1
|
Li S, An J, Zhang T, Chen G, Zhang Z, Guo Z, Dai Z, Cheng X, Cheng S, Xiong X, Wang N, Jiang G, Xu B, Lei H. Integration of network pharmacology, UHPLC-Q exactive orbitrap HRMS technique and metabolomics to elucidate the active ingredients and mechanisms of compound danshen pills in treating hypercholesterolemic rats. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118759. [PMID: 39209003 DOI: 10.1016/j.jep.2024.118759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/18/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hypercholesterolemia (HLC) was a key risk factor for cardiovascular disease (CVD) characterized by elevated cholesterol levels, particularly LDL. While traditional Chinese medicine preparations Compound Danshen Pills(CDP) has been clinically used for hypercholesterolemia and coronary heart disease, its specific therapeutic effect on HLC remains understudied, necessitating further investigation into its mechanisms. AIM OF THE STUDY The aim of this study was to explore the potential of CDP in treating HLC and elucidate its underlying mechanisms and active components. MATERIALS AND METHODS A hypercholesterolemic lipemia rat model induced by a high-fat diet was employed. Network pharmacology combined with UHPLC-Q exactive orbitrap HRMS technique was used to predict the active components, targets and mechanisms of CDP for HLC. Histological analysis and serum biochemical assays were used to assess the therapeutic effect of CDP and its main active ingredient Sa B on hypercholesterolemic lipemia rat model. Immunofluorescence assays and western blotting were used to verify the mechanism of CDP and Sa B in the treatment of HLC. Metabolomics approach was used to demonstrate that CDP and Sa B affected the metabolic profile of HLC. RESULTS Our findings demonstrated that both CDP and its main active ingredient Sa B significantly ameliorated hypercholesterolemic lipemic lesions, reducing levels of TC, LDL, AST, ALT, and ALP. Histological analysis revealed a decrease in lipid droplet accumulation and collagen fiber deposition in the liver, as well as reduced collagen fiber deposition in the aorta. Network pharmacology predicted potential targets such as PPARα and CYP27A1. Immunofluorescence assays and western blotting confirmed that CDP and Sa B upregulated the expression of Adipor1, PPARα and CYP27A1. Metabolomics analyses further indicated improvements in ABC transporters metabolic pathways, with differential metabolites such as riboflavin, taurine, and choline showed regression in levels after CDP treatment and riboflavin, L-Threonine, Thiamine, L-Leucine, and Adenosine showed improved expression after Sa B treatment. CONCLUSION CDP and Sa B have been shown to alleviate high-fat diet-induced hypercholesterolemia by activating the PPAR pathway and improving hepatic lipid metabolism. Our study demonstrated, for the first time, the complex mechanism of CDP, Sa B in the treatment of hypercholesterolemia at the protein and metabolic levels and provided a new reference that could elucidate the pharmacological effects of traditional Chinese medicine on hypercholesterolemia from multiple perspectives.
Collapse
Affiliation(s)
- Shanlan Li
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Jin An
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Tong Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Guangyun Chen
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Zixuan Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Zhuoqian Guo
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Ziqi Dai
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Xuehao Cheng
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Sijin Cheng
- School of Nursing, Beijing University of Chinese Medicine, Beijing, 102488, China
| | | | - Nan Wang
- Aimin Pharmaceutical Group, Henan, 463500, China
| | | | - Bing Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| |
Collapse
|
2
|
Yilihamu Y, Xu R, Jia W, Kukun H, Aihemaiti D, Chang Y, Ding S, Wang Y. Role of long non-coding RNA TCONS_02443383 in regulating cell adhesion and peroxisome proliferator-activated receptor (PPAR) signaling genes in atherosclerosis: A New Zealand white rabbit model study. Gene 2024; 927:148694. [PMID: 38878987 DOI: 10.1016/j.gene.2024.148694] [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: 03/19/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
OBJECTIVE In this study, we performed RNA sequencing (RNA-seq) on the abdominal aorta tissue of New Zealand rabbits and investigated the potential association of lncRNA TCONS_02443383 with the development of AS through bioinformatics analysis of the sequencing data. The obtained results were further validated using quantitative real-time polymerase chain reaction (qRT-PCR). METHOD We induced an AS model in New Zealand rabbits by causing balloon injury to the abdominal aorta vascular wall and administering a high-fat diet. We then upregulated the expression level of the lncRNA TCONS_02443383 by injecting lentiviral plasmids through the ear vein. RNA sequencing (RNA-seq) was performed on the abdominal aorta tissues. We conducted Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway and Gene Ontology (GO) analyses. RESULT The overexpression of the lncRNA TCONS_02443383 led to an upregulation of peroxisome proliferator-activated receptor (PPAR) signaling pathways as well as genes related to cell adhesion. CONCLUSION The overexpression of the lncRNA TCONS_02443383 can inhibit the occurrence and development of AS by upregulating peroxisome proliferator-activated receptor (PPAR) signaling pathways and genes related to cell adhesion.
Collapse
Affiliation(s)
- Yilinuer Yilihamu
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China
| | - Rui Xu
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China
| | - Wenxiao Jia
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China
| | - Hanjiaerbieke Kukun
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China
| | - Dilinuerkezi Aihemaiti
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China
| | - Yifan Chang
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China
| | - Shuang Ding
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China.
| | - Yunling Wang
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China.
| |
Collapse
|
3
|
Wang M, Yang Y, Xu Y. Brain nuclear receptors and cardiovascular function. Cell Biosci 2023; 13:14. [PMID: 36670468 PMCID: PMC9854230 DOI: 10.1186/s13578-023-00962-3] [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: 12/21/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Brain-heart interaction has raised up increasing attentions. Nuclear receptors (NRs) are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of cardiovascular diseases (CVDs), including hypertension, heart failure, atherosclerosis, etc. In this review, we will elaborate recent findings that have established the physiological relevance of brain NRs in the context of cardiovascular function. In addition, we will discuss the currently available evidence regarding the distinct neuronal populations that respond to brain NRs in the cardiovascular control. These findings suggest connections between cardiac control and brain dynamics through NR signaling, which may lead to novel tools for the treatment of pathological changes in the CVDs.
Collapse
Affiliation(s)
- Mengjie Wang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yongjie Yang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
4
|
Upregulated PPARG2 facilitates interaction with demethylated AKAP12 gene promoter and suppresses proliferation in prostate cancer. Cell Death Dis 2021; 12:528. [PMID: 34023860 PMCID: PMC8141057 DOI: 10.1038/s41419-021-03820-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCA) is one of the most common male genitourinary tumors. However, the molecular mechanisms involved in the occurrence and progression of PCA have not been fully clarified. The present study aimed to investigate the biological function and molecular mechanism of the nuclear receptor peroxisome proliferator-activated receptor gamma 2 (PPARG2) in PCA. Our results revealed that PPARG2 was downregulated in PCA, and overexpression of PPARG2 inhibited cell migration, colony formation, invasion and induced cell cycle arrest of PCA cells in vitro. In addition, PPARG2 overexpression modulated the activation of the Akt signaling pathway, as well as inhibited tumor growth in vivo. Moreover, mechanistic analysis revealed that PPARG2 overexpression induced increased expression level of miR-200b-3p, which targeted 3′ UTR of the downstream targets DNMT3A/3B, and facilitated interaction with demethylated AKAP12 gene promoter and suppressed cell proliferation in PCA. Our findings provided the first evidence for a novel PPARG2-AKAP12 axis mediated epigenetic regulatory network. The study identified a molecular mechanism involving an epigenetic modification that could be possibly targeted as an antitumoral strategy against prostate cancer.
Collapse
|
5
|
Zhou Y, Guo Y, Zhu Y, Sun Y, Li W, Li Z, Wei L. Dual PPARγ/ɑ agonist oroxyloside suppresses cell cycle progression by glycolipid metabolism switch-mediated increase of reactive oxygen species levels. Free Radic Biol Med 2021; 167:205-217. [PMID: 33713839 DOI: 10.1016/j.freeradbiomed.2021.02.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/26/2021] [Accepted: 02/24/2021] [Indexed: 12/15/2022]
Abstract
Cancer cells prefers to rely on aerobic glycolysis than pyruvate oxidation to meet the high demand of energy for rapidly proliferation. Peroxisome proliferator-activated receptors (PPARs) are a kind of important ligand-inducible transcription factors and play crucial roles in glucose and lipid metabolism. Careful designing of novel agonists for PPARs, may show improvement with the side effects and also increase the therapeutic value for cancer and other metabolic disorder diseases. Compared with normal human liver cells, lower expression or acitivity of PPARs is observed in hepatocellular carcinoma (HCC). In this study, we show that oroxyloside (OAG) is a new dual agonist of PPARγ/ɑ, and inhibits cell proliferation of HCC based on metabolic switch. Via both PPAR-dependent and PPAR-independent regulations on glycolipid metabolic enzymes, OAG shuts down the catabolism of glucose and promotes fatty acids oxidation to generate acetyl-CoA for TCA cycle and oxidative phosphorylation. The metabolic switch induced by OAG results in a marked increase of reactive oxygen species (ROS) levels, leading to rapid dephosphorylation of RB and cell-cycle arrest in G1 phase. Pyruvate dehydrogenase kinase 4 (PDK4) and β-Oxidation are required for the suppression of cell cycle progression by OAG. Together, our findings provide a new drug candidate and a viable therapeutic strategy for HCC based on metabolic reprogram.
Collapse
Affiliation(s)
- Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Yongjian Guo
- School of Biopharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Yejin Zhu
- School of Medicine & Holistic Integrative Medcine, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, PR China
| | - Yuening Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Wei Li
- Research Center of Basic Medical College, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, PR China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China.
| |
Collapse
|
6
|
Mirza AZ, Althagafi II, Shamshad H. Role of PPAR receptor in different diseases and their ligands: Physiological importance and clinical implications. Eur J Med Chem 2019; 166:502-513. [DOI: 10.1016/j.ejmech.2019.01.067] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 12/15/2022]
|
7
|
Hamilton A, Ly J, Robinson JR, Corder KR, DeMoranville KJ, Schaeffer PJ, Huss JM. Conserved transcriptional activity and ligand responsiveness of avian PPARs: Potential role in regulating lipid metabolism in mirgratory birds. Gen Comp Endocrinol 2018; 268:110-120. [PMID: 30114400 DOI: 10.1016/j.ygcen.2018.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 06/18/2018] [Accepted: 08/07/2018] [Indexed: 01/04/2023]
Abstract
Migratory birds undergo metabolic remodeling in tissues, including increased lipid storage in white adipose and fatty acid uptake and oxidation in skeletal muscle, to optimize energy substrate availability and utilization in preparation for long-distance flight. Different tissues undergo gene expression changes in keeping with their specialized functions and driven by tissue specific transcriptional pathways. Peroxisome proliferator-activated receptors (PPARs) are lipid-activated nuclear receptors that regulate metabolic pathways involved in lipid and glucose utilization or storage in mammals. To examine whether PPARs might mediate fatty acid activation of metabolic gene programs that would be relevant during pre-migratory fattening, we used gray catbird as the focal species. PPAR isoforms cloned from catbird share high amino acid identity with mammalian homologs (% vs human): gcPPARα (88.1%), gcPPARδ (87.3%), gcPPARγ (91.2%). We tested whether gcPPARs activated fatty acid (FA) utilization genes using Lpl and Cpt1b gene promoter-luciferase reporters in mammalian cell lines. In C2C12 mouse myocytes gcPPARα was broadly activated by the saturated and unsaturated FAs tested; while gcPPARδ showed highest activation by the mono-unsaturated FA, 18:1 oleic acid (+80%). In CV-1 monkey kidney cells gcPPARγ responded to the poly-unsaturated fatty acid, 20:5 eicosapentaenoic acid (+60%). Moreover, in agreement with their structural conservation, gcPPARs were activated by isoform selective synthetic agonists similar to the respective mammalian isoform. Adenoviral mediated over-expression of PPARα in C2C12 myocytes induced expression of genes involved in fatty acid transport, including Cd36/Fat, as well as Cpt1b, which mediates a key rate limiting step of mitochondrial β-oxidation. These gene expression changes correlated with increased lipid droplet accumulation in C2C12 myoblasts and differentiated myotubes and enhanced β-oxidation in myotubes. Collectively, the data predict that the PPARs play a conserved role in gray catbirds to regulate lipid metabolism in target tissues that undergo metabolic remodeling throughout the annual migratory cycle.
Collapse
Affiliation(s)
- Angelica Hamilton
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jennifer Ly
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jasmine R Robinson
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Keely R Corder
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | | | | | - Janice M Huss
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
8
|
Sun H, Shao W, Liu H, Jiang Z. Exposure to 2,4-dichlorophenoxyacetic acid induced PPARβ-dependent disruption of glucose metabolism in HepG2 cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:17050-17057. [PMID: 29633193 DOI: 10.1007/s11356-018-1921-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/02/2018] [Indexed: 06/08/2023]
Abstract
2,4-Dichlorophenoxyacetic acid is one of the most widely used herbicides. Its impact on health is increasingly attracting great attentions. This study aimed to investigate the effect of 2,4-dichlorophenoxyacetic acid on glucose metabolism in HepG2 cells and the underlying mechanism. After 24 h exposure to 2,4-dichlorophenoxyacetic acid, glycogen was measured by PAS staining and glucose by ELISA in HepG2 cells. The expression of genes involved in glucose metabolism was measured by real-time PCR, Western blotting, and immunofluorescence. HepG2 cells presented more extracellular glucose consumption and glycogen content after exposed to 2,4-dichlorophenoxyacetic acid. Expression of gluconeogenesis-related genes, FoxO1, and CREB is significantly elevated. Moreover, PPARβ was up-regulated dose-dependently. SiRNA knockdown of PPARβ completely rescued the increase of glycogen accumulation and glucose uptake, and the up-regulation of FOXO1 and CREB expression. Our findings propose novel mechanisms that 2,4-dichlorophenoxyacetic acid causes glucose metabolism dysfunction through PPARβ in HepG2 cells.
Collapse
Affiliation(s)
- Haidong Sun
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 125 Ruijin Er Road, Shanghai, 200025, China
| | - Wentao Shao
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Hui Liu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Zhaoyan Jiang
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 125 Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
9
|
Zha W, Ho HTB, Hu T, Hebert MF, Wang J. Serotonin transporter deficiency drives estrogen-dependent obesity and glucose intolerance. Sci Rep 2017; 7:1137. [PMID: 28442777 PMCID: PMC5430688 DOI: 10.1038/s41598-017-01291-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/27/2017] [Indexed: 12/31/2022] Open
Abstract
Depression and use of antidepressant medications are both associated with increased risk of obesity, potentially attributed to a reduced serotonin transporter (SERT) function. However, how SERT deficiency promotes obesity is unknown. Here, we demonstrated that SERT−/− mice display abnormal fat accumulation in both white and brown adipose tissues, glucose intolerance and insulin resistance while exhibiting suppressed aromatase (Cyp19a1) expression and reduced circulating 17β-estradiol levels. 17β-estradiol replacement in SERT−/− mice reversed the obesity and glucose intolerance, supporting a role for estrogen in SERT deficiency-associated obesity and glucose intolerance. Treatment of wild type mice with paroxetine, a chemical inhibitor of SERT, also resulted in Cyp19a1 suppression, decreased circulating 17β-estradiol levels, abnormal fat accumulation, and glucose intolerance. Such effects were not observed in paroxetine-treated SERT−/− mice. Conversely, pregnant SERT−/− mice displayed normalized estrogen levels, markedly reduced fat accumulation, and improved glucose tolerance, which can be eliminated by an antagonist of estrogen receptor α (ERα). Together, these findings support that estrogen suppression is involved in SERT deficiency-induced obesity and glucose intolerance, and suggest approaches to restore 17β-estradiol levels as a novel treatment option for SERT deficiency associated obesity and metabolic abnormalities.
Collapse
Affiliation(s)
- Weibin Zha
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Horace T B Ho
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Tao Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Mary F Hebert
- Department of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA. .,Nutrition Obesity Research Center, University of Washington, Seattle, WA, USA.
| |
Collapse
|
10
|
An HJ, Lee B, Kim DH, Lee EK, Chung KW, Park MH, Jeong HO, Kim SM, Moon KM, Kim YR, Kim SJ, Yun HY, Chun P, Yu BP, Moon HR, Chung HY. Physiological characterization of a novel PPAR pan agonist, 2-(4-(5,6-methylenedioxybenzo[d]thiazol-2-yl)-2-methylphenoxy)-2-methylpropanoic acid (MHY2013). Oncotarget 2017; 8:16912-16924. [PMID: 28129657 PMCID: PMC5370010 DOI: 10.18632/oncotarget.14818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/27/2016] [Indexed: 02/03/2023] Open
Abstract
Recently, agonists targeting multiple peroxisome proliferator-activated receptors (PPARs) have been developed to improve metabolic disorders and minimize the side effects of selective PPAR agonists such as weight gain and dyslipidemia. We newly synthesized six 2-methyl-2-(o-tolyloxy)propanoic acid derivatives based on the structure of a well-known PPAR pan agonist, bezafibrate. Of six compounds, MHY2013 was screened as the strongest activator of three PPAR subtypes based on protein docking simulation and luciferase assays. When treated orally in db/db mice, MHY2013 ameliorated obesity-induced insulin resistance, dyslipidemia, and hepatic steatosis without changes of the body weight and levels of liver and kidney injury markers. MHY2013 decreased the serum triglyceride and fatty acid levels, which is associated with an increase in fatty acid oxidation signaling in the liver and thermogenic signaling on white adipose tissue, respectively. Furthermore, MHY2013 markedly increased serum levels of insulin-sensitizing hormones including fibroblast growth factor 21 (FGF21) and adiponectin. In conclusion, this study suggests that, MHY2013 is a novel PPAR pan agonist that improves obesity-induced insulin resistance, dyslipidemia and hepatic steatosis and elevates insulin-sensitizing hormones in the blood.
Collapse
Affiliation(s)
- Hye Jin An
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Bonggi Lee
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea.,Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea
| | - Dae Hyun Kim
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Eun Kyeong Lee
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Ki Wung Chung
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Min Hi Park
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Hyoung Oh Jeong
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Seong Min Kim
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Kyoung Mi Moon
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Ye Ra Kim
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Seong Jin Kim
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Hwi Young Yun
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Pusoon Chun
- College of Pharmacy, Inje University, Gyeongsangnam-do 50834, Republic of Korea
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | - Hyung Ryong Moon
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| | - Hae Young Chung
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
11
|
Peroxisome Proliferator-Activated Receptor Modulation during Metabolic Diseases and Cancers: Master and Minions. PPAR Res 2016; 2016:6517313. [PMID: 28115924 PMCID: PMC5225385 DOI: 10.1155/2016/6517313] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/12/2016] [Indexed: 12/11/2022] Open
Abstract
The prevalence of obesity and metabolic diseases (such as type 2 diabetes mellitus, dyslipidaemia, and cardiovascular diseases) has increased in the last decade, in both industrialized and developing countries. This also coincided with our observation of a similar increase in the prevalence of cancers. The aetiology of these diseases is very complex and involves genetic, nutritional, and environmental factors. Much evidence indicates the central role undertaken by peroxisome proliferator-activated receptors (PPARs) in the development of these disorders. Due to the fact that their ligands could become crucial in future target-therapies, PPARs have therefore become the focal point of much research. Based on this evidence, this narrative review was written with the purpose of outlining the effects of PPARs, their actions, and their prospective uses in metabolic diseases and cancers.
Collapse
|
12
|
Pleiotropic Actions of Peroxisome Proliferator-Activated Receptors (PPARs) in Dysregulated Metabolic Homeostasis, Inflammation and Cancer: Current Evidence and Future Perspectives. Int J Mol Sci 2016; 17:ijms17070999. [PMID: 27347932 PMCID: PMC4964375 DOI: 10.3390/ijms17070999] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/13/2016] [Accepted: 06/21/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) have demonstrated a lot of important effects in the regulation of glucose and lipid metabolism and in the correct functioning of adipose tissue. Recently, many studies have evaluated a possible effect of PPARs on tumor cells. The purpose of this review is to describe the effects of PPARs, their action and their future prospective; METHODS Narrative review aimed to synthesize cutting-edge evidence retrieved from searches of computerized databases; RESULTS PPARs play a key role in metabolic diseases, which include several cardiovascular diseases, insulin resistance, type 2 diabetes, metabolic syndrome, impaired immunity and the increasing risk of cancer; in particular, PPARα and PPARβ/δ mainly enable energy combustion, while PPARγ contributes to energy storage by enhancing adipogenesis; CONCLUSION PPAR agonists could represent interesting types of molecules that can treat not only metabolic diseases, but also inflammation and cancer. Additional research is needed for the identification of high-affinity, high-specificity agonists for the treatment of obesity, type 2 diabetes (T2DM) and other metabolic diseases. Further studies are needed also to elucidate the role of PPARs in cancer.
Collapse
|
13
|
Teng Z, Jiang L, Hu Q, He Y, Guo Z, Wu Y, Huang Z, Cao F, Cheng C, Sun X, Guo Z. Peroxisome Proliferator-Activated Receptor β/δ Alleviates Early Brain Injury After Subarachnoid Hemorrhage in Rats. Stroke 2015; 47:196-205. [PMID: 26628385 DOI: 10.1161/strokeaha.115.011701] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/19/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Early brain injury is proposed to be the primary cause of the poor outcome after subarachnoid hemorrhage (SAH), which is closely related to the neural apoptosis. To date, the relationship between peroxisome proliferator-activated receptor β/δ (PPARβ/δ) and nuclear factor-κB/matrix metalloproteinase-9 (NF-κB/MMP-9) pathway, both of which are closely related to apoptotic effects, has been poorly studied in SAH. The present study was undertaken to evaluate the effects of PPARβ/δ on early brain injury and NF-κB/MMP-9 pathway after SAH in rats. METHODS SAH model was established by injecting nonheparinized autologous arterial blood into the prechiasmatic cistern in male Sprague-Dawley rats. Adenoviruses or small interfering RNAs were injected into the right lateral cerebral ventricle to, respectively, up- or downregulate PPARβ/δ expression before SAH. All animals were assessed with a neurological score and then killed at 24 hours after SAH surgery. The indexes of brain water content, blood-brain barrier permeability, and apoptosis were used to detect brain injury. The expression of PPARβ/δ, NF-κB, and MMP-9 were measured by immunohistochemistry, gelatin zymography, and Western Blot methods, respectively. In addition, GW0742, a specific agonist of PPARβ/δ, was used to treat SAH in rats, the effects of which were evaluated by neurological scoring and Evans blue extravasation. RESULTS Overexpression of PPARβ/δ by adenoviruses treatment significantly ameliorated brain injury with improvement in neurological deficits, brain edema, blood-brain barrier impairment, and neural cell apoptosis at 24 hours after SAH in rats, whereas downregulation of PPARβ/δ by small interfering RNAs administration resulted in the reverse effects of the above. The expression levels of NF-κB and MMP-9 were markedly downregulated when PPARβ/δ increased after PPARβ/δ adenovirus transfection and upregulated when PPARβ/δ decreased by PPARβ/δ small interfering RNAs treatment. Moreover, GW0742 improved neurological deficits and reduced Evans blue extravasation at 24 hours after SAH. CONCLUSIONS PPARβ/δ's overexpression may attenuate early brain injury after rats' SAH administration, which reduces neural apoptosis possibly through blocking NF-κB/MMP-9 pathway.
Collapse
Affiliation(s)
- Zhipeng Teng
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Li Jiang
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Qin Hu
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Yue He
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Zhenni Guo
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Yue Wu
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Zhijian Huang
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Fang Cao
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Chongjie Cheng
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Xiaochuan Sun
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo).
| | - Zongduo Guo
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo).
| |
Collapse
|
14
|
Zebrafish as a Model to Study the Role of Peroxisome Proliferating-Activated Receptors in Adipogenesis and Obesity. PPAR Res 2015; 2015:358029. [PMID: 26697060 PMCID: PMC4677228 DOI: 10.1155/2015/358029] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/29/2015] [Accepted: 11/05/2015] [Indexed: 02/05/2023] Open
Abstract
The Peroxisome Proliferator-Activated Receptors (PPARs) PPARA and PPARD are regulators of lipid metabolism with important roles in energy release through lipid breakdown, while PPARG plays a key role in lipid storage and adipogenesis. The aim of this review is to describe the role of PPARs in lipid metabolism, adipogenesis, and obesity and evaluate the zebrafish as an emerging vertebrate model to study the function of PPARs. Zebrafish are an appropriate model to study human diseases, including obesity and related metabolic diseases, as pathways important for adipogenesis and lipid metabolism which are conserved between mammals and fish. This review synthesizes knowledge on the role of PPARs in zebrafish and focuses on the putative function of PPARs in zebrafish adipogenesis. Using in silico analysis, we confirm the presence of five PPARs (pparaa, pparab, pparda, ppardb, and pparg) in the zebrafish genome with 67–74% identity to human and mouse PPARs. During development, pparda/b paralogs and pparg show mRNA expression around the swim bladder and pancreas, the region where adipocytes first develop, whereas pparg is detectable in adipocytes at 15 days post fertilization (dpf). This review indicates that the zebrafish is a promising model to investigate the specific functions of PPARs in adipogenesis and obesity.
Collapse
|
15
|
Rachid TL, Penna-de-Carvalho A, Bringhenti I, Aguila MB, Mandarim-de-Lacerda CA, Souza-Mello V. Fenofibrate (PPARalpha agonist) induces beige cell formation in subcutaneous white adipose tissue from diet-induced male obese mice. Mol Cell Endocrinol 2015; 402:86-94. [PMID: 25576856 DOI: 10.1016/j.mce.2014.12.027] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 12/07/2014] [Accepted: 12/30/2014] [Indexed: 12/18/2022]
Abstract
Browning is characterized by the formation of beige/brite fat depots in subcutaneous white adipose tissue (sWAT). This study aimed to examine whether the chronic activation of PPARalpha by fenofibrate could induce beige cell depots in the sWAT of diet-induced obese mice. High-fat fed animals presented overweight, insulin resistance and displayed adverse sWAT remodeling. Fenofibrate significantly attenuated these parameters. Treated groups demonstrated active UCP-1 beige cell clusters within sWAT, confirmed through higher gene expression of PPARalpha, PPARbeta, PGC1alpha, BMP8B, UCP-1, PRDM16 and irisin in treated groups. PPARalpha activation seems to be pivotal to trigger browning through irisin induction and UCP-1 transcription, indicating that fenofibrate increased the expression of genes typical of brown adipose tissue (BAT) in the sWAT, characterizing the formation of beige cells. These findings put forward a possible role of PPARalpha as a promising therapeutic for metabolic diseases via beige cell induction.
Collapse
Affiliation(s)
- Tamiris Lima Rachid
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Aline Penna-de-Carvalho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Isabele Bringhenti
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Marcia B Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Fu H, Desvergne B, Ferrari S, Bonnet N. Impaired musculoskeletal response to age and exercise in PPARβ(-/-) diabetic mice. Endocrinology 2014; 155:4686-96. [PMID: 25279796 PMCID: PMC4239426 DOI: 10.1210/en.2014-1585] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Fragility fractures are recognized complication of diabetes, but yet the underlying mechanisms remain poorly understood. This is particularly pronounced in type 2 diabetes in which the propensity to fall is increased but bone mass is not necessarily low. Thus, whether factors implicated in the development of insulin resistance and diabetes directly impact on the musculoskeletal system remains to be investigated. PPARβ(-/-) mice have reduced metabolic activity and are glucose intolerant. We examined changes in bone and muscle in PPARβ(-/-) mice and investigated both the mechanism behind those changes with age as well as their response to exercise. Compared with their wild type, PPARβ(-/-) mice had an accelerated and parallel decline in both muscle and bone strength with age. These changes were accompanied by increased myostatin expression, low bone formation, and increased resorption. In addition, mesenchymal cells from PPARβ(-/-) had a reduced proliferation capacity and appeared to differentiate into more of an adipogenic phenotype. Concomitantly we observed an increased expression of PPARγ, characteristic of adipocytes. The anabolic responses of muscle and bone to exercise were also diminished in PPARβ(-/-) mice. The periosteal bone formation response to direct bone compression was, however, maintained, indicating that PPARβ controls periosteal bone formation through muscle contraction and/or metabolism. Taken together, these data indicate that PPARβ deficiency leads to glucose intolerance, decreased muscle function, and reduced bone strength. On a molecular level, PPARβ appears to regulate myostatin and PPARγ expression in muscle and bone, thereby providing potential new targets to reverse bone fragility in patients with metabolic disturbances.
Collapse
Affiliation(s)
- He Fu
- Division of Bone Diseases (S.F., N.B.), Department of Internal Medicine Specialties, Geneva University Hospital, and Faculty of Medicine, Geneva 14, CH-1211 Geneva, Switzerland; and Center of Integrative Genomics (H.F., B.D.), Genopode, Lausanne Faculty of Biology and Medicine, CH-1015 Lausanne, Switzerland
| | | | | | | |
Collapse
|
17
|
Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications--a review. Nutr J 2014; 13:17. [PMID: 24524207 PMCID: PMC3943808 DOI: 10.1186/1475-2891-13-17] [Citation(s) in RCA: 802] [Impact Index Per Article: 80.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/07/2014] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptors are expressed in many tissues, including adipocytes, hepatocytes, muscles and endothelial cells; however, the affinity depends on the isoform of PPAR, and different distribution and expression profiles, which ultimately lead to different clinical outcomes. Because they play an important role in lipid and glucose homeostasis, they are called lipid and insulin sensors. Their actions are limited to specific tissue types and thus, reveal a characteristic influence on target cells. PPARα mainly influences fatty acid metabolism and its activation lowers lipid levels, while PPARγ is mostly involved in the regulation of the adipogenesis, energy balance, and lipid biosynthesis. PPARβ/δ participates in fatty acid oxidation, mostly in skeletal and cardiac muscles, but it also regulates blood glucose and cholesterol levels. Many natural and synthetic ligands influence the expression of these receptors. Synthetic ligands are widely used in the treatment of dyslipidemia (e.g. fibrates--PPARα activators) or in diabetes mellitus (e.g. thiazolidinediones--PPARγ agonists). New generation drugs--PPARα/γ dual agonists--reveal hypolipemic, hypotensive, antiatherogenic, anti-inflammatory and anticoagulant action while the overexpression of PPARβ/δ prevents the development of obesity and reduces lipid accumulation in cardiac cells, even during a high-fat diet. Precise data on the expression and function of natural PPAR agonists on glucose and lipid metabolism are still missing, mostly because the same ligand influences several receptors and a number of reports have provided conflicting results. To date, we know that PPARs have the capability to accommodate and bind a variety of natural and synthetic lipophilic acids, such as essential fatty acids, eicosanoids, phytanic acid and palmitoylethanolamide. A current understanding of the effects of PPARs, their molecular mechanisms and the role of these receptors in nutrition and therapeutic treatment are delineated in this paper.
Collapse
Affiliation(s)
- Bogna Grygiel-Górniak
- Department of Bromatology and Human Nutrition, University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
18
|
Abstract
Diabetes is a complex disease defined by hyperglycaemia; however, strong associations with abdominal obesity, hypertension and dyslipidaemia contribute to the high risk of cardiovascular disease. Although aggressive glycaemic control reduces microvascular complications, the evidence for macrovascular complications is less certain. The theoretical benefits of the mode of action of peroxisome proliferator-activated receptor (PPAR) agonists are clear. In clinical practice, PPAR-α agonists such as fibrates improve dyslipidaemia, while PPAR-γ agonists such as thiazolidinediones improve insulin resistance and diabetes control. However, although these agents are traditionally classed according to their target, they have different and sometimes conflicting clinical benefit and adverse event profiles. It is speculated that this is because of differing properties and specificities for the PPAR receptors (each of which targets specific genes). This is most obvious in the impact on cardiovascular outcomes--in clinical trials pioglitazone appeared to reduce cardiovascular events, whereas rosiglitazone potentially increased the risk of myocardial infarction. The development of a dual PPAR-α/γ agonist may prove beneficial in effectively managing glycaemic control and improving dyslipidaemia in patients with type 2 diabetes. Yet, development of agents such as muraglitazar and tesaglitazar has been hindered by various serious adverse events. Aleglitazar, a balanced dual PPAR-α/γ agonist, is currently the most advanced in clinical development and has shown promising results in phase II clinical trials with beneficial effects on glucose and lipid variables. A phase III study, ALECARDIO, is ongoing and will establish whether improvements in laboratory test profiles translate into an improvement in cardiovascular outcomes.
Collapse
Affiliation(s)
- J P H Wilding
- Department of Obesity & Endocrinology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
19
|
Toth PM, Naruhn S, Pape VFS, Dörr SMA, Klebe G, Müller R, Diederich WE. Development of improved PPARβ/δ inhibitors. ChemMedChem 2011; 7:159-70. [PMID: 22025402 DOI: 10.1002/cmdc.201100408] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Indexed: 11/08/2022]
Abstract
GSK0660 (1) is the first peroxisome proliferator-activated receptor (PPAR) β/δ-selective inhibitory ligand described in the literature. Based on its structure, we designed and synthesized a series of modified compounds to establish preliminary structure-activity relationships. Most beneficial for increased binding affinity towards the PPARβ/δ ligand binding domain was the replacement of the 4'-aminophenyl substituent by medium-length n-alkyl chains, such as n-butyl or iso-pentyl. These compounds show activity down to the one-digit nanomolar range, thus possessing up to a tenfold higher binding affinity compared with GSK0660. Additionally, the subtype-specific inhibition of PPARβ/δ was confirmed in a cell-based assay making these compounds invaluable tools for the further exploration of the functions of PPARβ/δ.
Collapse
Affiliation(s)
- Philipp M Toth
- Institut für Pharmazeutische Chemie, Philipps-Universität Marburg, Marburg, Germany
| | | | | | | | | | | | | |
Collapse
|