1
|
Mansouri M, Imenshahidi M, Rameshrad M, Hosseinzadeh H. Effects of Tinospora cordifolia (giloy) on metabolic syndrome components: a mechanistic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4979-5009. [PMID: 39731594 DOI: 10.1007/s00210-024-03642-2] [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: 09/10/2024] [Accepted: 11/15/2024] [Indexed: 12/30/2024]
Abstract
Metabolic syndrome is a cluster of some conditions such as high blood sugar, high blood triglycerides, low HDL cholesterol, abdominal obesity, and high blood pressure. Introducing a drug or a food that manages the majority of these medical conditions is invaluable. Tinospora cordifolia, known as guduchi and giloy, is a medicinal herb in ayurvedic medicine that is used in the treatment of various diseased conditions and also as a food for the maintenance of health. Here, we reviewed the current evidence supporting the role of giloy in the development and treatment of metabolic syndrome components. Appropriate articles that have been published until May 2024 were carefully extracted from PubMed, Scopus, and WOS databases to write a narrative review systematically. Gathered data showed the beneficial effects of giloy on metabolic syndrome components: hyperlipidemia, obesity, atherosclerosis, hypertension, and especially diabetes mellitus. As diabetes and insulin resistance seem to be a central feature of metabolic syndrome and in turn, can cause dyslipidemia, obesity, and, atherosclerosis, these beneficial effects are predictable with the anti-diabetogenic property of giloy. In this review, the main mechanisms of action of giloy in metabolic syndrome components are discussed. Based on the results, although giloy has been less investigated, considerable studies provide evidence of its beneficial effects on different components of metabolic syndrome. Relevant clinical trials are necessary to validate the mentioned effects, safety, and optimum dose of this herbal medicine and its components in managing different components of metabolic syndrome and transition from bench to bedside.
Collapse
Affiliation(s)
- Mehran Mansouri
- Student Research Committee, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Imenshahidi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rameshrad
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Odimegwu CL, Uwaezuoke SN, Chikani UN, Mbanefo NR, Adiele KD, Nwolisa CE, Eneh CI, Ndiokwelu CO, Okpala SC, Ogbuka FN, Odo KE, Ohuche IO, Obiora-Izuka CE. Targeting the Epigenetic Marks in Type 2 Diabetes Mellitus: Will Epigenetic Therapy Be a Valuable Adjunct to Pharmacotherapy? Diabetes Metab Syndr Obes 2024; 17:3557-3576. [PMID: 39323929 PMCID: PMC11423826 DOI: 10.2147/dmso.s479077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/03/2024] [Indexed: 09/27/2024] Open
Abstract
Although genetic, environmental, and lifestyle factors largely contribute to type 2 diabetes mellitus (T2DM) risk, the role of epigenetics in its pathogenesis is now well established. The epigenetic mechanisms in T2DM mainly consist of DNA methylation, histone modifications and regulation by noncoding RNAs (ncRNAs). For instance, DNA methylation at CpG islands in the promoter regions of specific genes encoding insulin signaling and glucose metabolism suppresses these genes. Modulating the enzyme mediators of these epigenetic marks aims to restore standard gene expression patterns and improve glycemic control. In targeting these epigenetic marks, using epigenetic drugs such as DNA methyltransferase (DNAMT), histone deacetylase (HDAC) and histone acetyltransferase (HAT) inhibitors has led to variable success in humans and experimental murine models. Specifically, the United States' Food and Drug Administration (US FDA) has approved DNAMT inhibitors like 5-azacytidine and 5-aza-2'-deoxycytidine for use in diabetic retinopathy: a T2DM microvascular complication. These DNAMT inhibitors block the genes for methylation of mitochondrial superoxide dismutase 2 (SOD2) and matrix metallopeptidase 9 (MMP-9): the epigenetic marks in diabetic retinopathy. Traditional pharmacotherapy with metformin also have epigenetic effects in T2DM and positively alter disease outcomes when combined with epigenetic drugs like DNAMT and HDAC inhibitors, raising the prospect of using epigenetic therapy as a valuable adjunct to pharmacotherapy. However, introducing small interfering RNAs (siRNAs) in cells to silence specific target genes remains in the exploratory phase. Future research should focus on regulating gene expression in T2DM using long noncoding RNA (lncRNA) molecules, another type of ncRNA. This review discusses the epigenetics of T2DM and that of its macro- and microvascular complications, and the potential benefits of combining epigenetic therapy with pharmacotherapy for optimal results.
Collapse
Affiliation(s)
- Chioma Laura Odimegwu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Samuel Nkachukwu Uwaezuoke
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ugo N Chikani
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ngozi Rita Mbanefo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ken Daberechi Adiele
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | - Chizoma Ihuarula Eneh
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Chibuzo Obiora Ndiokwelu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Somkenechi C Okpala
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Francis N Ogbuka
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Kenneth E Odo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | | |
Collapse
|
3
|
Kumagai H, Kim SJ, Miller B, Natsume T, Wan J, Kumagai ME, Ramirez R, Lee SH, Sato A, Mehta HH, Yen K, Cohen P. Mitochondrial-derived microprotein MOTS-c attenuates immobilization-induced skeletal muscle atrophy by suppressing lipid infiltration. Am J Physiol Endocrinol Metab 2024; 326:E207-E214. [PMID: 38170165 PMCID: PMC11196098 DOI: 10.1152/ajpendo.00285.2023] [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: 09/01/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024]
Abstract
Mitochondrial open reading frame of the 12S ribosomal RNA type-c (MOTS-c), a mitochondrial microprotein, has been described as a novel regulator of glucose and lipid metabolism. In addition to its role as a metabolic regulator, MOTS-c prevents skeletal muscle atrophy in high fat-fed mice. Here, we examined the preventive effect of MOTS-c on skeletal muscle mass, using an immobilization-induced muscle atrophy model, and explored its underlying mechanisms. Male C57BL/6J mice (10 wk old) were randomly assigned to one of the three experimental groups: nonimmobilization control group (sterilized water injection), immobilization control group (sterilized water injection), and immobilization and MOTS-c-treated group (15 mg/kg/day MOTS-c injection). We used casting tape for the immobilization experiment. After 8 days of the experimental period, skeletal muscle samples were collected and used for Western blotting, RNA sequencing, and lipid and collagen assays. Immobilization reduced ∼15% of muscle mass, whereas MOTS-c treatment attenuated muscle loss, with only a 5% reduction. MOTS-c treatment also normalized phospho-AKT, phospho-FOXO1, and phospho-FOXO3a expression levels and reduced circulating inflammatory cytokines, such as interleukin-1b (IL-1β), interleukin-6 (IL-6), chemokine C-X-C motif ligand 1 (CXCL1), and monocyte chemoattractant protein 1 (MCP-1), in immobilized mice. Unbiased RNA sequencing and its downstream analyses demonstrated that MOTS-c modified adipogenesis-modulating gene expression within the peroxisome proliferator-activated receptor (PPAR) pathway. Supporting this observation, muscle fatty acid levels were lower in the MOTS-c-treated group than in the casted control mice. These results suggest that MOTS-c treatment inhibits skeletal muscle lipid infiltration by regulating adipogenesis-related genes and prevents immobilization-induced muscle atrophy.NEW & NOTEWORTHY MOTS-c, a mitochondrial microprotein, attenuates immobilization-induced skeletal muscle atrophy. MOTS-c treatment improves systemic inflammation and skeletal muscle AKT/FOXOs signaling pathways. Furthermore, unbiased RNA sequencing and subsequent assays revealed that MOTS-c prevents lipid infiltration in skeletal muscle. Since lipid accumulation is one of the common pathologies among other skeletal muscle atrophies induced by aging, obesity, cancer cachexia, and denervation, MOTS-c treatment could be effective in other muscle atrophy models as well.
Collapse
Affiliation(s)
- Hiroshi Kumagai
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Su-Jeong Kim
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Brendan Miller
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Toshiharu Natsume
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
- Faculty of Medicine, Tokai University, Kanagawa, Japan
| | - Junxiang Wan
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Michi Emma Kumagai
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, California, United States
| | - Ricardo Ramirez
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Shin Hyung Lee
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Ayaka Sato
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Hemal H Mehta
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Kelvin Yen
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| | - Pinchas Cohen
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States
| |
Collapse
|
4
|
Kwon J, Aoki Y, Takahashi H, Nakata R, Kawarasaki S, Ni Z, Yu R, Inoue H, Inoue K, Kawada T, Goto T. Inflammation-induced nitric oxide suppresses PPARα expression and function via downregulation of Sp1 transcriptional activity in adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194987. [PMID: 37739218 DOI: 10.1016/j.bbagrm.2023.194987] [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: 06/14/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
The activation of peroxisome proliferator-activated receptor alpha (PPARα), a ligand-dependent transcription factor that regulates lipid oxidation-related genes, has been employed to treat hyperlipidemia. Emerging evidence indicates that Ppara gene expression decreases in adipose tissue under obese conditions; however, the underlying molecular mechanisms remain elusive. Here, we demonstrate that nitric oxide (NO) suppresses Ppara expression by regulating its promoter activity via suppression of specificity protein 1 (Sp1) transcriptional activity in adipocytes. NO derived from lipopolysaccharide (LPS) -activated macrophages or a NO donor (NOR5) treatment, suppressed Ppara mRNA expression in 10T1/2 adipocytes. In addition, Ppara transcript levels were reduced in the white adipose tissue (WAT) in both acute and chronic inflammation mouse models; however, such suppressive effects were attenuated via a nitric oxide synthase 2 (NOS2) inhibitor. Endoplasmic reticulum (ER) stress inhibitors attenuated the NO-induced repressive effects on Ppara gene expression in 10T1/2 adipocytes. Promoter mutagenesis and chromatin immunoprecipitation assays revealed that NO decreased the Sp1 occupancy in the proximal promoter regions of the Ppara gene, which might partially result from the reduced Sp1 expression levels by NO. This study delineated the molecular mechanism that modulates Ppara gene transcription upon NO stimulation in white adipocytes, suggesting a possible mechanism for the transcriptional downregulation of Ppara in WAT under obese conditions.
Collapse
Affiliation(s)
- Jungin Kwon
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Yumeko Aoki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Haruya Takahashi
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Rieko Nakata
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Satoko Kawarasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Zheng Ni
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Hiroyasu Inoue
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan; Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| | - Teruo Kawada
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan; Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan; Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
5
|
Lee S, Chung MJ, Ahn M, Park HJ, Wang EK, Guon T, Kee HJ, Ku CR, Na K. Surfactant-like photosensitizer for endoscopic duodenal ablation: Modulating meal-stimulated incretin hormones in obese and type 2 diabetes. Biomaterials 2023; 302:122336. [PMID: 37778055 DOI: 10.1016/j.biomaterials.2023.122336] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Duodenal ablation improves glycaemic control and weight loss, so it has been applied using hydrothermal catheters in obese and type 2 diabetes patients, indicating similar mechanisms and therapeutic effects as bariatric surgeries. Endoscopic photodynamic therapy is an innovative procedure that easily accessible to endocrine or gastrointestinal organs, so it is critical for the sprayed photosensitizer (PS) to long-term interact with target tissues for enhancing its effects. Surfactant-like PS was more stable in a wide range of pH and 2.8-fold more retained in the duodenum at 1 h than hydrophilic PS due to its amphiphilic property. Endoscopic duodenal ablation using surfactant-like PS was performed in high fat diet induced rat models, demonstrating body weight loss, enhanced insulin sensitivity, and modulation of incretin hormones. Locoregional ablation of duodenum could affect the profiles of overall intestinal cells secreting meal-stimulated hormones and further the systemic glucose and lipid metabolism, regarding gut-brain axis. Our strategy suggests a potential for a treatment of minimally invasive bariatric and metabolic therapy if accompanied by detailed clinical trials.
Collapse
Affiliation(s)
- Sanghee Lee
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Minji Ahn
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Hyun Jin Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eun Kyung Wang
- Endocrinology, Institute of Endocrine Research, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Taeeun Guon
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun Jung Kee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Cheol Ryong Ku
- Endocrinology, Institute of Endocrine Research, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Kun Na
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
| |
Collapse
|
6
|
Wang X, Zhao T, Ma A. Genetic Mechanism of Tissue-Specific Expression of PPAR Genes in Turbot ( Scophthalmus maximus) at Different Temperatures. Int J Mol Sci 2022; 23:ijms232012205. [PMID: 36293062 PMCID: PMC9603064 DOI: 10.3390/ijms232012205] [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: 08/18/2022] [Revised: 09/15/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we used PCR to measure the levels of the peroxisome proliferator activated receptor genes PPARα1, PPARα2, PPARβ, and PPARγ in the intestine, liver, gill, heart, kidney, brain, muscle, spleen, skin, and stomach of turbot (Scophthalmus maximus) cultured under different temperature conditions (14, 20, 23, 25, and 28 °C). We used split-split-plot (SSP) analysis of variance, additive main effects and multiplicative interaction (AMMI) analysis, and genotype main effects and genotype × environment interaction (GGE) biplot analysis to evaluate the genotype × tissue interaction effects on gene expression. The results of the SSP analysis of variance showed that temperature and tissue × gene have highly significant (p < 0.01) effect on the expression of S. maximus PPAR genes. The AMMI analysis results revealed that the expression of PPAR genes at the appropriate temperature (14 °C) mainly depended on genotype × tissue interaction and tissue effects. Under stress temperatures, genotype effects, tissue effects, and genotype × tissue interaction, all had significant effects on the expression of PPAR genes. The contribution of the genotype effect slowly increased with increasing temperature; it increased faster at 20 °C and then slowly declined at 25 °C. The contribution of the tissue effect slowly increased from 14 to 20 °C, where it sharply decreased, and then it stabilized after a slight fluctuation. The contribution of the genotype × tissue interaction effect showed a fluctuating upward trend throughout the experiment, and it had a significant impact on PPAR gene expression. The key temperature at which the three effects changed was 20 °C, indicating that it is the limit temperature for active lipid metabolism under high-temperature stress. The GGE biplot analysis results showed that under suitable water temperature, the expression difference of PPAR genes in the liver was the largest; at 20 and 23 °C, the expression difference in the gill was the largest; and at 25 and 28 °C, the expression difference in the brain was the largest. Overall, our results suggest that the mechanism responsible for PPAR gene expression under the three high temperatures (23, 25, and 28 °C) was relatively consistent, but it differed from that at 20 °C.
Collapse
Affiliation(s)
- Xinan Wang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| | - Tingting Zhao
- School of Fisheries, Zhejiang Ocean University, Zhoushan 316022, China
| | - Aijun Ma
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
- Correspondence:
| |
Collapse
|
7
|
Baumann A, Burger K, Brandt A, Staltner R, Jung F, Rajcic D, Lorenzo Pisarello MJ, Bergheim I. GW9662, a peroxisome proliferator-activated receptor gamma antagonist, attenuates the development of non-alcoholic fatty liver disease. Metabolism 2022; 133:155233. [PMID: 35654114 DOI: 10.1016/j.metabol.2022.155233] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Insulin resistance is among the key risk factors for the development of non-alcoholic fatty liver disease (NAFLD). Recently, it has been reported that GW9662, shown to be a potent peroxisome proliferator-activated receptor gamma (PPARγ) antagonist, may improve insulin sensitivity in settings of type 2 diabetes. Here, we determined the effects of GW9662 on the development of NAFLD and molecular mechanisms involved. METHODS Female C57BL/6J mice were pair-fed either a liquid control diet (C) or a fat-, fructose- and cholesterol-rich diet (FFC) for 8 weeks while either being treated with GW9662 (1 mg/kg body weight; C+GW9662 and FFC+GW9662) or vehicle (C and FFC) i.p. three times weekly. Indices of liver damage and inflammation, parameters of glucose metabolism and portal endotoxin levels were determined. Lipopolysaccharide (LPS)-challenged J774A.1 cells were treated with 10 μM GW9662. RESULTS Despite similar caloric intake the development of NAFLD and insulin resistance were significantly attenuated in FFC+GW9662-treated mice when compared to FFC-fed animals. Bacterial endotoxin levels in portal plasma were almost similarly increased in both FFC-fed groups while expressions of toll-like receptor 4 (Tlr4), myeloid differentiation primary response 88 (Myd88) and interleukin 1 beta (Il1b) as well as nitrite (NO2-) concentration in liver were significantly higher in FFC-fed mice than in FFC+GW9662-treated animals. In J774A.1 cells, treatment with GW9662 significantly attenuated LPS-induced expression of Il1b, interleukin 6 (Il6) and inducible nitric oxide synthase (iNos) as well as NO2- formation. CONCLUSION In summary, our data suggest that the PPARγ antagonist GW9662 attenuates the development of a diet-induced NAFLD and that this is associated with a protection against the activation of the TLR4 signaling cascade.
Collapse
Affiliation(s)
- Anja Baumann
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Katharina Burger
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Annette Brandt
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Raphaela Staltner
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Finn Jung
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Dragana Rajcic
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | | | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
Kirk AB, Michelsen-Correa S, Rosen C, Martin CF, Blumberg B. PFAS and Potential Adverse Effects on Bone and Adipose Tissue Through Interactions With PPARγ. Endocrinology 2021; 162:6364127. [PMID: 34480479 PMCID: PMC9034324 DOI: 10.1210/endocr/bqab194] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Indexed: 01/06/2023]
Abstract
Perfluoroalkyl and polyfluoroalkyl substances (PFAS) are a widely dispersed, broad class of synthetic chemicals with diverse biological effects, including effects on adipose and bone differentiation. PFAS most commonly occur as mixtures and only rarely, if ever, as single environmental contaminants. This poses significant regulatory questions and a pronounced need for chemical risk assessments, analytical methods, and technological solutions to reduce the risk to public and environmental health. The effects of PFAS on biological systems may be complex. Each may have several molecular targets initiating multiple biochemical events leading to a number of different adverse outcomes. An exposure to mixtures or coexposures of PFAS complicates the picture further. This review illustrates how PFAS target peroxisome proliferator-activated receptors. Additionally, we describe how such activation leads to changes in cell differentiation and bone development that contributes to metabolic disorder and bone weakness. This discussion sheds light on the importance of seemingly modest outcomes observed in test animals and highlights why the most sensitive end points identified in some chemical risk assessments are significant from a public health perspective.
Collapse
Affiliation(s)
- Andrea B Kirk
- Correspondence: Andrea Kirk, PhD, US EPA Headquarters, William Jefferson Clinton Bldg, 1200 Pennsylvania Ave NW, Mail Code 5201P, Washington, DC 20460, USA.
| | - Stephani Michelsen-Correa
- EPA Office of Chemical Safety and Pollution Prevention, Biopesticides and Pollution Prevention Division, Washington, District of Columbia 20460, USA
| | - Cliff Rosen
- Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | - Bruce Blumberg
- University of California, Irvine, Irvine, California 92697, USA
| |
Collapse
|
9
|
Choi RY, Lee MK. Polygonum multiflorum Thunb. Hot Water Extract Reverses High-Fat Diet-Induced Lipid Metabolism of White and Brown Adipose Tissues in Obese Mice. PLANTS (BASEL, SWITZERLAND) 2021; 10:1509. [PMID: 34451554 PMCID: PMC8398201 DOI: 10.3390/plants10081509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/11/2021] [Accepted: 07/21/2021] [Indexed: 05/06/2023]
Abstract
The purpose of the present study was to determine whether an anti-obesity effect of a Polygonum multiflorum Thunb. hot water extract (PW) was involved in the lipid metabolism of white adipose tissue (WAT) and brown adipose tissue (BAT) in high-fat diet (HFD)-induced C57BL/6N obese mice. Mice freely received a normal diet (NCD) or an HFD for 12 weeks; HFD-fed mice were orally given PW (100 or 300 mg/kg) or garcinia cambogia (GC, 200 mg/kg) once a day. After 12 weeks, PW (300 mg/kg) or GC significantly alleviated adiposity by reducing body weight, WAT weights, and food efficiency ratio. PW (300 mg/kg) improved hyperinsulinemia and enhanced insulin sensitivity. In addition, PW (300 mg/kg) significantly down-regulated expression of carbohydrate-responsive element-binding protein (ChREBP) and diacylglycerol O-acyltransferase 2 (DGAT2) genes in WAT compared with the untreated HFD group. HFD increased BAT gene levels such as adrenoceptor beta 3 (ADRB3), peroxisome proliferator-activated receptor γ (PPARγ), hormone-sensitive lipase (HSL), cluster of differentiation 36 (CD36), fatty acid-binding protein 4 (FABP4), PPARγ coactivator 1-α (PGC-1α), PPARα, and carnitine palmitoyltransferase 1B (CPT1B) compared with the NCD group; however, PW or GC effectively reversed those levels. These findings suggest that the anti-obesity activity of PW was mediated via suppression of lipogenesis in WAT, leading to the normalization of lipid metabolism in BAT.
Collapse
Affiliation(s)
- Ra-Yeong Choi
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Korea;
| | - Mi-Kyung Lee
- Department of Food and Nutrition, Sunchon National University, Suncheon 57922, Korea
| |
Collapse
|
10
|
Gu H, Zhou Y, Yang J, Li J, Peng Y, Zhang X, Miao Y, Jiang W, Bu G, Hou L, Li T, Zhang L, Xia X, Ma Z, Xiong Y, Zuo B. Targeted overexpression of PPARγ in skeletal muscle by random insertion and CRISPR/Cas9 transgenic pig cloning enhances oxidative fiber formation and intramuscular fat deposition. FASEB J 2021; 35:e21308. [PMID: 33481304 DOI: 10.1096/fj.202001812rr] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 11/11/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a master regulator of adipogenesis and lipogenesis. To understand its roles in fiber formation and fat deposition in skeletal muscle, we successfully generated muscle-specific overexpression of PPARγ in two pig models by random insertion and CRISPR/Cas9 transgenic cloning procedures. The content of intramuscular fat was significantly increased in PPARγ pigs while had no changes on lean meat ratio. PPARγ could promote adipocyte differentiation by activating adipocyte differentiating regulators such as FABP4 and CCAAT/enhancer-binding protein (C/EBP), along with enhanced expression of LPL, FABP4, and PLIN1 to proceed fat deposition. Proteomics analyses demonstrated that oxidative metabolism of fatty acids and respiratory chain were activated in PPARγ pigs, thus, gathered more Ca2+ in PPARγ pigs. Raising of Ca2+ could result in increased phosphorylation of CAMKII and p38 MAPK in PPARγ pigs, which can stimulate MEF2 and PGC1α to affect fiber type and oxidative capacity. These results support that skeletal muscle-specific overexpression of PPARγ can promote oxidative fiber formation and intramuscular fat deposition in pigs.
Collapse
Affiliation(s)
- Hao Gu
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Ying Zhou
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Jinzeng Yang
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Jianan Li
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Yaxin Peng
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Xia Zhang
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Yiliang Miao
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Wei Jiang
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Guowei Bu
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Liming Hou
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Ting Li
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Lin Zhang
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Xiaoliang Xia
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Zhiyuan Ma
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Yuanzhu Xiong
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| |
Collapse
|
11
|
Activation of the Peroxisome Proliferator-Activated Receptors (PPAR- α/ γ) and the Fatty Acid Metabolizing Enzyme Protein CPT1A by Camel Milk Treatment Counteracts the High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease. PPAR Res 2021; 2021:5558731. [PMID: 34306045 PMCID: PMC8285205 DOI: 10.1155/2021/5558731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/30/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Camel milk (CM) has a unique composition rich in antioxidants, trace elements, immunoglobulins, insulin, and insulin-like proteins. Treatment by CM demonstrated protective effects against nonalcoholic fatty liver disease (NAFLD) induced by a high-fat cholesterol-rich diet (HFD-C) in rats. CM dampened the steatosis, inflammation, and ballooning degeneration of the hepatocytes. It also counteracted hyperlipidemia, insulin resistance (IR), glucose intolerance, and oxidative stress. The commencement of NAFLD triggered the peroxisome proliferator-activated receptor-α (PPAR-α), carnitine palmitoyl-transferase-1 (CPT1A), and fatty acid-binding protein-1 (FABP1) and decreased the PPAR-γ expression in the tissues of the animals on HFD-C. This was associated with increased levels of the inflammatory cytokines IL-6 and TNF-α and leptin and declined levels of the anti-inflammatory adiponectin. Camel milk treatment to the NAFLD animals remarkably upregulated PPARs (α, γ) and the downstream enzyme CPT1A in the metabolically active tissues involved in cellular uptake and beta-oxidation of fatty acids. The enhanced lipid metabolism in the CM-treated animals was linked with decreased expression of FABP1 and suppression of IL-6, TNF-α, and leptin release with augmented adiponectin production. The protective effects of CM against the histological and biochemical features of NAFLD are at least in part related to the activation of the hepatic and extrahepatic PPARs (α, γ) with consequent activation of the downstream enzymes involved in fat metabolism. Camel milk treatment carries a promising therapeutic potential to NAFLD through stimulating PPARs actions on fat metabolism and glucose homeostasis. This can protect against hepatic steatosis, IR, and diabetes mellitus in high-risk obese patients.
Collapse
|
12
|
MEHP interferes with mitochondrial functions and homeostasis in skeletal muscle cells. Biosci Rep 2021; 40:222590. [PMID: 32255176 PMCID: PMC7167251 DOI: 10.1042/bsr20194404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 11/17/2022] Open
Abstract
Di (2-ethylhexyl) phthalate (DEHP) is a plasticizer frequently leached out from polyvinyl chloride (PVC) products and is quickly metabolized to its monoester equivalent mono(2-ethylhexyl) phthalate (MEHP) once enters organisms. Exposure to DEHP/MEHP through food chain intake has been shown to modified metabolism but its effect on the development of metabolic myopathy of skeletal muscle (SKM) has not been revealed so far. Here, we found that MEHP repressed myogenic terminal differentiation of proliferating myoblasts (PMB) and confluent myoblasts (CMB) but had weak effect on this process once it had been initiated. The transition of mitochondria (MITO) morphology from high efficient filamentary network to low efficient vesicles was triggered by MEHP, implying its negative effects on MITO functions. The impaired MITO functions was further demonstrated by reduced MITO DNA (mtDNA) level and SDH enzyme activity as well as highly increased reactive oxygen species (ROS) in cells after MEHP treatment. The expression of metabolic genes, including PDK4, CPT1b, UCP2, and HO1, was highly increased by MEHP and the promoters of PDK4 and CPT1b were also activated by MEHP. Additionally, the stability of some subunits in the oxidative phosphorylation system (OXPHOS) complexes was found to be reduced by MEHP, implying defective oxidative metabolism in MITO and which was confirmed by repressed palmitic acid oxidation in MEHP-treated cells. Besides, MEHP also blocked insulin-induced glucose uptake. Taken together, our results suggest that MEHP is inhibitory to myogenesis and is harmful to MITO functions in SKM, so its exposure should be avoided or limited.
Collapse
|
13
|
Yang J, Tao D, Ma W, Liu S, Liao Y, Shu L, Zhang S, Li C, Du N, Shi Z. Sijunzi, Lizhong, and Fuzilizhong Decoction Alleviate Nonalcoholic Fatty Liver Disease through Activation of PPAR Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:6363748. [PMID: 33178320 PMCID: PMC7648686 DOI: 10.1155/2020/6363748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/23/2020] [Accepted: 10/16/2020] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Sijunzi, Lizhong, and Fuzilizhong decoction were traditional Chinese classic formulations, which are widely used in clinical treatment, and the underlying mechanism is unclear. In this study, we aim to investigate the molecular mechanisms underlying the protective effects of Sijunzi, Lizhong, and Fuzilizhong on nonalcoholic fatty liver disease (NAFLD). METHODS Male Wistar rats were fed a high-fat diet for four weeks to induce NAFLD and were thereafter administered Sijunzi (8 g/kg/d), Lizhong (10 g/kg/d), or Fuzilizhong (10 g/kg/d) by gavage for four weeks. Hepatic damage, lipid accumulation, inflammation, autophagy, and peroxisome proliferator-activated receptor-α signaling were evaluated. RESULTS The high-fat diet-fed rats showed typical symptoms of NAFLD, including elevated levels of hepatic damage indicators, increased hepatic lipid deposition and fibrosis, severe liver inflammation, and prominent autophagy. Upon administration of Sijunzi, Lizhong, and Fuzilizhong, liver health was improved remarkably, along with ameliorated symptoms of NAFLD. In addition, NAFLD-suppressed peroxisome proliferator-activated receptor-α signaling was reactivated after treatment with the three types of decoctions. CONCLUSIONS The results collectively signify the effective therapeutic and protective functions of Sijunzi, Lizhong, and Fuzilizhong against NAFLD and demonstrate the potential of Chinese herbal medication in mitigating the symptoms of liver diseases. Novelty of the Work. Traditional Chinese herbal medicine has been used for centuries to treat various diseases, but the molecular mechanisms of individual ingredients have rarely been studied. The novelty of our work lies in elucidating the specific signaling pathways involved in the control of NAFLD using three common Chinese herbal decoctions. We suggest that natural herbal formulations can be effective therapeutic agents to combat against NAFLD.
Collapse
Affiliation(s)
- Jiayao Yang
- Department of Gastroenterology, Wuhan Integrated TCM and Western Medicine Hospital, Wuhan, China
| | - Dongqing Tao
- Department of Endocrinology, The Third People's Hospital of Hubei Province, Wuhan, China
| | - Wei Ma
- Department of Center Laboratory, Wuhan Integrated TCM and Western Medicine Hospital, Wuhan, China
| | - Song Liu
- Department of Gastroenterology, Wuhan Integrated TCM and Western Medicine Hospital, Wuhan, China
| | - Yan Liao
- Department of Gastroenterology, Wuhan Integrated TCM and Western Medicine Hospital, Wuhan, China
| | - Lei Shu
- Department of Gastroenterology, Wuhan Integrated TCM and Western Medicine Hospital, Wuhan, China
| | - Shu Zhang
- Department of Gastroenterology, Wuhan Integrated TCM and Western Medicine Hospital, Wuhan, China
| | - Chenyu Li
- Hubei University of Traditional Chinese Medicine, Wuhan, China
| | - Nianlong Du
- Department of Gastroenterology, Wuhan Integrated TCM and Western Medicine Hospital, Wuhan, China
| | - Zhaohong Shi
- Department of Gastroenterology, Wuhan Integrated TCM and Western Medicine Hospital, Wuhan, China
| |
Collapse
|
14
|
Singh R, Chandel S, Dey D, Ghosh A, Roy S, Ravichandiran V, Ghosh D. Epigenetic modification and therapeutic targets of diabetes mellitus. Biosci Rep 2020; 40:BSR20202160. [PMID: 32815547 PMCID: PMC7494983 DOI: 10.1042/bsr20202160] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
The prevalence of diabetes and its related complications are increasing significantly globally. Collected evidence suggested that several genetic and environmental factors contribute to diabetes mellitus. Associated complications such as retinopathy, neuropathy, nephropathy and other cardiovascular complications are a direct result of diabetes. Epigenetic factors include deoxyribonucleic acid (DNA) methylation and histone post-translational modifications. These factors are directly related with pathological factors such as oxidative stress, generation of inflammatory mediators and hyperglycemia. These result in altered gene expression and targets cells in the pathology of diabetes mellitus without specific changes in a DNA sequence. Environmental factors and malnutrition are equally responsible for epigenetic states. Accumulated evidence suggested that environmental stimuli alter the gene expression that result in epigenetic changes in chromatin. Recent studies proposed that epigenetics may include the occurrence of 'metabolic memory' found in animal studies. Further study into epigenetic mechanism might give us new vision into the pathogenesis of diabetes mellitus and related complication thus leading to the discovery of new therapeutic targets. In this review, we discuss the possible epigenetic changes and mechanism that happen in diabetes mellitus type 1 and type 2 separately. We highlight the important epigenetic and non-epigenetic therapeutic targets involved in the management of diabetes and associated complications.
Collapse
Affiliation(s)
- Rajveer Singh
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Shivani Chandel
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Dhritiman Dey
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Arijit Ghosh
- Department of Chemistry, University of Calcutta, Kolkata 700009, India
| | - Syamal Roy
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Velayutham Ravichandiran
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Dipanjan Ghosh
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| |
Collapse
|
15
|
Oleanolic acid induces a dual agonist action on PPARγ/α and GLUT4 translocation: A pentacyclic triterpene for dyslipidemia and type 2 diabetes. Eur J Pharmacol 2020; 883:173252. [DOI: 10.1016/j.ejphar.2020.173252] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/21/2020] [Accepted: 06/05/2020] [Indexed: 12/25/2022]
|
16
|
Xu Y, Curtasu MV, Bach Knudsen KE, Hedemann MS, Theil PK, Lærke HN. Dietary fibre and protein do not synergistically influence insulin, metabolic or inflammatory biomarkers in young obese Göttingen minipigs. Br J Nutr 2020; 125:1-13. [PMID: 32778179 DOI: 10.1017/s0007114520003141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The effects of dietary fibre (DF) and protein on insulin response, lipidaemia and inflammatory biomarkers were studied in a model experiment with juvenile obese Göttingen minipigs. After 20 weeks feeding on a high-fat fructose-rich low-DF diet, forty-three 30-week-old minipigs (31·3 (sem 4·0) kg body weight) were allocated to low- or high-DF and -protein diets for 8 weeks in a 2 × 2 factorial design. High DF contents decreased (P = 0·006) while high protein increased (P < 0·001) the daily gain. High protein contents increased fasting plasma concentrations of glucose (P = 0·008), NEFA (P = 0·015), ghrelin (P = 0·008) and non-fasting LDL:HDL ratios (P = 0·015). High DF increased ghrelin (P = 0·036) and C-peptide levels (P = 0·011) in the non-fasting state. High protein increased the gene expression of fructose-bisphosphatase 1 in liver tissue (P = 0·043), whereas DF decreased fatty acid synthase expression in adipose tissue (P = 0·035). Interactions between DF and protein level were observed in the expression of leptin receptor in adipose tissue (P = 0·031) and of PPARγ in muscle (P = 0·018) and adipose tissue (P = 0·004). In conclusion, high DF intake reduced weight gain and had potential benefit on β-cell secretory function, but without effect on the lipid profile in this young obese model. High dietary protein by supplementing with whey protein did not improve insulin sensitivity or lipidaemia, and combining high DF with high protein did not alleviate the risk of metabolic abnormalities.
Collapse
Affiliation(s)
- Yetong Xu
- Department of Animal Science, Aarhus University, DK-8830Tjele, Denmark
| | | | | | | | | | | |
Collapse
|
17
|
Prashantha Kumar BR, Kumar AP, Jose JA, Prabitha P, Yuvaraj S, Chipurupalli S, Jeyarani V, Manisha C, Banerjee S, Jeyabalan JB, Mohankumar SK, Dhanabal SP, Justin A. Minutes of PPAR-γ agonism and neuroprotection. Neurochem Int 2020; 140:104814. [PMID: 32758586 DOI: 10.1016/j.neuint.2020.104814] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPAR-γ) is one of the ligand-activated transcription factors which regulates a number of central events and considered as a promising target for various neurodegenerative disease conditions. Numerous reports implicate that PPAR-γ agonists have shown neuroprotective effects by regulating genes transcription associated with the pathogenesis of neurodegeneration. In regards, this review critically appraises the recent knowledge of PPAR-γ receptors in neuroprotection in order to hypothesize potential neuroprotective mechanism of PPAR-γ agonism in chronic neurological conditions. Of note, the PPAR-γ's interaction dynamics with PPAR-γ coactivator-1α (PGC-1α) has gained significant attention for neuroprotection. Likewise, a plethora of studies suggest that the PPAR-γ pathway can be actuated by the endogenous ligands present in the CNS and thus identification and development of novel agonist for the PPAR-γ receptor holds a vow to prevent neurodegeneration. Together, the critical insights of this review enlighten the translational possibilities of developing novel neuroprotective therapeutics targeting PPAR-γ for various neurodegenerative disease conditions.
Collapse
Affiliation(s)
- B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Mysuru, Karnataka, India
| | - Ashwini Prem Kumar
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - Jincy A Jose
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - P Prabitha
- Department of Pharmaceutical Chemistry, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Mysuru, Karnataka, India
| | - S Yuvaraj
- Department of Pharmaceutical Chemistry, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Mysuru, Karnataka, India
| | - Sandhya Chipurupalli
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - Victoria Jeyarani
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - Chennu Manisha
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - Sayani Banerjee
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - Jeyaram Bharathi Jeyabalan
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - Suresh Kumar Mohankumar
- TIFAC CORE in HD, Department of Pharmacognosy, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - S P Dhanabal
- TIFAC CORE in HD, Department of Pharmacognosy, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India
| | - Antony Justin
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, Nilgiris, Tamilnadu, India.
| |
Collapse
|
18
|
Stanford BC, Clake DJ, Morris MR, Rogers SM. The power and limitations of gene expression pathway analyses toward predicting population response to environmental stressors. Evol Appl 2020; 13:1166-1182. [PMID: 32684953 PMCID: PMC7359838 DOI: 10.1111/eva.12935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Rapid environmental changes impact the global distribution and abundance of species, highlighting the urgency to understand and predict how populations will respond. The analysis of differentially expressed genes has elucidated areas of the genome involved in adaptive divergence to past and present environmental change. Such studies however have been hampered by large numbers of differentially expressed genes and limited knowledge of how these genes work in conjunction with each other. Recent methods (broadly termed "pathway analyses") have emerged that aim to group genes that behave in a coordinated fashion to a factor of interest. These methods aid in functional annotation and uncovering biological pathways, thereby collapsing complex datasets into more manageable units, providing more nuanced understandings of both the organism-level effects of modified gene expression, and the targets of adaptive divergence. Here, we reanalyze a dataset that investigated temperature-induced changes in gene expression in marine-adapted and freshwater-adapted threespine stickleback (Gasterosteus aculeatus), using Weighted Gene Co-expression Network Analysis (WGCNA) with PANTHER Gene Ontology (GO)-Slim overrepresentation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Six modules exhibited a conserved response and six a divergent response between marine and freshwater stickleback when acclimated to 7°C or 22°C. One divergent module showed freshwater-specific response to temperature, and the remaining divergent modules showed differences in height of reaction norms. PPARAa, a transcription factor that regulates fatty acid metabolism and has been implicated in adaptive divergence, was located in a module that had higher expression at 7°C and in freshwater stickleback. This updated methodology revealed patterns that were not found in the original publication. Although such methods hold promise toward predicting population response to environmental stressors, many limitations remain, particularly with regard to module expression representation, database resources, and cross-database integration.
Collapse
Affiliation(s)
| | - Danielle J. Clake
- Department of Biological SciencesUniversity of CalgaryCalgaryABCanada
| | | | - Sean M. Rogers
- Department of Biological SciencesUniversity of CalgaryCalgaryABCanada
- Bamfield Marine Sciences CentreBamfieldBCCanada
| |
Collapse
|
19
|
Sharma P, Joshi T, Joshi T, Chandra S, Tamta S. In silico screening of potential antidiabetic phytochemicals from Phyllanthus emblica against therapeutic targets of type 2 diabetes. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112268. [PMID: 31593813 DOI: 10.1016/j.jep.2019.112268] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 07/29/2019] [Accepted: 09/30/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phyllanthus emblica Linn. (Syn. Emblica officinalis Gaertn.), has been used to cure many ailments of human beings. Literature survey demonstrates that it has many pharmacological activities i.e. antidiabetic, antioxidant, anti-microbial, antifungal, antiallergic, antiviral, and anticancer properties. AIM OF THE STUDY The present study aimed to identify the novel plant-derived antidiabetic compounds from P. emblica to understand the molecular basis of antidiabetic activities. MATERIAL AND METHODS Text mining analysis of P. emblica and its disease association was carried out using server DLAD4U. Due to the highest score of P. emblica with diabetes, the virtual screening of a phytochemical library of P. emblica against three targets of diabetes was carried out. After that FAF-Drug4, admetSAR and DruLiTo servers were used for drug-likeness prediction. Additionally, pharmacophore modeling was also carried out to understand the antidiabetic activity of screened compounds. RESULTS The docking scores, drug-likeness and pharmacophore studies found that Ellagic acid, Estradiol, Sesamine, Kaempferol, Zeatin, Quercetin, and Leucodelphinidin are potential antidiabetic compounds. CONCLUSIONS Our study shows that phytochemicals of P. emblica are very potential antidiabetic candidates. Using the modern techniques these molecules could be used to develop an effective antidiabetic drugs from a natural resource.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Botany, D.S.B. Campus, Kumaun University, Nainital, 263002, Uttarakhand, India.
| | - Tushar Joshi
- Department of Biotechnology, Bhimtal Campus, Kumaun University, Nainital, 263136, Uttarakhand, India.
| | - Tanuja Joshi
- Department of Botany, S.S.J Campus, Almora, Kumaun University, Nainital, 263601, Uttarakhand, India.
| | - Subhash Chandra
- Department of Botany, S.S.J Campus, Almora, Kumaun University, Nainital, 263601, Uttarakhand, India.
| | - Sushma Tamta
- Department of Botany, D.S.B. Campus, Kumaun University, Nainital, 263002, Uttarakhand, India.
| |
Collapse
|
20
|
de Mendonça M, de Sousa É, da Paixão AO, Araújo Dos Santos B, Roveratti Spagnol A, Murata GM, Araújo HN, Imamura de Lima T, Passos Simões Fróes Guimarães DS, Silveira LR, Rodrigues AC. MicroRNA miR-222 mediates pioglitazone beneficial effects on skeletal muscle of diet-induced obese mice. Mol Cell Endocrinol 2020; 501:110661. [PMID: 31770568 DOI: 10.1016/j.mce.2019.110661] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/19/2019] [Accepted: 11/19/2019] [Indexed: 01/23/2023]
Abstract
Pioglitazone belongs to the class of drugs thiazolidinediones (TZDs) and is an oral hypoglycemic drug, used in the treatment of type 2 diabetes, which improves insulin sensitivity in target tissues. Adipose tissue is the main target of pioglitazone, a PPARg and PPARa agonist; however, studies also point to skeletal muscle as a target. Non-PPAR targets of TZDs have been described, thus we aimed to study the direct effects of pioglitazone on skeletal muscle and the possible role of microRNAs as targets of this drug. Pioglitazone treatment of obese mice increased insulin-mediated glucose transport as a result of increased fatty acid oxidation and mitochondrial activity. PPARg blockage by treatment with GW9662 nullified pioglitazone's effect on systemic and muscle insulin sensitivity and citrate synthase activity of obese mice. After eight weeks of high-fat diet, miR-221-3p expression in soleus muscle was similar among the groups and miR-23b-3p and miR-222-3p were up-regulated in obese mice compared to the control group, and treatment with pioglitazone was able to reverse this condition. In vitro studies in C2C12 cells suggest that inhibition of miR-222-3p protects C2C12 cells from insulin resistance and increased non-mitochondrial respiration induced by palmitate. Together, these data demonstrate a role of pioglitazone in the downregulation of microRNAs that is not dependent on PPARg. Moreover, miR-222 may be a novel PPARg-independent mechanism through which pioglitazone improves insulin sensitivity in skeletal muscle.
Collapse
MESH Headings
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Animals
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Down-Regulation/drug effects
- Glucose/metabolism
- Glucose Tolerance Test
- Hypoglycemic Agents
- Insulin/metabolism
- Insulin Resistance/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Obese
- MicroRNAs/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Obesity/drug therapy
- Obesity/metabolism
- PPAR alpha/metabolism
- PPAR gamma/metabolism
- Palmitates/pharmacology
- Pioglitazone/pharmacology
- Thiazolidinediones/pharmacology
- Up-Regulation/drug effects
Collapse
Affiliation(s)
| | - Érica de Sousa
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Ailma O da Paixão
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, SP, Brazil
| | | | | | - Gilson M Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Hygor N Araújo
- Obesity and Comorbidities Research Center, Campinas, Sao Paulo, Brazil; Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Tanes Imamura de Lima
- Obesity and Comorbidities Research Center, Campinas, Sao Paulo, Brazil; Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Dimitrius Santiago Passos Simões Fróes Guimarães
- Obesity and Comorbidities Research Center, Campinas, Sao Paulo, Brazil; Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center, Campinas, Sao Paulo, Brazil; Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Alice C Rodrigues
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
21
|
Jo YH, Peng DQ, Kim WS, Kim SJ, Kim NY, Kim SH, Ghassemi Nejad J, Lee JS, Lee HG. The effects of vitamin A supplementation during late-stage pregnancy on longissimus dorsi muscle tissue development, birth traits, and growth performance in postnatal Korean native calves. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 33:742-752. [PMID: 32054186 PMCID: PMC7206403 DOI: 10.5713/ajas.19.0413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study investigated the effects of vitamin A (VA) supplementation during late-stage pregnancy on longissimus dorsi muscle tissue development, birth traits, and growth performance of postnatal Korean native calves. METHODS In the preliminary experiment, twenty-six pregnant cattle (initial body weight [BW] = 319 kg (standard deviation [SD] = 30.1; 1st parity) were randomly assigned to the control and treatment groups. The treatment group received VA supplementation at 24,000 IU/d from gestational day 225 until delivery. In the main experiment, twelve pregnant cattle (initial BW = 317 kg [SD = 31.3]; 1st parity) were treated with VA supplementation at 24,000 IU/d (gestational days 150 to 225) and at 78,000 IU/d (gestational day 225 until delivery). Serum VA levels were analyzed in pregnant cattle, and the growth performance, gene expression, and serum VA levels were analyzed in the offspring. RESULTS Serum VA levels in pregnant cattle decreased the late gestation in both experiments (p<0.001). In the main experiment, pregnant cattle at parturition and offspring at birth in the treatment group had higher serum VA levels than those in the control group (p<0.05). In the treatment groups, an increased birth weight was observed in the main experimental group (p = 0.022), and a tendency (p = 0.088) toward an increased birth weight was observed in the preliminary experimental group. However, no differences were observed in the feed intake, average daily gain, gain-to-feed ratio, or BW of 31-day-old calves. Gene expression was analyzed in longissimus dorsi muscles of 31-day-old calves. VA supplementation in pregnant cattle stimulated postnatal muscle development in offspring by elevating myogenic factor 5 (MYF5), MYF6, and myoblast determination levels (p<0.05). Moreover, preadipocyte-related marker genes such as extracellular signal-regulated kinase 2 and krüppel-like factor 2 were higher in the treatment group than in the control group (p<0.05). CONCLUSION VA supplementation (78,000 IU/d) in late-stage pregnant cattle maintained serum VA levels. In addition, 78,000 IU/d VA supplementation increased the birth weight and expression of genes related to muscle and preadipocyte development in offspring. Overall, 78,000 IU/d VA supplementation in pregnant cattle is beneficial to newborn calves.
Collapse
Affiliation(s)
- Yong Ho Jo
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| | - Dong Qiao Peng
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| | - Won Seob Kim
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| | - Seong Jin Kim
- Asia Pacific Ruminant Institute, Icheon 467814, Korea
| | - Na Yeon Kim
- Asia Pacific Ruminant Institute, Icheon 467814, Korea
| | - Sung Hak Kim
- Department of Animal Science, Chonnam National University, Gwangju 61186, Korea
| | - Jalil Ghassemi Nejad
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| | - Jae Sung Lee
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Hong Gu Lee
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
22
|
Ozkan H, Yakan A. Dietary high calories from sunflower oil, sucrose and fructose sources alters lipogenic genes expression levels in liver and skeletal muscle in rats. Ann Hepatol 2019; 18:715-724. [PMID: 31204236 DOI: 10.1016/j.aohep.2019.03.013] [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: 01/22/2019] [Revised: 03/25/2019] [Accepted: 03/19/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES The objectives of this study were to investigate the underlying mechanism of PPARα, LXRα, ChREBP, and SREBP-1c at the level of gene and protein expression with high-energy diets in liver and skeletal muscle. MATERIALS AND METHODS Metabolic changes with consumption of high fat (Hfat), high sucrose (Hsuc) and high fructose (Hfru) diets were assessed. Levels of mRNA and protein of PPARα, LXRα, ChREBP, and SREBP-1c were investigated. Body weight changes, histological structure of liver and plasma levels of some parameters were also examined. RESULTS In Hfru group, body weights were higher than other groups (P<0.05). In liver, LXRα levels of Hsuc and Hfru groups were upregulated as 1.87±0.30 (P<0.05) and 2.01±0.29 (P<0.01). SREBP-1c levels were upregulated as 4.52±1.25 (P<0.05); 4.05±1.11 (P<0.05) and 3.85±1.04 (P<0.05) in Hfat, Hsuc, and Hfru groups, respectively. In skeletal muscle, LXRα and SREBP-1c were upregulated as 1.77±0.30 (P<0.05) and 2.71±0.56 (P<0.05), in the Hfru group. Protein levels of ChREBP (33.92±8.84ng/mg protein (P<0.05)) and SREBP-1c (135.16±15.57ng/mg protein (P<0.001)) in liver were higher in Hfru group. In skeletal muscle, LXRα, ChREBP and SREBP-1c in Hfru group were 6.67±0.60, 7.11±1.29 and 43.17±6.37ng/mg, respectively (P<0.05; P<0.01; P<0.05). The rats in Hfru group had the most damaged livers. CONCLUSION Besides liver, fructose consumption significantly effects skeletal muscle and leads to weight gain, triggers lipogenesis and metabolic disorders.
Collapse
Affiliation(s)
- Huseyin Ozkan
- Department of Genetic, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, Hatay, Turkey.
| | - Akin Yakan
- Department of Animal Breeding, Faculty of Veterinary Medicine, University of Erciyes, Kayseri, Turkey
| |
Collapse
|
23
|
Scalzo RL, Rafferty D, Schauer I, Huebschmann AG, Cree-Green M, Reusch JEB, Regensteiner JG. Sitagliptin improves diastolic cardiac function but not cardiorespiratory fitness in adults with type 2 diabetes. J Diabetes Complications 2019; 33:561-566. [PMID: 31182338 PMCID: PMC7278036 DOI: 10.1016/j.jdiacomp.2019.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 04/17/2019] [Accepted: 05/05/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND People with type 2 diabetes mellitus (T2D) have preclinical cardiac and vascular dysfunction associated with low cardiorespiratory fitness (CRF). This is especially concerning because CRF is a powerful predictor of cardiovascular mortality, a primary issue in T2D management. Glucagon-like pepetide-1 (GLP-1) augments cardiovascular function and our previous data in rodents demonstrate that potentiating the GLP-1 signal with a dipeptidyl peptidase-4 (DPP4) inhibitor augments CRF. Lacking are pharmacological treatments which can target T2D-specific physiological barriers to exercise to potentially permit adaptations necessary to improve CRF and thereby health outcomes in people with T2D. We therefore hypothesized that administration of a DPP4-inhibitor (sitagliptin) would improve CRF in adults with T2D. METHODS AND RESULTS Thirty-eight participants (64 ± 1 years; mean ± SE) with T2D were randomized in a double-blinded study to receive 100 mg/day sitagliptin, 2 mg/day glimepiride, or placebo for 3 months after baseline measurements. Fasting glucose decreased with both glimepiride and sitagliptin compared with placebo (P = 0.002). CRF did not change in any group (Placebo: Pre: 15.4 ± 0.9 vs. Post: 16.1 ± 1.1 ml/kg/min vs. Glimepiride: 18.5 ± 1.0 vs. 17.7 ± 1.2 ml/kg/min vs. Sitagliptin: 19.1 ± 1.2 vs. 18.3 ± 1.1 ml/kg/min; P = 0.3). Sitagliptin improved measures of cardiac diastolic function, however, measures of vascular function did not change with any treatment. CONCLUSIONS Three months of sitagliptin improved diastolic cardiac function, however, CRF did not change. These data suggest that targeting the physiological contributors to CRF with sitagliptin alone is not an adequate strategy to improve CRF in people with T2D. CLINICAL TRIALS REGISTRATION www.clinicaltrials.gov NCT01951339.
Collapse
Affiliation(s)
- Rebecca L Scalzo
- Division of Endocrinology, Department of Medicine, University of Colorado School of Medicine, United States of America; Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, United States of America; Rocky Mountain Regional Veterans Administration Medical Center, United States of America.
| | - Deirdre Rafferty
- Division of General Internal Medicine, Department of Medicine, University of Colorado School of Medicine, United States of America
| | - Irene Schauer
- Division of Endocrinology, Department of Medicine, University of Colorado School of Medicine, United States of America; Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, United States of America; Rocky Mountain Regional Veterans Administration Medical Center, United States of America
| | - Amy G Huebschmann
- Division of General Internal Medicine, Department of Medicine, University of Colorado School of Medicine, United States of America; Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, United States of America
| | - Melanie Cree-Green
- Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, United States of America; Division of Pediatric Endocrinology, University of Colorado School of Medicine, United States of America
| | - Jane E B Reusch
- Division of Endocrinology, Department of Medicine, University of Colorado School of Medicine, United States of America; Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, United States of America; Rocky Mountain Regional Veterans Administration Medical Center, United States of America
| | - Judith G Regensteiner
- Division of General Internal Medicine, Department of Medicine, University of Colorado School of Medicine, United States of America; Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, United States of America
| |
Collapse
|
24
|
Cao Y, Zhang Q, Chen J, Li Z, Zhou Z, Shen J, Wang D, Pan D, Wang Z, Ke D, Wang X, Lu D, Zhao Y, Cheng S, Shi Y. Polymorphism of the PPARD Gene and Dynamic Balance Performance in Han Chinese Children. Hereditas 2019; 156:15. [PMID: 31148953 PMCID: PMC6533762 DOI: 10.1186/s41065-019-0092-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/15/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Athletic performances are complex traits with heritability of ~66%. Dynamic balance is one of the most important athletic performances, and there has been little studies for it in sports genomics. The candidate PPARD gene was reported to be able to affect muscle development for balance predisposition and influence the athletic performance including skiing triumph in the Caucasian population. This study aims to investigate whether the PPARD gene is a susceptibility gene for dynamic balance performance in Han Chinese children. RESULTS A total 2244 children were recruited and their balance beam performances were measured. Five polymorphisms in the PPARD gene were genotyped through the MassARRAY Sequenom platform. Rs2016520 exerted significant association with dynamic balance performance (minor allele C, P = 0.015, Pcorrected < 0.05) and was affirmed in a meta-analysis by combining previously reported Caucasian cohorts (OR = 1.57, 95% CI = [1.30, 1.91], P < 10 -5) . Another polymorphism, rs2267668, was also significantly associated with dynamic balance performance (minor allele G, P = 0.015, Pcorrected < 0.05). In the dichotomous study, 321 cases (61% boys and 39% girls) and 370 controls (49% boys and 51% girls) in our samples were selected as representatives, and the thresholds were the mean velocity (0.737 m/s) ± standard deviation (0.264 m/s), in which rs2016520-C and rs2267668-G still remained significant (CI =1.41 [1.11~1.79], P = 0.004, Pcorrected < 0.016; CI =1.45 [1.14~1.86], P = 0.002, Pcorrected < 0.016). In different genders, consistent OR direction was observed for each variant. CONCLUSIONS Our results suggested that the PPARD gene is associated with dynamic balance performance of human being, and further studies to reveal its etiology is strongly suggested.
Collapse
Affiliation(s)
- Yixuan Cao
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Qiyue Zhang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Jianhua Chen
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030 China
| | - Zhiqiang Li
- Qingdao University, Metabolic Disease Institute, Qingdao, 266003 China
| | - Zhaowei Zhou
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Jiawei Shen
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Dong Wang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Dun Pan
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Zhuo Wang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Dandan Ke
- Department of Human Sports Science, Shanghai University of Sport, Shanghai, 200438 China
| | - Xiaofei Wang
- Department of Human Sports Science, Shanghai University of Sport, Shanghai, 200438 China
| | - Dajiang Lu
- Department of Human Sports Science, Shanghai University of Sport, Shanghai, 200438 China
| | - Ying Zhao
- Physical Education Department, Shanghai Jiao Tong University, Shanghai, 200240 China
| | - Shulin Cheng
- Physical Education Department, Shanghai Jiao Tong University, Shanghai, 200240 China
| | - Yongyong Shi
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030 China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| |
Collapse
|
25
|
Hare KS, Wood KM, Fitzsimmons C, Penner GB. Oversupplying metabolizable protein in late gestation for beef cattle: effects on postpartum ruminal fermentation, blood metabolites, skeletal muscle catabolism, colostrum composition, milk yield and composition, and calf growth performance. J Anim Sci 2019; 97:437-455. [PMID: 30371794 DOI: 10.1093/jas/sky413] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/25/2018] [Indexed: 12/15/2022] Open
Abstract
The objective of the study was to determine whether oversupplying MP prepartum affects postpartum cow BW, colostrum composition, milk production and composition, protein catabolism in the dam, and calf growth. Crossbred Hereford heifers were individually fed a control treatment designed to meet MP requirements (CON; n = 10) or 133% of the MP requirement (HMP; n = 11) from day -55 ± 4 until parturition. All cows were provided a common postpartum diet. Cow BW was measured on days 7 ± 1, 14 ± 2, 28 ± 3, 57 ± 4, 82 ± 5, and 111 ± 3 relative to parturition. DMI and ruminal pH were measured daily and summarized by week until day 33. Milk yield was estimated based on a 12-h two-quarter milk yield on days 7 ± 1, 12 ± 1, 28 ± 3, 33 ± 3, 70 ± 3, and 112 ± 3. Urine samples were collected from cows over a 6-d period starting on days 7 ± 1 and 28 ± 3 and the composited samples were analyzed for 3-methylhistidine (3-MH) and creatinine. Muscle samples were collected from cows on day 13 ± 1 while calf muscle samples were collected on days 2 and 111 ± 3 of age. Muscle samples from cows were analyzed for markers of protein catabolism, and calf muscle samples were analyzed for genes regulating cell growth and differentiation. Data were analyzed as a randomized complete block design using the MIXED procedure of SAS accounting for repeated measures when necessary. Postpartum BW did not differ (P ≥ 0.30) by treatment, day, or the interaction of treatment and day (T × D), but rump fat decreased (P = 0.011) as lactation progressed. DMI decreased during weeks 2 and 3 compared to 1 and 4, whereas ruminal pH was less during weeks 2, 3, and 4 relative to week 1. Colostrum fat concentration was less (P = 0.003) for HMP than CON; but, milk production was not affected by treatment. Milk yield was greatest from days 7 to 33 and decreased thereafter (P < 0.01). Urinary 3-MH and the 3-MH:creatinine ratio did not differ by treatment, day, or the T × D (P ≥ 0.22) interaction, nor was there a difference (P ≥ 0.13) in the abundance of catabolic proteins. Calf growth was not affected by treatment, but HMP calves had greater expression (T × D, P = 0.05) of PPARG while PKM expression increased for CON calves (T × D, P = 0.04) at day 111 compared to their expression at day 2. Overfeeding MP during late gestation does not improve postpartum indicators of N balance or maternal muscle turnover but may alter colostrum composition and calf gene expression at weaning.
Collapse
Affiliation(s)
- Koryn S Hare
- Department of Animal and Poultry Science, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, SK, Canada
| | - Katie M Wood
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
| | - Carolyn Fitzsimmons
- Agriculture and Agri-Food Canada, Edmonton, AB, Canada.,$Department of Agriculture, Food and Nutritional Sciences, Faculty of Agricultural Life and Environmental Sciences, University of Alberta, Edmonton, AB, Canada
| | - Gregory B Penner
- Department of Animal and Poultry Science, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
26
|
Understanding Peroxisome Proliferator-Activated Receptors: From the Structure to the Regulatory Actions on Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1127:39-57. [DOI: 10.1007/978-3-030-11488-6_3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
27
|
Kim J, Wellmann KB, Smith ZK, Johnson BJ. All-trans retinoic acid increases the expression of oxidative myosin heavy chain through the PPARδ pathway in bovine muscle cells derived from satellite cells. J Anim Sci 2018; 96:2763-2776. [PMID: 29688535 DOI: 10.1093/jas/sky155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/19/2018] [Indexed: 11/15/2022] Open
Abstract
All-trans retinoic acid (ATRA) has been associated with various physiological phenomenon in mammalian adipose tissue and skeletal muscle. We hypothesized that ATRA may affect skeletal muscle fiber type in bovine satellite cell culture through various transcriptional processes. Bovine primary satellite cell (BSC) culture experiments were conducted to determine dose effects of ATRA on expression of genes and protein levels related to skeletal muscle fiber type and metabolism. The semimembranosus from crossbred steers (n = 2 steers), aged approximately 24 mo, were used to isolate BSC for 3 separate assays. Myogenic differentiation was induced using 3% horse serum upon cultured BSC with increasing doses (0, 1, 10, 100, and 1,000 nM) of ATRA. After 96 h of incubation, cells were harvested and used to measure the gene expression of protein kinase B (Akt), AMP-activated protein kinase alpha (AMPK), glucose transporter 4 (GLUT4), myogenin, lipoprotein lipase (LPL), myosin heavy chain (MHC) I, MHC IIA, MHC IIX, insulin like growth factor-1 (IGF-1), Peroxisome proliferator activated receptor gamma (PPARγ), PPARδ, and Smad transcription factor 3 (SMAD3) mRNA relative to ribosomal protein subunit 9 (RPS9). The mRNA expression of LPL was increased (P < 0.05) with 100 and 1,000 nM of ATRA. Expression of GLUT4 was altered (P < 0.05) by ATRA. The treatment of ATRA (1,000 nM) also increased (P < 0.05) mRNA gene expression of SMAD3. The gene expression of both PPARδ and PPARγ were increased (P < 0.05) with 1,000 nM of ATRA. Protein level of PPARδ was also affected (P < 0.05) by 1,000 nM of ATRA and resulted in a greater (P < 0.05) protein level of PPARδ compared to CON. All-trans retinoic acid (10 nM) increased gene expression of MHC I (P < 0.05) compared to CON. Expression of MHC IIA was also influenced (P < 0.05) by ATRA. The mRNA expression of MHC IIX was decreased (P < 0.05) with 100 and 1,000 nM of ATRA. In muscle cells, ATRA may cause muscle fibers to transition towards the MHC isoform that prefers oxidative metabolism, as evidenced by increased expression of genes associated with the MHC I isoform. These changes in MHC isoforms appeared to be brought about by changing PPARδ gene expression and protein levels.
Collapse
Affiliation(s)
- Jongkyoo Kim
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| | | | - Zachary K Smith
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| | - Bradley J Johnson
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| |
Collapse
|
28
|
Takahashi H, Sanada K, Nagai H, Li Y, Aoki Y, Ara T, Seno S, Matsuda H, Yu R, Kawada T, Goto T. Over-expression of PPARα in obese mice adipose tissue improves insulin sensitivity. Biochem Biophys Res Commun 2017; 493:108-114. [PMID: 28919422 DOI: 10.1016/j.bbrc.2017.09.067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 09/13/2017] [Indexed: 01/22/2023]
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is important in the regulation of lipid metabolism and expressed at high levels in the liver. Although PPARα is also expressed in adipose tissue, little is known about the relationship between its activation and the regulation of glucose metabolism. In this study, we developed adipose tissue specific PPARα over-expression (OE) mice. Metabolomics and insulin tolerance tests showed that OE induces branched-chain amino acid (BCAA) profile and improvement of insulin sensitivity. Furthermore, LC-MS and PCR analyses revealed that OE changes free fatty acid (FFA) profile and reduces obesity-induced inflammation. These findings suggested that PPARα activation in adipose tissue contributes to the improvement of glucose metabolism disorders via the enhancement of BCAA and FFA metabolism.
Collapse
Affiliation(s)
- Haruya Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Kohei Sanada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Hiroyuki Nagai
- Gifu Prefectural Research Institute for Health and Environmental Science, Gifu, Japan
| | - Yongjia Li
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Yumeko Aoki
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Takeshi Ara
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Shigeto Seno
- Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
| | - Hideo Matsuda
- Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, South Korea
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan; Research Unit for Physiological Chemistry, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan; Research Unit for Physiological Chemistry, Kyoto University, Kyoto, Japan.
| |
Collapse
|
29
|
Smith ZK, Chung KY, Parr SL, Johnson BJ. Anabolic payout of terminal implant alters adipogenic gene expression of the longissimus muscle in beef steers. J Anim Sci 2017; 95:1197-1204. [PMID: 28380538 DOI: 10.2527/jas.2016.0630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This experiment evaluated the dose and payout pattern of trenbolone acetate (TBA) and estradiol-17β (E) on LM mRNA expression of adenosine monophosphate-activated protein kinase-ɑ (-ɑ), β, G protein-coupled receptor 41(), G protein-coupled receptor 43 (), γ, and stearoyl CoA desaturase () in finishing feedlot steers as indicators of adipogenesis and marbling development. British × Continental steers (n = 168; 14 pens/treatment; initial BW = 362 kg) were used in a randomized complete block design. Treatments included: no implant (NI), Revalor-S (REV-S; 120 mg TBA + 24 mg E), or Revalor-XS (REV-X; delayed release implant: 80 mg TBA + 16 mg E [uncoated], 120 mg TBA + 24 mg E [coated], 200 mg TBA + 40 mg E [total]). Steers were fed 1 time daily for an average of 164 d. The LM biopsies were collected (1 steer/pen) on d -1, 27, 55, and 111 relative to timing of implant. Total RNA was isolated from each sample and real-time quantitative PCR was used to measure quantity of -ɑ, β, , ,it, γ, and mRNA. No implant × day interactions were detected ( ≥ 0.19) in this experiment. Day impacted the mRNA expression of all adipogenic genes ( ≤ 0.02). The main effect of implant tended ( = 0.09) to influence expression of -ɑ, REV-X had an 8.8% increase over NI and an 18.7% increase over REV-S. Implant influenced ( = 0.03) mRNA expression of , expression of for the REV-X treatment was not different ( > 0.10) from NI, and both were greater ( ≤ 0.05) than REV-S (1.13, 1.00, and 0.67 ± 0.224 arbitrary units) for REV-X, NI, and REV-S, respectively. Implant also influenced ( = 0.02) expression of , expression of for REV-X was not different ( > 0.10) from NI, and both were greater ( ≤ 0.05) than REV-S (1.27, 1.07, and 0.72 ± 0.234 arbitrary units) for REV-X, NI, and REV-S, respectively. Implant influenced ( = 0.02) mRNA expression of γ in LM tissue, expression of γ for REV-X was not different ( > 0.10) from NI, and both were greater ( ≤ 0.05) than REV-S (1.09, 1.02, and 0.69 ± 0.195 arbitrary units) for REV-X, NI, and REV-S, respectively. The REV-X steers received the greatest anabolic dose of TBA + E without detriment to marbling scores. The increased mRNA expression of adipogenic genes for REV-X steers suggest that the delayed and gradual release of anabolic stimulants associated with REV-X might have mitigated decreases in marbling generally attributed to multiple combined TBA + E implants.
Collapse
|
30
|
Abstract
Obesity is a worldwide epidemic that predisposes individuals to cardiometabolic complications, such as type 2 diabetes mellitus (T2DM) and nonalcoholic fatty liver disease (NAFLD), which are all related to inappropriate ectopic lipid deposition. Identification of the pathogenic molecular mechanisms and effective therapeutic approaches are highly needed. The peroxisome proliferator-activated receptors (PPARs) modulate several biological processes that are perturbed in obesity, including inflammation, lipid and glucose metabolism and overall energy homeostasis. Here, we review how PPARs regulate the functions of adipose tissues, such as adipogenesis, lipid storage and adaptive thermogenesis, under healthy and pathological conditions. We also discuss the clinical use and mechanism of PPAR agonists in the treatment of obesity comorbidities such as dyslipidaemia, T2DM and NAFLD. First generation PPAR agonists, primarily those acting on PPARγ, are associated with adverse effects that outweigh their clinical benefits, which led to the discontinuation of their development. An improved understanding of the physiological roles of PPARs might, therefore, enable the development of safe, new PPAR agonists with improved therapeutic potential.
Collapse
Affiliation(s)
- Barbara Gross
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Michal Pawlak
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, 02-109 Warsaw, Poland
| | - Philippe Lefebvre
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Bart Staels
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| |
Collapse
|
31
|
Nicoletti CF, Pinhel MAS, de Oliveira BAP, Marchini JS, Salgado Junior W, Silva Junior WA, Nonino CB. The Genetic Predisposition Score of Seven Obesity-Related Single Nucleotide Polymorphisms Is Associated with Better Metabolic Outcomes after Roux-en-Y Gastric Bypass. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2016; 9:222-230. [PMID: 27806373 DOI: 10.1159/000452129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 09/29/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Genetic variants associated with obesity have cumulative effects on obesity risk and related phenotypes. This study aimed to estimate the contribution of a genetic predisposition score (GPS) calculated from 7 obesity-related polymorphisms to the improvement of biochemical parameters 1 year after Roux-en-Y gastric bypass (RYGB). METHODS Obese patients (n = 150; aged 47.2 ± 10.5 years) were enrolled and weight, body mass index (BMI), and biochemical parameters (glycemia and lipid profile) were evaluated preoperatively and 1 year after RYGB. A GPS was calculated with the polymorphisms rs1801282 of PPARG2, rs4994 of ADRB3, rs1800592 of UCP1, rs659366 and rs669339 of UCP2, rs7121 of GNAS1, and rs5443 of GNB3. We observed that 66.3% of the patients has a GPS >5. RESULTS During the preoperative period, the GPS showed a significant association with weight (β = -0.163; p = 0.020), BMI (β = -0.169; p = 0.038), and glucose concentrations (β = -0.177; p = 0.036). After sex and age adjustment, a higher GPS was associated with a greater reduction in glycemia (β = -0.158; p = 0.048), triglycerides (β = -0.256; p = 0.002), and total cholesterol (β = -0.172; p = 0.038) concentrations 1 year after surgery. CONCLUSION Our data elucidated that a higher GPS provides a greater metabolic benefit of RYGB.
Collapse
|
32
|
Tokach RJ, Ribeiro FRB, Chung KY, Rounds W, Johnson BJ. Chromium Propionate Enhances Adipogenic Differentiation of Bovine Intramuscular Adipocytes. Front Vet Sci 2015; 2:26. [PMID: 26664955 PMCID: PMC4672234 DOI: 10.3389/fvets.2015.00026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/17/2015] [Indexed: 01/24/2023] Open
Abstract
In vitro experiments were performed to determine the effects of increasing concentrations of chromium propionate (CrPro) on mRNA and protein abundance of different enzymes and receptors. Intramuscular (IM) and subcutaneous (SC) preadipocytes and bovine satellite cells were isolated from the longissimus muscle to determine the effect of treatment on glucose transporter type 4 (GLUT4) and peroxisome proliferator-activated receptor γ mRNA and GLUT4 protein abundance. Preadipocyte cultures were treated with differentiation media plus either sodium propionate or different concentrations of CrPro for 96, 120, and 144 h before harvest. This study indicated that adipogenesis of the bovine IM adipocytes were more sensitive to the treatment of CrPro as compared to SC adipocytes. Enhancement of adenosine monophosphate-activated protein kinase and GLUT4 mRNA by CrPro treatment may enhance glucose uptake in IM adipocytes. CrPro decreased GLUT4 protein levels in muscle cell cultures suggesting that those cells have increased efficiency of glucose uptake due to exposure to increased levels of CrPro. In contrast, each of the two adipogenic lines had opposing responses to the CrPro. It appeared that CrPro had the most stimulative effect of GLUT4 response in the IM adipocytes as compared to SC adipocytes. These findings indicated opportunities to potentially augment marbling in beef cattle fed CrPro during the finishing phase.
Collapse
Affiliation(s)
- Rebecca J. Tokach
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX, USA
| | | | - Ki Yong Chung
- Hanwoo Research Institute, National Institute of Animal Science, Rural Development Administration, Kangwon, South Korea
| | - Whitney Rounds
- Kemin Animal Nutrition and Health North America, Des Moines, IA, USA
| | - Bradley J. Johnson
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
33
|
MacNeil LG, Glover E, Bergstra TG, Safdar A, Tarnopolsky MA. The order of exercise during concurrent training for rehabilitation does not alter acute genetic expression, mitochondrial enzyme activity or improvements in muscle function. PLoS One 2014; 9:e109189. [PMID: 25289940 PMCID: PMC4188604 DOI: 10.1371/journal.pone.0109189] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 09/08/2014] [Indexed: 01/09/2023] Open
Abstract
Concurrent exercise combines different modes of exercise (e.g., aerobic and resistance) into one training protocol, providing stimuli meant to increase muscle strength, aerobic capacity and mass. As disuse is associated with decrements in strength, aerobic capacity and muscle size concurrent training is an attractive modality for rehabilitation. However, interference between the signaling pathways may result in preferential improvements for one of the exercise modes. We recruited 18 young adults (10 ♂, 8 ♀) to determine if order of exercise mode during concurrent training would differentially affect gene expression, protein content and measures of strength and aerobic capacity after 2 weeks of knee-brace induced disuse. Concurrent exercise sessions were performed 3x/week for 6 weeks at gradually increasing intensities either with endurance exercise preceding (END>RES) or following (RES>END) resistance exercise. Biopsies were collected from the vastus lateralis before, 3 h after the first exercise bout and 48 h after the end of training. Concurrent exercise altered the expression of genes involved in mitochondrial biogenesis (PGC-1α, PRC, PPARγ), hypertrophy (PGC-1α4, REDD2, Rheb) and atrophy (MuRF-1, Runx1), increased electron transport chain complex protein content, citrate synthase and mitochondrial cytochrome c oxidase enzyme activity, muscle mass, maximum isometric strength and VO2peak. However, the order in which exercise was completed (END>RES or RES>END) only affected the protein content of mitochondrial complex II subunit. In conclusion, concurrent exercise training is an effective modality for the rehabilitation of the loss of skeletal muscle mass, maximum strength, and peak aerobic capacity resulting from disuse, regardless of the order in which the modes of exercise are performed.
Collapse
Affiliation(s)
- Lauren G. MacNeil
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Elisa Glover
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - T. Graham Bergstra
- Department of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Adeel Safdar
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Mark A. Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
34
|
Liu HW, Tsai YT, Chang SJ. Toona sinensis leaf extract inhibits lipid accumulation through up-regulation of genes involved in lipolysis and fatty acid oxidation in adipocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:5887-5896. [PMID: 24884355 DOI: 10.1021/jf500714c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Toona sinensis leaf (TSL) has been shown to lower plasma triacylglycerol levels and diminish the size of visceral fat cells in vivo. The molecular mechanism of TSL ethanol extract (TSL-E) on lipid metabolism in 3T3-L1 adipocytes was investigated in this study. Oil Red O staining as well as immunoblotting, real-time PCR, and dual-Luciferase reporter system were performed to investigate the effect of TSL-E on lipid accumulation and the regulation of lipid metabolism, respectively. In addition, active compounds in the TSL-E were analyzed by HPLC. TSL-E significantly decreased lipid accumulation, stimulated free fatty acid (FFA) release, and up-regulated peroxisome proliferator-activated receptor-α (PPARα) and genes involved in peroxisomal (acyl-CoA oxidase) and mitochondrial (uncouple protein 3) fatty acid oxidation. TSL-E also up-regulated cytoplasmic triacylglycerol hydrolysis gene (adipose triglyceride lipase) and genes related to fatty acid oxidation (AMP-activated protein kinase, acetyl-CoA carboxylase, carnitine palmitoyltransferase I, PPARγ, and adiponectin). The major constituents directly inducing PPARα transactivity in TSL-E are gallic acid, rutin, palmitic acid, linoleic acid, and α-linolenic acid. These results indicate that the inhibitory effect of TSL-E on lipid accumulation was through PPARα activation and further up-regulation of PPARα-mediated genes plus up-regulation of cytoplasmic genes involved in lipid catabolism.
Collapse
Affiliation(s)
- Hung-Wen Liu
- Department of Life Sciences, National Cheng Kung University , Tainan, Taiwan
| | | | | |
Collapse
|
35
|
Black RNA, Ennis CN, Young IS, Hunter SJ, Atkinson AB, Bell PM. The peroxisome proliferator-activated receptor alpha agonist fenofibrate has no effect on insulin sensitivity compared to atorvastatin in type 2 diabetes mellitus; a randomised, double-blind controlled trial. J Diabetes Complications 2014; 28:323-7. [PMID: 24560135 DOI: 10.1016/j.jdiacomp.2014.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/02/2013] [Accepted: 01/02/2014] [Indexed: 11/28/2022]
Abstract
AIMS Assess insulin sensitivity after treatment with a selective PPAR-alpha agonist compared to an HMG CoA reductase inhibitor in human subjects with type 2 diabetes mellitus. METHODS Thirteen subjects with Type 2 diabetes mellitus were studied in a double-blind crossover design with 4-week placebo run-in and washout and 12-week treatment periods, randomised to micronised fenofibrate 267 mg or atorvastatin 10mg daily followed by the alternate drug in the second period. Insulin resistance was measured using the isoglycaemic hyperinsulinaemic clamp method with isotope dilution. RESULTS Weight, physical activity and other medications did not change. Total cholesterol (mean +/- standard error) was 4.60+/-0.21 versus 3.9+/-0.22 mmol/L after fenofibrate and atorvastatin respectively, p<0.05. LDL was 2.70+/-0.19 versus 1.95+/-0.23 mmol/L, p<0.05 and triglyceride 1.64+/-0.23 versus 1.84+/-0.26 mmol/L, p<0.05. Insulin-stimulated whole-body glucose disposal (35.4+/-3.1 versus 33.2+/-3.0 μmol/kg/min) and nadir endogenous glucose production (6.2+/-1.4 versus 7.0+/-1.1 μmol/kg/min) revealed no significant differences in effects of the treatments. CONCLUSIONS In human subjects with Type 2 diabetes mellitus there were characteristic differences in lipid profile changes but no difference in insulin sensitivity after treatment with micronised fenofibrate compared to atorvastatin. This study finds no evidence of increased insulin sensitivity using this selective PPAR-alpha agonist over a commonly used statin at these doses.
Collapse
Affiliation(s)
- R Neil A Black
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK.
| | - Cieran N Ennis
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| | - Ian S Young
- Nutrition and Metabolism Group, The Queen's University of Belfast, Belfast, UK
| | - Steven J Hunter
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| | - A Brew Atkinson
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| | - Patrick M Bell
- Regional Centre for Endocrinology & Diabetes, Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| |
Collapse
|
36
|
The formation of brown adipose tissue induced by transgenic over-expression of PPARγ2. Biochem Biophys Res Commun 2014; 446:959-64. [PMID: 24642257 DOI: 10.1016/j.bbrc.2014.03.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 03/10/2014] [Indexed: 11/22/2022]
Abstract
Brown adipose tissue (BAT) is specialized to dissipate energy as heat, therefore reducing fat deposition and counteracting obesity. Brown adipocytes arise from myoblastic progenitors during embryonic development by the action of transcription regulator PRDM16 binding to PPARγ, which promotes BAT-like phenotype in white adipose tissue. To investigate the capability of converting white adipose tissue to BAT or browning by PPARγ in vivo, we generated transgenic mice with over-expressed PPARγ2. The transgenic mice showed strong brown fat features in subcutaneous fat in morphology and histology. To provide molecular evidences on browning characteristics of the adipose tissue, we employed quantitative real-time PCR to determine BAT-specific gene expressions. The transgenic mice had remarkably elevated mRNA level of UCP1, Elovl3, PGC1α and Cebpα in subcutaneous fat. Compared with wild-type mice, UCP1 protein levels were increased significantly in transgenic mice. ATP concentration was slightly decreased in the subcutaneous fat of transgenic mice. Western blotting analysis also confirmed that phosphorylated AMPK and ACC proteins were significantly (P<0.01) increased in the transgenic mice. Therefore, this study demonstrated that over-expression of PPARγ2 in skeletal muscle can promote conversion of subcutaneous fat to brown fat formation, which can have beneficial effects on increasing energy metabolisms and combating obesity.
Collapse
|
37
|
Yin SN, Liu M, Jing DQ, Mu YM, Lu JM, Pan CY. Telmisartan increases lipoprotein lipase expression via peroxisome proliferator-activated receptor-alpha in HepG2 cells. Endocr Res 2014; 39:66-72. [PMID: 24067162 DOI: 10.3109/07435800.2013.828741] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In addition to their hypotensive properties, angiotensin receptor blockers (ARBs) have been shown to exert clinical antidyslipidemic effects. The mechanism underlying these ARB lipid metabolic effects remains unclear. Some ARBs, for example, telmisartan, activate peroxisome proliferator-activated activated receptor-gamma (PPAR-gamma). We hypothesized that PPAR-gamma-activating ARBs might exert antidyslipidemic effects via PPAR-alpha. In this study, we assessed the effect of telmisartan on the expression of PPAR-alpha and lipoprotein lipase (LPL). PPAR-alpha expression was detected by reverse-transcription polymerase chain reaction and Western blot in HepG2 hepatocytes as well as differentiated C2C12 myocytes treated with increasing concentrations of telmisartan (0.1-10 μmol/L) for 48 h. Results showed that 1 μmol/L and 10 μmol/L telmisartan significantly increased the expression of PPAR-alpha mRNA and protein in HepG2 cells (p < 0.01). No effect was shown in differentiated C2C12 cells. Similarly, 1 µmol/L and 10 μmol/L telmisartan significantly increased the expression of LPL mRNA and protein in HepG2 cells (p < 0.01), and this increase was significantly (p < 0.01) inhibited by the PPAR-alpha-specific antagonist MK886. These results indicate that certain of the antidyslipidemic effects of telmisartan might be mediated via increased PPAR-alpha-dependent induction of LPL expression.
Collapse
Affiliation(s)
- Shi Nan Yin
- State Key Laboratory of Endocrine and Metabolic Diseases, Chinese PLA General Hospital , Beijing , China
| | | | | | | | | | | |
Collapse
|
38
|
Metabolic disturbance in PCOS: clinical and molecular effects on skeletal muscle tissue. ScientificWorldJournal 2013; 2013:178364. [PMID: 23844380 PMCID: PMC3687487 DOI: 10.1155/2013/178364] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 02/04/2013] [Indexed: 12/21/2022] Open
Abstract
Polycystic ovary syndrome is a complex hormonal disorder affecting the reproductive and metabolic systems with signs and symptoms related to anovulation, infertility, menstrual irregularity and hirsutism.
Skeletal muscle plays a vital role in the peripheral glucose uptake. Since PCOS is associated with defects in the activation and pancreatic dysfunction of β-cell insulin, it is important to understand the molecular mechanisms of insulin resistance in PCOS. Studies of muscle tissue in patients with PCOS reveal defects in insulin signaling. Muscle biopsies performed during euglycemic hyperinsulinemic clamp showed a significant reduction in glucose uptake, and insulin-mediated IRS-2 increased significantly in skeletal muscle. It is recognized that the etiology of insulin resistance in PCOS is likely to be as complicated as in type 2 diabetes and it has an important role in metabolic and reproductive phenotypes of this syndrome. Thus, further evidence regarding the effect of nonpharmacological approaches (e.g., physical exercise) in skeletal muscle of women with PCOS is required for a better therapeutic approach in the management of various metabolic and reproductive problems caused by this syndrome.
Collapse
|
39
|
Conn CA, Vaughan RA, Garver WS. Nutritional Genetics and Energy Metabolism in Human Obesity. Curr Nutr Rep 2013. [DOI: 10.1007/s13668-013-0046-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
40
|
Effect of Opuntia humifusa supplementation and acute exercise on insulin sensitivity and associations with PPAR-γ and PGC-1α protein expression in skeletal muscle of rats. Int J Mol Sci 2013; 14:7140-54. [PMID: 23538842 PMCID: PMC3645680 DOI: 10.3390/ijms14047140] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/21/2013] [Accepted: 03/25/2013] [Indexed: 12/15/2022] Open
Abstract
This study examined whether Opuntia humifusa (O. humifusa), which is a member of the Cactaceae family, supplementation and acute swimming exercise affect insulin sensitivity and associations with PPAR-γ and PGC-1α protein expression in rats. Thirty-two rats were randomly divided into four groups (HS: high fat diet sedentary group, n = 8; HE: high fat diet acute exercise group, n = 8; OS: 5% O. humifusa supplemented high fat diet sedentary group, n = 8; OE: 5% O. humifusa supplemented high fat diet acute exercise group, n = 8). Rats in the HE and OE swam for 120 min. before being sacrificed. Our results indicated that serum glucose level, fasting insulin level and homeostasis model assessment of insulin resistance (HOMA-IR) in OS were significantly lower compared to those of the HS (p < 0.01, p < 0.05, p < 0.05). In addition, PPAR-γ protein expression in the OS and OE was significantly higher than that of the HS and HE, respectively (p < 0.05, p < 0.01). PGC-1α and GLUT-4 protein expressions in the OS were significantly higher compared to those of the HS (p < 0.05, p < 0.05). From these results, O. humifusa supplementation might play an important role for improving insulin sensitivity through elevation of PPAR-γ, PGC-1α, and GLUT-4 protein expression in rat skeletal muscle.
Collapse
|
41
|
Sangeetha MK, Priya CDM, Vasanthi HR. Anti-diabetic property of Tinospora cordifolia and its active compound is mediated through the expression of Glut-4 in L6 myotubes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:246-248. [PMID: 23290487 DOI: 10.1016/j.phymed.2012.11.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/01/2012] [Accepted: 11/11/2012] [Indexed: 06/01/2023]
Abstract
Tinospora cordifolia is a well reported plant possessing numerous medicinal values including anti-diabetic property. Aim of the present study is to study the mechanism of action of Tinospora cordifolia and its active compound in differentiated myocytes, L6 cells. Key marker of diabetes in cells is the insulin dependent glucose transporter-4 (Glut-4) which also responds to exogenous chemicals, and is over expressed up to 5- and 4-fold, by Tinospora cordifolia and palmatine, respectively. Next to Glut-4, the predominant protein influencing glucose metabolism is PPARα and γ whose expressions were also positively modulated. Further, the inhibitors of insulin pathway prevented glucose uptake mediated by Tinospora cordifolia and palmatine which shows that the activity is majorly mediated through insulin pathway.
Collapse
Affiliation(s)
- M K Sangeetha
- Herbal and Indian Medicine Research Laboratory, Department of Biochemistry, Sri Ramachandra University, Chennai, India
| | | | | |
Collapse
|
42
|
Cornall LM, Mathai ML, Hryciw DH, Simcocks AC, O'Brien PE, Wentworth JM, McAinch AJ. GPR119 regulates genetic markers of fatty acid oxidation in cultured skeletal muscle myotubes. Mol Cell Endocrinol 2013; 365:108-18. [PMID: 23069642 DOI: 10.1016/j.mce.2012.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 10/01/2012] [Accepted: 10/03/2012] [Indexed: 12/16/2022]
Abstract
Gene knockout and agonist studies indicate that activation of the G protein-coupled receptor, GPR119, protects against diet-induced obesity and insulin resistance. It is not known if GPR119 activation in skeletal muscle mediates these effects. To address this uncertainty, we measured GPR119 expression in skeletal muscle and determined the effects of PSN632408, a GPR119 agonist, on the expression of genes and proteins required for fatty acid and glucose oxidation in cultured myotubes. GPR119 expression was readily detected in rat skeletal muscle and mRNAs were induced by 12 weeks of high-fat feeding. Treatment of cultured mouse C₂C₁₂ myotubes with 5 μM PSN632408 or 0.5 mM palmitate reduced expression of mRNAs encoding fatty acid oxidation genes to similar extents. More so, treatment with PSN632408 decreased AMPKα (Thr172 phosphorylation) activity in the absence of palmitate and ACC (Ser79 phosphorylation) activity in the presence of palmitate. In human primary myotubes PSN632408 decreased expression of PDK4 and AMPKα2 mRNA in myotubes derived from obese donors. These data suggest GPR119 activation in skeletal muscle may impair fatty acid and glucose oxidation.
Collapse
MESH Headings
- Acids, Heterocyclic/pharmacology
- Adult
- Animals
- Body Mass Index
- Cells, Cultured
- Clone Cells
- Fatty Acids, Nonesterified/metabolism
- Female
- Gene Expression Regulation/drug effects
- Genetic Markers
- Glucose/metabolism
- Humans
- Male
- Mice
- Middle Aged
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Obesity, Morbid/genetics
- Obesity, Morbid/metabolism
- Obesity, Morbid/pathology
- Oxadiazoles/pharmacology
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
Collapse
Affiliation(s)
- L M Cornall
- Biomedical and Lifestyle Diseases Unit, School of Biomedical and Health Sciences, Victoria University, Melbourne 8001, Australia.
| | | | | | | | | | | | | |
Collapse
|
43
|
Wang H, Wu H, Rocuts F, Gu Z, Bach FH, Otterbein LE. Activation of Peroxisome Proliferator-Activated Receptor γ Prolongs Islet Allograft Survival. Cell Transplant 2012; 21:2111-8. [DOI: 10.3727/096368911x637399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Exposing donor mice to carbon monoxide (CO) protects transplanted islet allografts from immune rejection after transplantation (referred as the “donor” effect). In an attempt to understand the mechanisms of the donor effect of CO, we found that donor treatment with CO upregulates expression of peroxisome proliferatoractivated receptor γ (PPARγ), a transcriptional regulator, in isolated islets. In this study, we evaluated whether PPARγ contributes to the survival and function of transplanted islets and whether PPARγ mediates the protective effect of CO in a major mismatch islet allogeneic transplantation model. BALB/c (H-2d) islets in which PPARγ activity was induced by its agonists, 15-deoxy-Δ12–14-prostaglandin J2 (15d-PGJ2) or troglitazone were transplanted into C57BL/6 (H-2b) recipients that had been rendered diabetic by streptozotocin (STZ). Blood glucose levels of recipients were monitored to determine the function of transplanted islets. Our data indicated that PPARγ activation in islets led to a high percentage of BALB/c islets survived long-term in C57BL/6 recipients. Activation of PPARγ in the donor suppresses expressions of proinflammatory cytokines including tumor necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS) in transplanted islets. Blocking PPARγ activity by its antagonist, GW9662, abrogated the donor effect of CO in vivo and in vitro. Our data demonstrate that PPARγ plays a critical role in the survival and function of transplanted islets after transplantation in the recipient. The protective effects of CO are at least in part mediated by PPARγ.
Collapse
Affiliation(s)
- Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hongju Wu
- Department of Obstetrics and Gynecology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Fredy Rocuts
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zhuoying Gu
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Fritz H. Bach
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Leo E. Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Rikimaru K, Wakabayashi T, Abe H, Tawaraishi T, Imoto H, Yonemori J, Hirose H, Murase K, Matsuo T, Matsumoto M, Nomura C, Tsuge H, Arimura N, Kawakami K, Sakamoto J, Funami M, Mol CD, Snell GP, Bragstad KA, Sang BC, Dougan DR, Tanaka T, Katayama N, Horiguchi Y, Momose Y. Structure–activity relationships and key structural feature of pyridyloxybenzene-acylsulfonamides as new, potent, and selective peroxisome proliferator-activated receptor (PPAR) γ Agonists. Bioorg Med Chem 2012; 20:3332-58. [DOI: 10.1016/j.bmc.2012.03.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Revised: 03/13/2012] [Accepted: 03/14/2012] [Indexed: 10/28/2022]
|
45
|
Hu S, Yao J, Howe AA, Menke BM, Sivitz WI, Spector AA, Norris AW. Peroxisome proliferator-activated receptor γ decouples fatty acid uptake from lipid inhibition of insulin signaling in skeletal muscle. Mol Endocrinol 2012; 26:977-88. [PMID: 22474127 DOI: 10.1210/me.2011-1253] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is expressed at low levels in skeletal muscle, where it protects against adiposity and insulin resistance via unclear mechanisms. To test the hypothesis that PPARγ directly modulates skeletal muscle metabolism, we created two models that isolate direct PPARγ actions on skeletal myocytes. PPARγ was overexpressed in murine myotubes by adenotransfection and in mouse skeletal muscle by plasmid electroporation. In cultured myotubes, PPARγ action increased fatty acid uptake and incorporation into myocellular lipids, dependent upon a 154 ± 20-fold up-regulation of CD36 expression. PPARγ overexpression more than doubled insulin-stimulated thymoma viral proto-oncogene (AKT) phosphorylation during low lipid availability. Furthermore, in myotubes exposed to palmitate levels that inhibit insulin signaling, PPARγ overexpression increased insulin-stimulated AKT phosphorylation and glycogen synthesis over 3-fold despite simultaneously increasing myocellular palmitate uptake. The insulin signaling enhancement was associated with an increase in activating phosphorylation of phosphoinositide-dependent protein kinase 1 and a normalized expression of palmitate-induced genes that antagonize AKT phosphorylation. In vivo, PPARγ overexpression more than doubled insulin-dependent AKT phosphorylation in lipid-treated mice but did not augment insulin-stimulated glucose uptake. We conclude that direct PPARγ action promotes myocellular storage of energy by increasing fatty acid uptake and esterification while simultaneously enhancing insulin signaling and glycogen formation. However, direct PPARγ action in skeletal muscle is not sufficient to account for the hypoglycemic actions of PPARγ agonists during lipotoxicity.
Collapse
Affiliation(s)
- Shanming Hu
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Haddad PS, Musallam L, Martineau LC, Harris C, Lavoie L, Arnason JT, Foster B, Bennett S, Johns T, Cuerrier A, Coon Come E, Coon Come R, Diamond J, Etapp L, Etapp C, George J, Husky Swallow C, Husky Swallow J, Jolly M, Kawapit A, Mamianskum E, Petagumskum J, Petawabano S, Petawabano L, Weistche A, Badawi A. Comprehensive evidence-based assessment and prioritization of potential antidiabetic medicinal plants: a case study from canadian eastern james bay cree traditional medicine. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2011; 2012:893426. [PMID: 22235232 PMCID: PMC3247006 DOI: 10.1155/2012/893426] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 09/09/2011] [Indexed: 01/15/2023]
Abstract
Canadian Aboriginals, like others globally, suffer from disproportionately high rates of diabetes. A comprehensive evidence-based approach was therefore developed to study potential antidiabetic medicinal plants stemming from Canadian Aboriginal Traditional Medicine to provide culturally adapted complementary and alternative treatment options. Key elements of pathophysiology of diabetes and of related contemporary drug therapy are presented to highlight relevant cellular and molecular targets for medicinal plants. Potential antidiabetic plants were identified using a novel ethnobotanical method based on a set of diabetes symptoms. The most promising species were screened for primary (glucose-lowering) and secondary (toxicity, drug interactions, complications) antidiabetic activity by using a comprehensive platform of in vitro cell-based and cell-free bioassays. The most active species were studied further for their mechanism of action and their active principles identified though bioassay-guided fractionation. Biological activity of key species was confirmed in animal models of diabetes. These in vitro and in vivo findings are the basis for evidence-based prioritization of antidiabetic plants. In parallel, plants were also prioritized by Cree Elders and healers according to their Traditional Medicine paradigm. This case study highlights the convergence of modern science and Traditional Medicine while providing a model that can be adapted to other Aboriginal realities worldwide.
Collapse
Affiliation(s)
- Pierre S. Haddad
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
- Department of Pharmacology, Université de Montréal and Montreal Diabetes Research Center, P.O. Box 6128, Downtown Postal Station, Montreal, QC, Canada H3C 3J7
| | - Lina Musallam
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
- Department of Pharmacology, Université de Montréal and Montreal Diabetes Research Center, P.O. Box 6128, Downtown Postal Station, Montreal, QC, Canada H3C 3J7
| | - Louis C. Martineau
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
| | - Cory Harris
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
- School of Dietetics and Human Nutrition and Center for Indigenous Peoples' Nutrition and Environment, McGill University, Sainte-Anne-de-Bellevue, QC, Canada H9X 3V9
| | - Louis Lavoie
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
| | - John T. Arnason
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
- Department of Biology, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | - Brian Foster
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
- Department of Cellular and Molecular Medicine, University of Ottawa and Therapeutic Products Directorate, Health Canada, Ottawa, ON, Canada K1A 1B6
| | - Steffany Bennett
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada K1H 8M5
| | - Timothy Johns
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
- School of Dietetics and Human Nutrition and Center for Indigenous Peoples' Nutrition and Environment, McGill University, Sainte-Anne-de-Bellevue, QC, Canada H9X 3V9
| | - Alain Cuerrier
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines, Montreal, QC, Canada H3C 317
- Plant Biology Research Institute, Université de Montréal and Montreal Botanical Garden, Montreal, QC, Canada H1X 2B2
| | - Emma Coon Come
- Cree Nation of Mistissini, Eeyou Istchii, QC, Canada GOW 1CO
| | - Rene Coon Come
- Cree Nation of Mistissini, Eeyou Istchii, QC, Canada GOW 1CO
| | - Josephine Diamond
- The Crees of Waskaganish First Nation, Eeyou Istchii, QC, Canada JON 1RO
| | - Louise Etapp
- Cree Nation of Mistissini, Eeyou Istchii, QC, Canada GOW 1CO
| | - Charlie Etapp
- Cree Nation of Mistissini, Eeyou Istchii, QC, Canada GOW 1CO
| | - Jimmy George
- Whapmagoostui First Nation, Eeyou Istchii, QC, Canada JOM 1GO
| | | | | | - Mary Jolly
- Cree Nation of Nemaska, Nemaska, QC, Canada JLY 3BO
| | - Andrew Kawapit
- Whapmagoostui First Nation, Eeyou Istchii, QC, Canada JOM 1GO
| | | | | | | | | | - Alex Weistche
- The Crees of Waskaganish First Nation, Eeyou Istchii, QC, Canada JON 1RO
| | - Alaa Badawi
- Office of Biotechnology, Genomics, and Population Health, Public Health Agency of Canada, Toronto, ON, Canada M5V 3L7
| |
Collapse
|
47
|
Duchesne E, Tremblay MH, Côté CH. Mast cell tryptase stimulates myoblast proliferation; a mechanism relying on protease-activated receptor-2 and cyclooxygenase-2. BMC Musculoskelet Disord 2011; 12:235. [PMID: 21999702 PMCID: PMC3207928 DOI: 10.1186/1471-2474-12-235] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 10/14/2011] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Mast cells contribute to tissue repair in fibrous tissues by stimulating proliferation of fibroblasts through the release of tryptase which activates protease-activated receptor-2 (PAR-2). The possibility that a tryptase/PAR-2 signaling pathway exists in skeletal muscle cell has never been investigated. The aim of this study was to evaluate whether tryptase can stimulate myoblast proliferation and determine the downstream cascade. METHODS Proliferation of L6 rat skeletal myoblasts stimulated with PAR-2 agonists (tryptase, trypsin and SLIGKV) was assessed. The specificity of the tryptase effect was evaluated with a specific inhibitor, APC-366. Western blot analyses were used to evaluate the expression and functionality of PAR-2 receptor and to assess the expression of COX-2. COX-2 activity was evaluated with a commercial activity assay kit and by measurement of PGF2α production. Proliferation assays were also performed in presence of different prostaglandins (PGs). RESULTS Tryptase increased L6 myoblast proliferation by 35% above control group and this effect was completely inhibited by APC-366. We confirmed the expression of PAR-2 receptor in vivo in skeletal muscle cells and in satellite cells and in vitro in L6 cells, where PAR-2 was found to be functional. Trypsin and SLIGKV increased L6 cells proliferation by 76% and 26% above control, respectively. COX-2 activity was increased following stimulation with PAR-2 agonist but its expression remained unchanged. Inhibition of COX-2 activity by NS-398 abolished the stimulation of cell proliferation induced by tryptase and trypsin. Finally, 15-deoxy-Δ-12,14-prostaglandin J2 (15Δ-PGJ2), a product of COX-2-derived prostaglandin D2, stimulated myoblast proliferation, but not PGE2 and PGF2α. CONCLUSIONS Taken together, our data show that tryptase can stimulate myoblast proliferation and this effect is part of a signaling cascade dependent on PAR-2 activation and on the downstream activation of COX-2.
Collapse
Affiliation(s)
- Elise Duchesne
- CHUQ Research Center and Faculty of Medicine, Laval University, 2705 boul, Laurier, Québec, Québec G1V 4G2, Canada
| | | | | |
Collapse
|
48
|
Perreault L, Bergman BC, Hunerdosse DM, Howard DJ, Eckel RH. Fenofibrate administration does not affect muscle triglyceride concentration or insulin sensitivity in humans. Metabolism 2011; 60:1107-14. [PMID: 21306746 PMCID: PMC3135750 DOI: 10.1016/j.metabol.2010.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 11/24/2010] [Accepted: 12/09/2010] [Indexed: 11/21/2022]
Abstract
Animal data suggest that males, in particular, rely on peroxisome proliferator activated receptor-α activity to maintain normal muscle triglyceride metabolism. We sought to examine whether this was also true in men vs women and its relationship to insulin sensitivity (Si). Normolipidemic obese men (n = 9) and women (n = 9) underwent an assessment of Si (intravenous glucose tolerance test) and intramuscular triglyceride (IMTG) metabolism (gas chromatography/mass spectrometry and gas chromatography-combustion isotope ratio mass spectrometry from plasma and muscle biopsies taken after infusion of [U-(13)C]palmitate) before and after 12 weeks of fenofibrate treatment. Women were more insulin sensitive (Si: 5.2 ± 0.7 vs 2.4 ± 0.4 ×10(-4)/ μU/mL, W vs M, P < .01) at baseline despite similar IMTG concentration (41.9 ± 15.5 vs 30.8 ± 5.1 μg/mg dry weight, W vs M, P = .43) and IMTG fractional synthesis rate (FSR) (0.27%/h ± 0.07%/h vs 0.35%/h ± 0.06%/h, W vs M, P = .41) as men. Fenofibrate enhanced FSR in men (0.35 ± 0.06 to 0.54 ± 0.06, P = .05), with no such change seen in women (0.27 ± 0.07 to 0.32 ± 0.13, P = .73) and no change in IMTG concentration in either group (23.0 ± 3.9 in M, P = .26 vs baseline; 36.3 ± 12.0 in W, P = .79 vs baseline). Insulin sensitivity was unaffected by fenofibrate (P ≥ .68). Lower percentage saturation of IMTG in women vs men before (29.1% ± 2.3% vs 35.2% ± 1.7%, P = .06) and after (27.3% ± 2.8% vs 35.1% ± 1.9%, P = .04) fenofibrate most closely related to their greater Si (R(2) = 0.34, P = .10) and was largely unchanged by the drug. Peroxisome proliferator activated receptor-α agonist therapy had little effect on IMTG metabolism in men or women. Intramuscular triglyceride saturation, rather than IMTG concentration or FSR, most closely (but not significantly) related to Si and was unchanged by fenofibrate administration.
Collapse
Affiliation(s)
- Leigh Perreault
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver School of Medicine, Aurora, CO 80045, USA.
| | | | | | | | | |
Collapse
|
49
|
Mynarcik DC, McNurlan MA, Melendez MM, Vosswinkel JA, Gelato MC. Rosiglitazone-Mediated Effects on Skeletal Muscle Gene Expression Correlate with Improvements in Insulin Sensitivity in Individuals with HIV-Insulin Resistance. PATHOLOGY RESEARCH INTERNATIONAL 2011; 2011:736425. [PMID: 21559208 PMCID: PMC3090220 DOI: 10.4061/2011/736425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 02/21/2011] [Indexed: 11/20/2022]
Abstract
Rosiglitazone, an agonist of peroxisome proliferator activated receptor (PPARγ), improves insulin sensitivity by increasing insulin-stimulated glucose uptake into muscle tissue. This study was undertaken to assess changes in expression of PPAR-regulated genes in muscle tissue following treatment of HIV-associated insulin resistance with rosiglitazone. Muscle gene expression was assessed in twenty-two seronegative HIV subjects (control), 21 HIV-infected individuals with normal insulin sensitivity (HIV-IS) and 19 HIV-infected individuals with insulin resistance (HIV-IR). A subset of the HIV-IR group (N=10) were re-evaluated 12 weeks after treatment with 8 mg/d of rosiglitazone. The HIV-IR group's rosiglitazone-mediated improvement in insulin sensitivity was highly correlated with increased expression of PPARγ and carnitine palmitoyl transferase-1 (CPT-1), (r=0.87, P<.001) and (r=0.95, P<.001), respectively. The changes in PPARγ expression were also correlated with the changes in CPT1 expression (r=0.75, P=.009). The results suggest that rosiglitazone; may have a direct effect on muscle tissue to improve insulin sensitivity.
Collapse
Affiliation(s)
- Dennis C. Mynarcik
- Division of Endocrinology, Department of Medicine, Stony Brook University Medical Center, HSC T15-060, Stony Brook, NY 11794-8154, USA
| | - Margaret A. McNurlan
- Department of Surgery, Stony Brook University Medical Center, Stony Brook, NY 11794-8154, USA
| | - Mark M. Melendez
- Department of Surgery, Stony Brook University Medical Center, Stony Brook, NY 11794-8154, USA
| | - James A. Vosswinkel
- Department of Surgery, Stony Brook University Medical Center, Stony Brook, NY 11794-8154, USA
| | - Marie C. Gelato
- Division of Endocrinology, Department of Medicine, Stony Brook University Medical Center, HSC T15-060, Stony Brook, NY 11794-8154, USA
| |
Collapse
|
50
|
Goto T, Lee JY, Teraminami A, Kim YI, Hirai S, Uemura T, Inoue H, Takahashi N, Kawada T. Activation of peroxisome proliferator-activated receptor-alpha stimulates both differentiation and fatty acid oxidation in adipocytes. J Lipid Res 2011; 52:873-84. [PMID: 21324916 PMCID: PMC3073464 DOI: 10.1194/jlr.m011320] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Peroxisome proliferator-activated receptor-α (PPARα) is a dietary lipid sensor, whose activation results in hypolipidemic effects. In this study, we investigated whether PPARα activation affects energy metabolism in white adipose tissue (WAT). Activation of PPARα by its agonist (bezafibrate) markedly reduced adiposity in KK mice fed a high-fat diet. In 3T3-L1 adipocytes, addition of GW7647, a highly specific PPARα agonist, during adipocyte differentiation enhanced glycerol-3-phosphate dehydrogenase activity, insulin-stimulated glucose uptake, and adipogenic gene expression. However, triglyceride accumulation was not increased by PPARα activation. PPARα activation induced expression of target genes involved in FA oxidation and stimulated FA oxidation. In WAT of KK mice treated with bezafibrate, both adipogenic and FA oxidation-related genes were significantly upregulated. These changes in mRNA expression were not observed in PPARα-deficient mice. Bezafibrate treatment enhanced FA oxidation in isolated adipocytes, suppressing adipocyte hypertrophy. Chromatin immunoprecipitation (ChIP) assay revealed that PPARα was recruited to promoter regions of both adipogenic and FA oxidation-related genes in the presence of GW7647 in 3T3-L1 adipocytes. These findings indicate that the activation of PPARα affects energy metabolism in adipocytes, and PPARα activation in WAT may contribute to the clinical effects of fibrate drugs.
Collapse
Affiliation(s)
- Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|