne AN, Morrey ME, Sanchez-Sotelo J, Berry DJ, Dudakovic A, Abdel MP. The Safety of Adiponectin Receptor Agonist AdipoRon in a Rabbit Model of Arthrofibrosis. Tissue Eng Part C Methods 2023; 29:154-159. [PMID: 36924279 PMCID: PMC10122264 DOI: 10.1089/ten.tec.2023.0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
AdipoRon is an adiponectin receptor 1, 2 (ADIPOR1 and ADIPOR2) agonist with numerous reported physiological benefits in murine models of human disease, including a proposed reduction in fibrosis. However, AdipoRon has never been investigated in rabbits, which provide a robust model for orthopedic conditions. We examined the safety of intravenous (IV) AdipoRon in New Zealand White (NZW) female rabbits surgically stressed by a procedure that mimics human arthrofibrosis. Fifteen female NZW rabbits were prospectively studied using increasing AdipoRon doses based on established literature. AdipoRon was dissolved in dimethyl sulfoxide (DMSO), diluted in normal saline, and administered IV preoperatively and for 5 subsequent days postoperatively. The primary outcome was overall toxicity to rabbits, whereas secondary outcomes were change in rabbit weights and hemodynamics and defining acid-base characteristics of the drug formulation. Two rabbits expired during preoperative drug administration at 25 mg/kg. Remaining rabbits received preoperative doses of DMSO (vehicle), 2.5, 5, or 10 mg/kg of AdipoRon without complications. On postoperative day 1, one rabbit sustained a tonic-clonic seizure after their second dose of 10 mg/kg AdipoRon. The remaining 12 rabbits (4 in each group) received six serial doses of vehicle, 2.5, or 5 mg/kg of AdipoRon without adverse effects. All formulations of AdipoRon were within safe physiological pH ranges (4-5). We are the first to report the use of IV AdipoRon in a surgically stressed rabbit model of orthopedic disease. AdipoRon doses of 5 mg/kg or less appear to be well-tolerated in female NZW rabbits. Impact statement We provided the first in vivo toxicity assessment and dose optimization of a new antifibrotic experimental medication, AdipoRon, in a surgically stressed rabbit model of knee arthrofibrosis.
Collapse
Affiliation(s)
- Harold I. Salmons
- Department of Orthopedic Surgery and Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher Gow
- Department of Comparative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Afton K. Limberg
- Department of Orthopedic Surgery and Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mason F. Carstens
- Department of Orthopedic Surgery and Mayo Clinic, Rochester, Minnesota, USA
| | - Ashley N. Payne
- Department of Orthopedic Surgery and Mayo Clinic, Rochester, Minnesota, USA
| | - Mark E. Morrey
- Department of Orthopedic Surgery and Mayo Clinic, Rochester, Minnesota, USA
| | | | - Daniel J. Berry
- Department of Orthopedic Surgery and Mayo Clinic, Rochester, Minnesota, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery and Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew P. Abdel
- Department of Orthopedic Surgery and Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
29
|
Metabolic landscape in cardiac aging: insights into molecular biology and therapeutic implications. Signal Transduct Target Ther 2023; 8:114. [PMID: 36918543 PMCID: PMC10015017 DOI: 10.1038/s41392-023-01378-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Cardiac aging is evident by a reduction in function which subsequently contributes to heart failure. The metabolic microenvironment has been identified as a hallmark of malignancy, but recent studies have shed light on its role in cardiovascular diseases (CVDs). Various metabolic pathways in cardiomyocytes and noncardiomyocytes determine cellular senescence in the aging heart. Metabolic alteration is a common process throughout cardiac degeneration. Importantly, the involvement of cellular senescence in cardiac injuries, including heart failure and myocardial ischemia and infarction, has been reported. However, metabolic complexity among human aging hearts hinders the development of strategies that targets metabolic susceptibility. Advances over the past decade have linked cellular senescence and function with their metabolic reprogramming pathway in cardiac aging, including autophagy, oxidative stress, epigenetic modifications, chronic inflammation, and myocyte systolic phenotype regulation. In addition, metabolic status is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and chronic inflammation. However, further elucidation of the metabolism involvement in cardiac degeneration is still needed. Thus, deciphering the mechanisms underlying how metabolic reprogramming impacts cardiac aging is thought to contribute to the novel interventions to protect or even restore cardiac function in aging hearts. Here, we summarize emerging concepts about metabolic landscapes of cardiac aging, with specific focuses on why metabolic profile alters during cardiac degeneration and how we could utilize the current knowledge to improve the management of cardiac aging.
Collapse
|
30
|
Moyce Gruber BL, Dolinsky VW. The Role of Adiponectin during Pregnancy and Gestational Diabetes. Life (Basel) 2023; 13:301. [PMID: 36836658 PMCID: PMC9958871 DOI: 10.3390/life13020301] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Pregnancy involves a range of metabolic adaptations to supply adequate energy for fetal growth and development. Gestational diabetes (GDM) is defined as hyperglycemia with first onset during pregnancy. GDM is a recognized risk factor for both pregnancy complications and long-term maternal and offspring risk of cardiometabolic disease development. While pregnancy changes maternal metabolism, GDM can be viewed as a maladaptation by maternal systems to pregnancy, which may include mechanisms such as insufficient insulin secretion, dysregulated hepatic glucose output, mitochondrial dysfunction and lipotoxicity. Adiponectin is an adipose-tissue-derived adipokine that circulates in the body and regulates a diverse range of physiologic mechanisms including energy metabolism and insulin sensitivity. In pregnant women, circulating adiponectin levels decrease correspondingly with insulin sensitivity, and adiponectin levels are low in GDM. In this review, we summarize the current state of knowledge about metabolic adaptations to pregnancy and the role of adiponectin in these processes, with a focus on GDM. Recent studies from rodent model systems have clarified that adiponectin deficiency during pregnancy contributes to GDM development. The upregulation of adiponectin alleviates hyperglycemia in pregnant mice, although much remains to be understood for adiponectin to be utilized clinically for GDM.
Collapse
Affiliation(s)
- Brittany L. Moyce Gruber
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM), Research Theme of the Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| | - Vernon W. Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM), Research Theme of the Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| |
Collapse
|
31
|
Kondratieva DS, Afanasiev SA, Muslimova EF. Diabetes mellitus — metabolic preconditioning in protecting the heart from ischemic damage? DIABETES MELLITUS 2022. [DOI: 10.14341/dm12933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The negative impact of diabetes mellitus (DM) on the cardiovascular system has been confirmed by numerous clinical studies. However, there are experimental studies that show an increase in the resistance of the heart to ischemic and reperfusion damage in animals with DM. This phenomenon is characterized by a smaller size of the infarct zone, better preservation of the contractile function of the myocardium, and a lower incidence of ischemic and reperfusion arrhythmias. It is assumed that at a certain stage in the development of DM, a “metabolic window” is formed, in which metabolic alterations at the cellular level trigger adaptive mechanisms that increase the viability of cardiomyocytes. Published data confirm that the magnitude of the protective effect induced by DM is comparable to, and in some cases even exceeds, the effect of the preconditioning phenomenon. It is recognized that the mechanisms that protect the heart from ischemic and reperfusion damage against the background of DM are universal and are associated with the modulation of the antioxidant system, apoptosis factors, pro-inflammatory cytokines, and signaling systems that ensure cell survival. The one of the main pathogenic factor in DM is hyperglycemia, but under stress it plays the role of an adaptive mechanism aimed at meeting the increased energy demand in pathological conditions. Probably, at a certain stage of DM, hyperglycemia becomes a trigger for the development of protective effects and activates not only signaling pathways, but also the restructuring of energy metabolism, which makes it possible to maintain ATP production at a sufficient level to maintain the vital activity of heart cells under ischemia/reperfusion conditions. It is possible that an increased level of glucose, accompanied by the activation of insulin-independent mechanisms of its entry into cells, as well as the availability of this energy substrate, will contribute to a better restoration of energy production in heart cells after a infarction, which, in turn, will significantly reduce the degree of myocardial damage and will help preserve the contractile function of the heart. Identification of the conditions and mechanisms of the cardioprotective phenomenon induced by DM will make it possible to simulate the metabolic state in which the protection of cardiomyocytes from damaging factors is realized.
Collapse
Affiliation(s)
- D. S. Kondratieva
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - S. A. Afanasiev
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - E. F. Muslimova
- Cardiology Research Institute, Tomsk National Research Medical Center
| |
Collapse
|
32
|
Guo J, Feng J, Qu H, Xu H, Zhou H. Potential Drug Targets for Ceramide Metabolism in Cardiovascular Disease. J Cardiovasc Dev Dis 2022; 9:434. [PMID: 36547431 PMCID: PMC9782850 DOI: 10.3390/jcdd9120434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease poses a significant threat to the quality of human life. Metabolic abnormalities caused by excessive caloric intake have been shown to lead to the development of cardiovascular diseases. Ceramides are structural molecules found in biological membranes; they are crucial for cell survival and lipid metabolism, as they maintain barrier function and membrane fluidity. Increasing evidence has demonstrated that ceramide has a strong correlation with cardiovascular disease progression. Nevertheless, it remains a challenge to develop sphingolipids as therapeutic targets to improve the prognosis of cardiovascular diseases. In this review, we summarize the three synthesis pathways of ceramide and other intermediates that are important in ceramide metabolism. Furthermore, mechanistic studies and therapeutic strategies, including clinical drugs and bioactive molecules based on these intermediates, are discussed.
Collapse
Affiliation(s)
- Jiaying Guo
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
| | - Jiling Feng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, No. 1200, Cailun Road, Shanghai 201203, China
| | - Huiyan Qu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
| | - Hongxi Xu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, No. 1200, Cailun Road, Shanghai 201203, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
33
|
Xu Y, Zhu H, Li W, Chen D, Xu Y, Xu A, Ye D. Targeting adipokines in polycystic ovary syndrome and related metabolic disorders: from experimental insights to clinical studies. Pharmacol Ther 2022; 240:108284. [PMID: 36162728 DOI: 10.1016/j.pharmthera.2022.108284] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) affects approximately 15% of women of reproductive age worldwide. It is the most prevalent endocrine disorder with marked risks for female infertility, type 2 diabetes mellitus (T2DM), psychiatric disorders and gynecological cancers. Although the pathophysiology of PCOS remains largely elusive, growing evidence suggests a close link with obesity and its related metabolic disorders. As a highly active endocrine cell population, hypertrophic adipocytes in obesity have disturbed production of a vast array of adipokines, biologically active peptides that exert pleiotropic effects on homeostatic regulation of glucose and lipid metabolism. In parallel with their crucial roles in the pathophysiology of obesity-induced metabolic diseases, adipokines have recently been identified as promising targets for novel therapeutic strategies for multiple diseases. Current treatments for PCOS are suboptimal with insufficient alleviation of all symptoms. Novel findings in adipokine-targeted agents may provide important insight into the development of new drugs for PCOS. This Review presents an overview of the current understanding of mechanisms that link PCOS to obesity and highlights emerging evidence of adipose-ovary crosstalk as a pivotal mediator of PCOS pathogenesis. We summarize recent findings of preclinical and clinical studies that reveal the therapeutic potential of adipokine-targeted novel approaches to PCOS and its related metabolic disorders. We also discuss the critical gaps in knowledge that need to be addressed to guide the development of adipokine-based novel therapies for PCOS.
Collapse
Affiliation(s)
- Yidan Xu
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huiqiu Zhu
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Weiwei Li
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Danxia Chen
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Xu
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Dewei Ye
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
34
|
Hydrogen Sulfide Plays an Important Role by Regulating Endoplasmic Reticulum Stress in Diabetes-Related Diseases. Int J Mol Sci 2022; 23:ijms23137170. [PMID: 35806174 PMCID: PMC9266787 DOI: 10.3390/ijms23137170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
Endoplasmic reticulum (ER) plays important roles in protein synthesis, protein folding and modification, lipid biosynthesis, calcium storage, and detoxification. ER homeostasis is destroyed by physiological and pharmacological stressors, resulting in the accumulation of misfolded proteins, which causes ER stress. More and more studies have shown that ER stress contributes to the pathogenesis of many diseases, such as diabetes, inflammation, neurodegenerative diseases, cancer, and autoimmune diseases. As a toxic gas, H2S has, in recent years, been considered the third most important gas signal molecule after NO and CO. H2S has been found to have many important physiological functions and to play an important role in many pathological and physiological processes. Recent evidence shows that H2S improves the body’s defenses to many diseases, including diabetes, by regulating ER stress, but its mechanism has not yet been fully understood. We therefore reviewed recent studies of the role of H2S in improving diabetes-related diseases by regulating ER stress and carefully analyzed its mechanism in order to provide a theoretical reference for future research.
Collapse
|