51
|
Clevenger TN, Hinman CR, Ashley Rubin RK, Smither K, Burke DJ, Hawker CJ, Messina D, Van Epps D, Clegg DO. Vitronectin-Based, Biomimetic Encapsulating Hydrogel Scaffolds Support Adipogenesis of Adipose Stem Cells. Tissue Eng Part A 2016; 22:597-609. [PMID: 26956095 DOI: 10.1089/ten.tea.2015.0550] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Soft tissue defects are relatively common, yet currently used reconstructive treatments have varying success rates, and serious potential complications such as unpredictable volume loss and reabsorption. Human adipose-derived stem cells (ASCs), isolated from liposuction aspirate have great potential for use in soft tissue regeneration, especially when combined with a supportive scaffold. To design scaffolds that promote differentiation of these cells down an adipogenic lineage, we characterized changes in the surrounding extracellular environment during adipogenic differentiation. We found expression changes in both extracellular matrix proteins, including increases in expression of collagen-IV and vitronectin, as well as changes in the integrin expression profile, with an increase in expression of integrins such as αVβ5 and α1β1. These integrins are known to specifically interact with vitronectin and collagen-IV, respectively, through binding to an Arg-Gly-Asp (RGD) sequence. When three different short RGD-containing peptides were incorporated into three-dimensional (3D) hydrogel cultures, it was found that an RGD-containing peptide derived from vitronectin provided strong initial attachment, maintained the desired morphology, and created optimal conditions for in vitro 3D adipogenic differentiation of ASCs. These results describe a simple, nontoxic encapsulating scaffold, capable of supporting the survival and desired differentiation of ASCs for the treatment of soft tissue defects.
Collapse
Affiliation(s)
- Tracy N Clevenger
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| | - Cassidy R Hinman
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California
| | - Rebekah K Ashley Rubin
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| | | | - Daniel J Burke
- 4 Materials Research Laboratory, University of California , Santa Barbara
| | - Craig J Hawker
- 4 Materials Research Laboratory, University of California , Santa Barbara
| | | | | | - Dennis O Clegg
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| |
Collapse
|
52
|
Korach-André M, Gustafsson JÅ. Liver X receptors as regulators of metabolism. Biomol Concepts 2016; 6:177-90. [PMID: 25945723 DOI: 10.1515/bmc-2015-0007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/01/2015] [Indexed: 11/15/2022] Open
Abstract
The liver X receptors (LXR) are crucial regulators of metabolism. After ligand binding, they regulate gene transcription and thereby mediate changes in metabolic pathways. Modulation of LXR and their downstream targets has appeared to be a promising treatment for metabolic diseases especially atherosclerosis and cholesterol metabolism. However, the complexity of LXR action in various metabolic tissues and the liver side effect of LXR activation have slowed down the interest for LXR drugs. In this review, we summarized the role of LXR in the main metabolically active tissues with a special focus on obesity and associated diseases in mammals. We will also discuss the dual interplay between the two LXR isoforms suggesting that they may collaborate to establish a fine and efficient system for the maintenance of metabolism homeostasis.
Collapse
|
53
|
Mysore R, Zhou Y, Sädevirta S, Savolainen-Peltonen H, Nidhina Haridas PA, Soronen J, Leivonen M, Sarin AP, Fischer-Posovszky P, Wabitsch M, Yki-Järvinen H, Olkkonen VM. MicroRNA-192* impairs adipocyte triglyceride storage. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1861:342-51. [PMID: 26747651 DOI: 10.1016/j.bbalip.2015.12.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 02/06/2023]
Abstract
We investigated the expression of miR-192* (miR-192-3p) in the visceral adipose tissue (VAT) of obese subjects and its function in cultured human adipocytes. This miRNA is a 3' arm derived from the same pre-miRNA as miR-192 (miR-192-5p) implicated in type 2 diabetes, liver disease and cancers, and is predicted to target key genes in lipid metabolism. In morbidly obese subjects undergoing bariatric surgery preceded by a very low calorie diet, miR-192* in VAT correlated negatively (r=-0.387; p=0.046) with serum triglyceride (TG) and positively with high-density lipoprotein (HDL) concentration (r=0.396; p=0.041). In a less obese patient cohort, the miRNA correlated negatively with the body mass index (r=-0.537; p=0.026). To characterize the function of miR-192*, we overexpressed it in cultured adipocytes and analyzed the expression of adipogenic differentiation markers as well as cellular TG content. Reduced TG and expression of the adipocyte marker proteins aP2 (adipocyte protein 2) and perilipin 1 were observed. The function of miR-192* was further investigated by transcriptomic profiling of adipocytes expressing this miRNA, revealing impacts on key lipogenic genes. A number of the mRNA alterations were validated by qPCR. Western analysis confirmed a marked reduction of the lipogenic enzyme SCD (stearoyl coenzyme A desaturase-1), the fatty aldehyde dehydrogenase ALDH3A2 (aldehyde dehydrogenase 3 family member A2) and the high-density lipoprotein receptor SCARB1 (scavenger receptor B, type I). SCD and ALDH3A2 were demonstrated to be direct targets of miR-192*. To conclude, the present data identify miR-192* as a novel controller of adipocyte differentiation and lipid homeostasis.
Collapse
Affiliation(s)
- Raghavendra Mysore
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland
| | - You Zhou
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland
| | - Sanja Sädevirta
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland; Department of Medicine, University of Helsinki, FI-00014 Finland
| | - Hanna Savolainen-Peltonen
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, FI-00029 HUS, Helsinki, Finland; Folkhälsan Research Center, FI-00290 Helsinki, Finland
| | - P A Nidhina Haridas
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland
| | - Jarkko Soronen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland; National Institute for Health and Welfare/Public Health Genomics Unit, Biomedicum, FI-00290 Helsinki, Finland
| | - Marja Leivonen
- Department of Surgery, Helsinki University Central Hospital, FI-000290 HUS, Helsinki, Finland
| | - Antti-Pekka Sarin
- National Institute for Health and Welfare/Public Health Genomics Unit, Biomedicum, FI-00290 Helsinki, Finland; Institute for Molecular Medicine Finland FIMM, University of Helsinki, FI-00014 Helsinki, Finland
| | - Pamela Fischer-Posovszky
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, D-89075 Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, D-89075 Ulm, Germany
| | - Hannele Yki-Järvinen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland; Department of Medicine, University of Helsinki, FI-00014 Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, FI-00290 Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|
54
|
Li X, Yang M, Li Z, Xue M, Shangguan Z, Ou Z, Liu M, Liu S, Yang S, Li X. Fructus xanthii improves lipid homeostasis in the epididymal adipose tissue of rats fed a high-fat diet. Mol Med Rep 2015; 13:787-95. [PMID: 26648271 PMCID: PMC4686102 DOI: 10.3892/mmr.2015.4628] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 11/06/2015] [Indexed: 12/15/2022] Open
Abstract
High fat diet (HFD)-induced obesity triggers common features of human metabolic syndrome in rats. Our previous study showed that Fructus xanthii (FX) attenuates HFD-induced hepatic steatosis. The present study was designed to investigate the effects of FX on lipid metabolism in epididymal fat (EF), and examine its underlying mechanisms. Aqueous extraction fractions of FX or vehicle were orally administered by gavage for 6 weeks to rats fed either a HFD or a normal chow diet (NCD). The levels of circulating free fatty acid (FFA) were determined in plasma, and the expression levels of lipid metabolism- and inflammation-associated genes in the EF were measured using reverse transcription-quantitative polymerase chain reaction analysis. The general morphology, size and number of adipocytes in the EF, and the levels of macrophage infiltration were evaluated using hematoxylin and eosin staining or immunohistochemical staining. FX decreased circulating levels of FFA, increased the expression levels of sterol-regulatory-element-binding protein-1c, FAS, acetyl coenzyme A carboxylase, diacylglycerol acyltransferase and lipoprotein lipase lipogenic genes in the EF. FX increased the numbers of adipocytes in the EF, and featured a shift towards smaller adipocyte size. Compared with the vehicle-treated rats, positive staining of F4/80 was more dispersed in the FX-treated rats, and the percentage of F4/80 positive cells was significantly decreased. FX attenuated HFD-induced lipid dyshomeostasis in the epididymal adipose tissue.
Collapse
Affiliation(s)
- Xiumin Li
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Mingxing Yang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhipeng Li
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Mei Xue
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhaoshui Shangguan
- Central Laboratory, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhimin Ou
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Ming Liu
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Suhuan Liu
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Shuyu Yang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Xuejun Li
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| |
Collapse
|
55
|
Tian Y, Tao L, Zhao S, Tai D, Liu D, Liu P. Isolation and morphological characterization of ovine amniotic fluid mesenchymal stem cells. Exp Anim 2015; 65:125-34. [PMID: 26616638 PMCID: PMC4873481 DOI: 10.1538/expanim.15-0031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are one of the most promising cell populations for tissue engineering and regenerative medicine. Of utmost importance to MSC research is identification of MSC sources that are easily obtainable and stable. Several studies have shown that MSCs can be isolated from amniotic fluid. The sheep is one of the main types of farm animal, and it has many biophysical and biochemical similarities to humans. Here, we obtained MSCs from ovine amniotic fluid and determined the expansion capacity, surface and intracellular marker expression, karyotype, and multilineage differentiation ability of these ovine amniotic fluid mesenchymal stem cells (oAF-MSCs). Moreover, expression levels of differentiation markers were measured using reverse transcription-qPCR (RT-qPCR). Our phenotypic analysis shows that the isolated oAF-MSCs are indeed MSCs.
Collapse
Affiliation(s)
- Yunyun Tian
- College of Life Sciences, Inner Mongolia University, Inner Mongolia, Hohhot 010021, P.R.China
| | | | | | | | | | | |
Collapse
|
56
|
Paredes JF, López-Olmeda JF, Martínez FJ, Sánchez-Vázquez FJ. Daily rhythms of lipid metabolic gene expression in zebra fish liver: Response to light/dark and feeding cycles. Chronobiol Int 2015; 32:1438-48. [PMID: 26595085 DOI: 10.3109/07420528.2015.1104327] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Despite numerous studies about fish nutrition and lipid metabolism, very little is known about the daily rhythm expression of lipogenesis and lipolysis genes. This research aimed to investigate the existence of daily rhythm expressions of the genes involved in lipid metabolism and their synchronization to different light/dark (LD) and feeding cycles in zebra fish liver. For this purpose, three groups of zebra fish were submitted to a 12:12 h LD cycle. A single daily meal was provided to each group at various times: in the middle of the light phase (ML); in the middle of the dark phase (MD); at random times. After 20 days of acclimation to these experimental conditions, liver samples were collected every 4 h in one 24-h cycle. The results revealed that most genes displayed a significant daily rhythm with an acrophase of expression in the dark phase. The acrophase of lipolytic genes (lipoprotein lipase - lpl, peroxisome proliferator-activated receptor - pparα and hydroxyacil CoA dehydrogenase - hadh) was displayed between ZT 02:17 h and ZT 18:31 h. That of lipogenic genes (leptin-a - lepa, peroxisome proliferator-activated receptor - pparγ, liver X receptor - lxr, insulin-like growth factor - igf1, sterol regulatory element-binding protein - srebp and fatty acid synthase - fas) was displayed between ZT 15:25 h and 20:06 h (dark phase). Feeding time barely influenced daily expression rhythms, except for lxr in the MD group, whose acrophase shifted by about 14 h compared with the ML group (ZT 04:31 h versus ZT 18:29 h, respectively). These results evidence a strong synchronization to the LD cycle, but not to feeding time, and most genes showed a nocturnal acrophase. These findings highlight the importance of considering light and feeding time to optimize lipid metabolism and feeding protocols in fish farming.
Collapse
Affiliation(s)
- J F Paredes
- a Department of Physiology , Faculty of Biology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia , Murcia , Spain
| | - J F López-Olmeda
- a Department of Physiology , Faculty of Biology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia , Murcia , Spain
| | - F J Martínez
- a Department of Physiology , Faculty of Biology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia , Murcia , Spain
| | - F J Sánchez-Vázquez
- a Department of Physiology , Faculty of Biology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia , Murcia , Spain
| |
Collapse
|
57
|
Li J, Papadopoulos V, Vihma V. Steroid biosynthesis in adipose tissue. Steroids 2015; 103:89-104. [PMID: 25846979 DOI: 10.1016/j.steroids.2015.03.016] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 12/25/2022]
Abstract
Tissue-specific expression of steroidogenic enzymes allows the modulation of active steroid levels in a local manner. Thus, the measurement of local steroid concentrations, rather than the circulating levels, has been recognized as a more accurate indicator of the steroid action within a specific tissue. Adipose tissue, one of the largest endocrine tissues in the human body, has been established as an important site for steroid storage and metabolism. Locally produced steroids, through the enzymatic conversion from steroid precursors delivered to adipose tissue, have been proven to either functionally regulate adipose tissue metabolism, or quantitatively contribute to the whole body's steroid levels. Most recently, it has been suggested that adipose tissue may contain the steroidogenic machinery necessary for the initiation of steroid biosynthesis de novo from cholesterol. This review summarizes the evidence indicating the presence of the entire steroidogenic apparatus in adipose tissue and discusses the potential roles of local steroid products in modulating adipose tissue activity and other metabolic parameters.
Collapse
Affiliation(s)
- Jiehan Li
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Biochemistry, McGill University, Montreal, Canada
| | - Vassilios Papadopoulos
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Biochemistry, McGill University, Montreal, Canada.
| | - Veera Vihma
- Folkhälsan Research Center, Helsinki, Finland; University of Helsinki and Helsinki University Central Hospital, Heart and Lung Center, Helsinki, Finland.
| |
Collapse
|
58
|
Ann JY, Eo H, Lim Y. Mulberry leaves (Morus alba L.) ameliorate obesity-induced hepatic lipogenesis, fibrosis, and oxidative stress in high-fat diet-fed mice. GENES AND NUTRITION 2015; 10:46. [PMID: 26463593 DOI: 10.1007/s12263-015-0495-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/29/2015] [Indexed: 01/06/2023]
Abstract
Obesity is associated with chronic diseases such as fatty liver, type 2 diabetes, cardiovascular disease, and severe metabolic syndrome. Obesity causes metabolic impairment including excessive lipid accumulation and fibrosis in the hepatic tissue as well as the increase in oxidative stress. In order to investigate the effect of mulberry leaf (Morus alba L.) extract (MLE) on obesity-induced oxidative stress, lipogenesis, and fibrosis in liver, MLE has been gavaged for 12 weeks in high-fat diet (HFD)-induced obese mice. MLE treatment significantly ameliorated LXRα-mediated lipogenesis and hepatic fibrosis markers such as α-smooth muscle actin, while MLE up-regulated lipolysis-associated markers such as lipoprotein lipase in the HFD-fed mice. Moreover, MLE normalized the activities of antioxidant enzymes including heme oxygenase-1 and glutathione peroxidase in accordance with protein levels of 4-hydroxynonenal in the HFD-fed mice. MLE has beneficial effects on obesity-related fatty liver disease by regulation of hepatic lipid metabolism, fibrosis, and antioxidant defense system. MLE supplementation might be a potential therapeutic approach for obesity-related disease including non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Ji-Young Ann
- Department of Food and Nutrition, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Hyeyoon Eo
- Department of Food and Nutrition, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Yunsook Lim
- Department of Food and Nutrition, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul, 130-701, Republic of Korea.
| |
Collapse
|
59
|
Protein kinase CK2 is necessary for the adipogenic differentiation of human mesenchymal stem cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2207-16. [DOI: 10.1016/j.bbamcr.2015.05.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 11/17/2022]
|
60
|
Lee B, Kwon M, Choi JS, Jeong HO, Chung HY, Kim HR. Kaempferol Isolated from Nelumbo nucifera Inhibits Lipid Accumulation and Increases Fatty Acid Oxidation Signaling in Adipocytes. J Med Food 2015; 18:1363-70. [PMID: 26280739 DOI: 10.1089/jmf.2015.3457] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Stamens of Nelumbo nucifera Gaertn have been used as a Chinese medicine due to its antioxidant, hypoglycemic, and antiatherogenic activity. However, the effects of kaempferol, a main component of N. nucifera, on obesity are not fully understood. We examined the effect of kaempferol on adipogenesis and fatty acid oxidation signaling pathways in 3T3-L1 adipocytes. Kaempferol reduced cytoplasmic triglyceride (TG) accumulation in dose and time-dependent manners during adipocyte differentiation. Accumulation of TG was rapidly reversed by retrieving kaempferol treatment. Kaempferol broadly decreased mRNA or protein levels of adipogenic transcription factors and their target genes related to lipid accumulation. Kaempferol also suppressed glucose uptake and glucose transporter GLUT4 mRNA expression in adipocytes. Furthermore, protein docking simulation suggests that Kaempferol can directly bind to and activate peroxisome proliferator-activated receptor (PPAR)-α by forming hydrophobic interactions with VAL324, THR279, and LEU321 residues of PPARα. The binding affinity was higher than a well-known PPARα agonist fenofibrate. Consistently, mRNA expression levels of PPARα target genes were increased. Our study indicates while kaempferol inhibits lipogenic transcription factors and lipid accumulation, it may bind to PPARα and stimulate fatty acid oxidation signaling in adipocytes.
Collapse
Affiliation(s)
- Bonggi Lee
- 1 Department of Food Science and Nutrition, Pukyong National University , Busan, Korea
| | - Misung Kwon
- 1 Department of Food Science and Nutrition, Pukyong National University , Busan, Korea
| | - Jae Sue Choi
- 1 Department of Food Science and Nutrition, Pukyong National University , Busan, Korea
| | - Hyoung Oh Jeong
- 2 Molecular Inflammation Research Center for Ageing Intervention (MRCA), Pusan National University , Busan, Korea
| | - Hae Young Chung
- 2 Molecular Inflammation Research Center for Ageing Intervention (MRCA), Pusan National University , Busan, Korea.,3 College of Pharmacy, Pusan National University , Busan, Korea
| | - Hyeung-Rak Kim
- 1 Department of Food Science and Nutrition, Pukyong National University , Busan, Korea
| |
Collapse
|
61
|
Lim D, Chai HH, Lee SH, Cho YM, Choi JW, Kim NK. Gene Expression Patterns Associated with Peroxisome Proliferator-activated Receptor (PPAR) Signaling in the Longissimus dorsi of Hanwoo (Korean Cattle). ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2015; 28:1075-83. [PMID: 26104514 PMCID: PMC4478474 DOI: 10.5713/ajas.14.0811] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/29/2014] [Accepted: 01/30/2015] [Indexed: 12/04/2022]
Abstract
Adipose tissue deposited within muscle fibers, known as intramuscular fat (IMF or marbling), is a major determinant of meat quality and thereby affects its economic value. The biological mechanisms that determine IMF content are therefore of interest. In this study, 48 genes involved in the bovine peroxisome proliferator-activated receptor signaling pathway, which is involved in lipid metabolism, were investigated to identify candidate genes associated with IMF in the longissimus dorsi of Hanwoo (Korean cattle). Ten genes, retinoid X receptor alpha, peroxisome proliferator-activated receptor gamma (PPARG), phospholipid transfer protein, stearoyl-CoA desaturase, nuclear receptor subfamily 1 group H member 3, fatty acid binding protein 3 (FABP3), carnitine palmitoyltransferase II, acyl-Coenzyme A dehydrogenase long chain (ACADL), acyl-Coenzyme A oxidase 2 branched chain, and fatty acid binding protein 4, showed significant effects with regard to IMF and were differentially expressed between the low- and high-marbled groups (p<0.05). Analysis of the gene co-expression network based on Pearson’s correlation coefficients identified 10 up-regulated genes in the high-marbled group that formed a major cluster. Among these genes, the PPARG-FABP4 gene pair exhibited the strongest correlation in the network. Glycerol kinase was found to play a role in mediating activation of the differentially expressed genes. We categorized the 10 significantly differentially expressed genes into the corresponding downstream pathways and investigated the direct interactive relationships among these genes. We suggest that fatty acid oxidation is the major downstream pathway affecting IMF content. The PPARG/RXRA complex triggers activation of target genes involved in fatty acid oxidation resulting in increased triglyceride formation by ATP production. Our findings highlight candidate genes associated with the IMF content of the loin muscle of Korean cattle and provide insight into the biological mechanisms that determine adipose deposition within muscle.
Collapse
Affiliation(s)
- Dajeong Lim
- Experiment Research Institute, National Agricultural Products Quality Management Service, Gimcheon 740-870, Korea
| | - Han-Ha Chai
- Experiment Research Institute, National Agricultural Products Quality Management Service, Gimcheon 740-870, Korea
| | - Seung-Hwan Lee
- Experiment Research Institute, National Agricultural Products Quality Management Service, Gimcheon 740-870, Korea
| | - Yong-Min Cho
- Experiment Research Institute, National Agricultural Products Quality Management Service, Gimcheon 740-870, Korea
| | - Jung-Woo Choi
- Experiment Research Institute, National Agricultural Products Quality Management Service, Gimcheon 740-870, Korea
| | - Nam-Kuk Kim
- Experiment Research Institute, National Agricultural Products Quality Management Service, Gimcheon 740-870, Korea
| |
Collapse
|
62
|
Varin A, Thomas C, Ishibashi M, Ménégaut L, Gautier T, Trousson A, Bergas V, de Barros JPP, Narce M, Lobaccaro JMA, Lagrost L, Masson D. Liver X receptor activation promotes polyunsaturated fatty acid synthesis in macrophages: relevance in the context of atherosclerosis. Arterioscler Thromb Vasc Biol 2015; 35:1357-65. [PMID: 25838428 DOI: 10.1161/atvbaha.115.305539] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/18/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Liver X receptors (LXRs) modulate cholesterol and fatty acid homeostasis as well as inflammation. This study aims to decipher the role of LXRs in the regulation of polyunsaturated fatty acid (PUFA) synthesis in macrophages in the context of atherosclerosis. APPROACH AND RESULTS Transcriptomic analysis in human monocytes and macrophages was used to identify putative LXR target genes among enzymes involved in PUFA biosynthesis. In parallel, the consequences of LXR activation or LXR invalidation on PUFA synthesis and distribution were determined. Finally, we investigated the impact of LXR activation on PUFA metabolism in vivo in apolipoprotein E-deficient mice. mRNA levels of acyl-CoA synthase long-chain family member 3, fatty acid desaturases 1 and 2, and fatty acid elongase 5 were significantly increased in human macrophages after LXR agonist treatment, involving both direct and sterol responsive element binding protein-1-dependent mechanisms. Subsequently, pharmacological LXR agonist increased long chain PUFA synthesis and enhanced arachidonic acid content in the phospholipids of human macrophages. Increased fatty acid desaturases 1 and 2 and acyl-CoA synthase long-chain family member 3 mRNA levels as well as increased arachidonic acid to linoleic acid and docosahexaenoic acid to eicosapentaenoic acid ratios were also found in atheroma plaque and peritoneal foam cells from LXR agonist-treated mice. By contrast, murine LXR-deficient macrophages displayed reduced expression of fatty acid elongase 5, acyl-CoA synthase long-chain family member 3 and fatty acid desaturases 1, as well as decreased cellular levels of docosahexaenoic acid and arachidonic acid. CONCLUSIONS Our results indicate that LXR activation triggers PUFA synthesis in macrophages, which results in significant alterations in the macrophage lipid composition. Moreover, we demonstrate here that LXR agonist treatment modulates PUFA metabolism in atherosclerotic arteries.
Collapse
Affiliation(s)
- Alexis Varin
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Charles Thomas
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Minako Ishibashi
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Louise Ménégaut
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Thomas Gautier
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Amalia Trousson
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Victoria Bergas
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Jean Paul Pais de Barros
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Michel Narce
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Jean Marc A Lobaccaro
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - Laurent Lagrost
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.)
| | - David Masson
- From the Centre de Recherche INSERM-UMR866, Université de Bourgogne, Dijon, France (A.V., C.T., M.I., L.M., T.G., V.B., J.P.P.d.B., M.N., L.L., D.M.); Centre Hospitalier Universitaire Dijon, Dijon, France (L.M., L.L., D.M.); Clermont Université, Université Blaise Pascal (A.T., J.M.A.L.) and Inserm, UMR 1103 (A.T., J.M.A.L.), GReD, Aubière, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France (A.T., J.M.A.L.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 6293, Aubière, France (A.T., J.M.A.L.).
| |
Collapse
|
63
|
Lee JE, Kim JM, Jang HJ, Lim SY, Choi SJ, Lee NH, Suh PG, Choi UK. Propyl gallate inhibits adipogenesis by stimulating extracellular signal-related kinases in human adipose tissue-derived mesenchymal stem cells. Mol Cells 2015; 38:336-42. [PMID: 25813451 PMCID: PMC4400308 DOI: 10.14348/molcells.2015.2238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 01/26/2023] Open
Abstract
Propyl gallate (PG) used as an additive in various foods has antioxidant and anti-inflammatory effects. Although the functional roles of PG in various cell types are well characterized, it is unknown whether PG has effect on stem cell differentiation. In this study, we demonstrated that PG could inhibit adipogenic differentiation in human adipose tissue-derived mesenchymal stem cells (hAMSCs) by decreasing the accumulation of intracellular lipid droplets. In addition, PG significantly reduced the expression of adipocyte-specific markers including peroxisome proliferator-activated receptor-γ (PPAR-γ), CCAAT enhancer binding protein-α (C/EBP-α), lipoprotein lipase (LPL), and adipocyte fatty acid-binding protein 2 (aP2). PG inhibited adipogenesis in hAMSCs through extracellular regulated kinase (ERK) pathway. Decreased adipogenesis following PG treatment was recovered in response to ERK blocking. Taken together, these results suggest a novel effect of PG on adipocyte differentiation in hAMSCs, supporting a negative role of ERK1/2 pathway in adipogenic differentiation.
Collapse
Affiliation(s)
- Jeung-Eun Lee
- School of Nano-Bioscience and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798,
Korea
| | - Jung-Min Kim
- School of Nano-Bioscience and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798,
Korea
| | - Hyun-Jun Jang
- School of Nano-Bioscience and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798,
Korea
- Division of Molecular and Life sciences, Pohang University of Science and Technology, Pohang,
Korea
| | - Se-young Lim
- School of Nano-Bioscience and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798,
Korea
| | - Seon-Jeong Choi
- Department of Food Science and Technology, Korea National University of Transportation, Chungbuk 368-701,
Korea
| | - Nan-Hee Lee
- Department of Food Nutrition and Cook, Daegu Science University, Daegu 702-723,
Korea
| | - Pann-Ghill Suh
- School of Nano-Bioscience and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798,
Korea
| | - Ung-Kyu Choi
- Department of Food Science and Technology, Korea National University of Transportation, Chungbuk 368-701,
Korea
| |
Collapse
|
64
|
Zhang GH, Lu JX, Chen Y, Guo PH, Qiao ZL, Feng RF, Chen SE, Bai JL, Huo SD, Ma ZR. ChREBP and LXRα mediate synergistically lipogenesis induced by glucose in porcine adipocytes. Gene 2015; 565:30-8. [PMID: 25827716 DOI: 10.1016/j.gene.2015.03.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/25/2015] [Accepted: 03/26/2015] [Indexed: 01/04/2023]
Abstract
Glucose is a substrate for fatty acid synthesis, and induces lipogenesis and expressions of lipogenic genes. It was proposed that transcriptional factor ChREBP, LXRα and SREBP-1c are key mediators in lipogenesis induced by glucose, however the underlying mechanism remains unclear in porcine adipocytes. In this study, glucose stimulated lipogenesis and expressions of ChREBP, LXRα, SREBP-1c and lipogenic genes FAS and ACC1 in primary porcine adipocytes. When ChREBP expression was knocked down by RNAi, lipogenesis and FAS and ACC1 expressions decreased significantly, and lipogenesis induced by glucose decreased by 75.6%, whereas neither the basal expressions under glucose-free nor glucose induced expressions of LXRα and SREBP-1c were evidently affected, suggesting that ChREBP was a main mediator of lipogenesis stimulated by glucose. Glucose promoted LXRα gene expression, and activation of LXRα by T0901317 increased SREBP-1c expression and enhanced the stimulation of glucose on lipogenesis, but this stimulatory effect of LXRα depended on glucose. Activated LXRα stimulated lipogenesis and ChREBP mRNA expression, which was much lower than that elevated by glucose, and was markedly lower in ChREBP-silencing than in unperturbed adipocytes. SREBP-1c activation blocked by fatostatin markedly decreased lipogenesis and expressions of FAS and ACC1 induced by glucose. Lipogenesis and lipogenic gene expression stimulated by LXRα activation were attenuated by fatostatin, however there was still a slightly increase in ChREBP-silencing adipocytes. These dates suggested that LXRα could directly or through SREBP-1c mediate the lipogenesis induced by glucose. Together, glucose induced lipogenesis and lipogenic gene expressions directly through ChREBP, and directly through LXRα or via SREBP-1c.
Collapse
Affiliation(s)
- Guo Hua Zhang
- College of Life Science and Engineering, Northwest University for Nationalities, Lanzhou, Gansu 730030, China
| | - Jian Xiong Lu
- College of Life Science and Engineering, Northwest University for Nationalities, Lanzhou, Gansu 730030, China.
| | - Yan Chen
- College of Life Science and Engineering, Northwest University for Nationalities, Lanzhou, Gansu 730030, China
| | - Peng Hui Guo
- College of Life Science and Engineering, Northwest University for Nationalities, Lanzhou, Gansu 730030, China
| | - Zi Lin Qiao
- Gansu Engineering Research Center for Animal Cell, Northwest University for Nationalities, Lanzhou, Gansu 730030, China
| | - Ruo Fei Feng
- Gansu Engineering Research Center for Animal Cell, Northwest University for Nationalities, Lanzhou, Gansu 730030, China
| | - Shi En Chen
- College of Life Science and Engineering, Northwest University for Nationalities, Lanzhou, Gansu 730030, China
| | - Jia Lin Bai
- Gansu Engineering Research Center for Animal Cell, Northwest University for Nationalities, Lanzhou, Gansu 730030, China
| | - Sheng Dong Huo
- College of Life Science and Engineering, Northwest University for Nationalities, Lanzhou, Gansu 730030, China
| | - Zhong Ren Ma
- Gansu Engineering Research Center for Animal Cell, Northwest University for Nationalities, Lanzhou, Gansu 730030, China.
| |
Collapse
|
65
|
Frisdal E, Le Lay S, Hooton H, Poupel L, Olivier M, Alili R, Plengpanich W, Villard EF, Gilibert S, Lhomme M, Superville A, Miftah-Alkhair L, Chapman MJ, Dallinga-Thie GM, Venteclef N, Poitou C, Tordjman J, Lesnik P, Kontush A, Huby T, Dugail I, Clement K, Guerin M, Le Goff W. Adipocyte ATP-binding cassette G1 promotes triglyceride storage, fat mass growth, and human obesity. Diabetes 2015; 64:840-55. [PMID: 25249572 DOI: 10.2337/db14-0245] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The role of the ATP-binding cassette G1 (ABCG1) transporter in human pathophysiology is still largely unknown. Indeed, beyond its role in mediating free cholesterol efflux to HDL, the ABCG1 transporter equally promotes lipid accumulation in a triglyceride (TG)-rich environment through regulation of the bioavailability of lipoprotein lipase (LPL). Because both ABCG1 and LPL are expressed in adipose tissue, we hypothesized that ABCG1 is implicated in adipocyte TG storage and therefore could be a major actor in adipose tissue fat accumulation. Silencing of Abcg1 expression by RNA interference in 3T3-L1 preadipocytes compromised LPL-dependent TG accumulation during the initial phase of differentiation. Generation of stable Abcg1 knockdown 3T3-L1 adipocytes revealed that Abcg1 deficiency reduces TG storage and diminishes lipid droplet size through inhibition of Pparγ expression. Strikingly, local inhibition of adipocyte Abcg1 in adipose tissue from mice fed a high-fat diet led to a rapid decrease of adiposity and weight gain. Analysis of two frequent ABCG1 single nucleotide polymorphisms (rs1893590 [A/C] and rs1378577 [T/G]) in morbidly obese individuals indicated that elevated ABCG1 expression in adipose tissue was associated with increased PPARγ expression and adiposity concomitant to increased fat mass and BMI (haplotype AT>GC). The critical role of ABCG1 in obesity was further confirmed in independent populations of severe obese and diabetic obese individuals. This study identifies for the first time a major role of adipocyte ABCG1 in adiposity and fat mass growth and suggests that adipose ABCG1 might represent a potential therapeutic target in obesity.
Collapse
Affiliation(s)
- Eric Frisdal
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | | | - Henri Hooton
- Université Pierre et Marie Curie-Paris 6, Paris, France INSERM, U872, Nutriomique Team 7, Cordeliers Research Center, Paris, France
| | - Lucie Poupel
- Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Maryline Olivier
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France
| | - Rohia Alili
- Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France INSERM, U872, Nutriomique Team 7, Cordeliers Research Center, Paris, France
| | - Wanee Plengpanich
- INSERM, UMR_S1166, Team 4, Paris, France King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Patumwan, Bangkok, Thailand
| | - Elise F Villard
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Sophie Gilibert
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Marie Lhomme
- Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Alexandre Superville
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | | | - M John Chapman
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France
| | | | - Nicolas Venteclef
- Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France INSERM, U872, Nutriomique Team 7, Cordeliers Research Center, Paris, France
| | - Christine Poitou
- Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France INSERM, U872, Nutriomique Team 7, Cordeliers Research Center, Paris, France Heart and Metabolism, Assistance-Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
| | - Joan Tordjman
- Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France INSERM, U872, Nutriomique Team 7, Cordeliers Research Center, Paris, France
| | - Philippe Lesnik
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Anatol Kontush
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Thierry Huby
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Isabelle Dugail
- Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France INSERM, U872, Nutriomique Team 7, Cordeliers Research Center, Paris, France
| | - Karine Clement
- Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France INSERM, U872, Nutriomique Team 7, Cordeliers Research Center, Paris, France Heart and Metabolism, Assistance-Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
| | - Maryse Guerin
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| | - Wilfried Le Goff
- INSERM, UMR_S1166, Team 4, Paris, France Université Pierre et Marie Curie-Paris 6, Paris, France Institute of Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
66
|
Regassa A, Kim WK. Transcriptome analysis of hen preadipocytes treated with an adipogenic cocktail (DMIOA) with or without 20(S)-hydroxylcholesterol. BMC Genomics 2015; 16:91. [PMID: 25765115 PMCID: PMC4347561 DOI: 10.1186/s12864-015-1231-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 01/12/2015] [Indexed: 11/17/2022] Open
Abstract
Background 20(S)-hydroxycholesterol (20(S)) potentially reduces adipogenesis in mammalian cells. The role of this oxysterol and molecular mechanisms underlying the adipogenesis of preadipocytes from laying hens have not been investigated. This study was conducted to 1. Analyze genes differentially expressed between preadipocytes treated with an adipogenic cocktail (DMIOA) containing 500 nM dexamethasone, 0.5 mM 3-isobutyl-1-methylxanthine, 20 μg/mL insulin and 300 μM oleic acid (OA) and control cells and 2. Analyze genes differentially expressed between preadipocytes treated with DMIOA and those treated with DMIOA + 20(S) using Affymetrix GeneChip® Chicken Genome Arrays. Results In experiment one, where we compared the gene expression profile of non-treated (control) cells with those treated with DMIOA, out of 1,221 differentially expressed genes, 755 were over-expressed in control cells, and 466 were over-expressed in cells treated with DMIOA. In experiment two, where we compared the gene expression profile of DMIOA treated cells with those treated with DMIOA+20(S), out of 212 differentially expressed genes, 90 were over-expressed in cells treated with DMIOA, and 122 were over-expressed in those treated with DMIOA+20(S). Genes over-expressed in control cells compared to those treated with DMIOA include those involved in cell-to-cell signaling and interaction (IL6, CNN2, ITGB3), cellular assembly and organization (BMP6, IGF1, ACTB), and cell cycle (CD4, 9, 38). Genes over-expressed in DMIOA compared to control cells include those involved in cellular development (ADAM22, ADAMTS9, FIGF), lipid metabolism (FABP3, 4 and 5), and molecular transport (MAP3K8, PDK4, AGTR1). Genes over-expressed in cells treated with DMIOA compared with those treated with DMIOA+20(S) include those involved in lipid metabolism (ENPP2, DHCR7, DHCR24), molecular transport (FADS2, SLC6A2, CD36), and vitamin and mineral metabolism (BCMO1, AACS, AR). Genes over-expressed in cells treated with DMIOA+20(S) compared with those treated with DMIOA include those involved in cellular growth and proliferation (CD44, CDK6, IL1B), cellular development (ADORA2B, ATP6VOD2, TNFAIP3), and cell-to-cell signaling and interaction (VCAM1, SPON2, VLDLR). Conclusion We identified important adipogenic regulators and key pathways that would help to understand the molecular mechanism of the in vitro adipogenesis in laying hens and demonstrated that 20(S) is capable of suppressing DMIOA-induced adipogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1231-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alemu Regassa
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Woo Kyun Kim
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada. .,Department of Poultry Science, University of Georgia, 303 Poultry Science Building, Athens, GA, 30602, U.S.A.
| |
Collapse
|
67
|
Na TY, Han YH, Ka NL, Park HS, Kang YP, Kwon SW, Lee BH, Lee MO. 22-S-Hydroxycholesterol protects against ethanol-induced liver injury by blocking the auto/paracrine activation of MCP-1 mediated by LXRα. J Pathol 2015; 235:710-20. [PMID: 25557254 PMCID: PMC6084351 DOI: 10.1002/path.4494] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 11/05/2014] [Accepted: 11/29/2014] [Indexed: 02/01/2023]
Abstract
Chronic ethanol consumption causes hepatic steatosis and inflammation, which are associated with liver hypoxia. Monocyte chemoattractant protein‐1 (MCP‐1) is a hypoxia response factor that determines recruitment and activation of monocytes to the site of tissue injury. The level of MCP‐1 is elevated in the serum and liver of patients with alcoholic liver disease (ALD); however, the molecular details regarding the regulation of MCP‐1 expression are not yet understood completely. Here, we show the role of liver X receptor α (LXRα) in the regulation of MCP‐1 expression during the development of ethanol‐induced fatty liver injury, using an antagonist, 22‐S‐hydroxycholesterol (22‐S‐HC). First, administration of 22‐S‐HC attenuated the signs of liver injury with decreased levels of MCP‐1 and its receptor CCR2 in ethanol‐fed mice. Second, hypoxic conditions or treatment with the LXRα agonist GW3965 significantly induced the expression of MCP‐1, which was completely blocked by treatment with 22‐S‐HC or infection by shLXRα lentivirus in the primary hepatocytes. Third, over‐expression of LXRα or GW3965 treatment increased MCP‐1 promoter activity by increasing the binding of hypoxia‐inducible factor‐1α to the hypoxia response elements, together with LXRα. Finally, treatment with recombinant MCP‐1 increased the level of expression of LXRα and LXRα‐dependent lipid droplet accumulation in both hepatocytes and Kupffer cells. These data show that LXRα and its ligand‐induced up‐regulation of MCP‐1 and MCP‐1‐induced LXRα‐dependent lipogenesis play a key role in the autocrine and paracrine activation of MCP‐1 in the pathogenesis of alcoholic fatty liver disease, and that this activation may provide a promising new target for ALD therapy.Copyright © 2014 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tae-Young Na
- College of Pharmacy, Seoul National University, Korea
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Xu H, Wang F, Wang J, Xu J, Wang Y, Xue C. The WNT/β-catenin pathway is involved in the anti-adipogenic activity of cerebrosides from the sea cucumber Cucumaria frondosa. Food Funct 2015; 6:2396-404. [DOI: 10.1039/c5fo00273g] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Both adipocyte hypertrophy and hyperplasia lead to obesity.
Collapse
Affiliation(s)
- Hui Xu
- College of Food Science and Engineering
- Ocean University of China
- Qingdao 266003
- China
| | - Fei Wang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao 266003
- China
| | - Jingfeng Wang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao 266003
- China
| | - Jie Xu
- College of Food Science and Engineering
- Ocean University of China
- Qingdao 266003
- China
| | - Yuming Wang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao 266003
- China
| | - Changhu Xue
- College of Food Science and Engineering
- Ocean University of China
- Qingdao 266003
- China
| |
Collapse
|
69
|
Lee HM, Rim HK, Seo JH, Kook YB, Kim SK, Oh CH, Yoo KH, Jin JS, An HJ. HOX-7 suppresses body weight gain and adipogenesis-related gene expression in high-fat-diet-induced obese mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:505. [PMID: 25515293 PMCID: PMC4320579 DOI: 10.1186/1472-6882-14-505] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/09/2014] [Indexed: 11/16/2022]
Abstract
Background HOX-7 is a newly developed dietary formula composed of traditional oriental herbal medicines. The formula was developed with the aim of improving weight control. We investigated the anti-obesity effect of HOX-7 on high-fat-diet (HFD)-induced obesity in C57BL/6 mice. Methods The mice were divided into four groups and were fed a normal diet (ND), HFD, or HFD with oral administration of HOX-7 at 100 or 200 mg/kg/day for 12 weeks. Body and fat weight, histological changes of fat tissue, and the expression of key adipogenic transcription factors were investigated. Results The body weight of mice fed the HFD with HOX-7 was significantly decreased compared to the HFD group. There were no obvious differences in weekly food intake among the 4 groups. The weight of the epididymal and total fat pads was reduced in mice fed the HFD with HOX-7. Treatment with HOX-7 also substantially attenuated the expression of key adipogenic transcription factors, including peroxisome proliferatoractivated receptor γ, CCAAT/enhancer binding protein α, sterol regulatory element binding protein 1c, adipocyte P2, liver X receptor, and lipoprotein lipase in the epididymal adipose tissue. Conclusion Overall, this study highlighted the anti-obesity effects of HOX-7, a finding that could contribute to the development of natural anti-obesity herbal medicines.
Collapse
|
70
|
|
71
|
He Q, Pu J, Yuan A, Lau WB, Gao E, Koch WJ, Ma XL, He B. Activation of liver-X-receptor α but not liver-X-receptor β protects against myocardial ischemia/reperfusion injury. Circ Heart Fail 2014; 7:1032-41. [PMID: 25277999 DOI: 10.1161/circheartfailure.114.001260] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Liver-X-receptors, LXRα (NR1H3) and LXRβ (NR1H2), encode 2 different but highly homologous isoforms of transcription factors belonging to the nuclear receptor superfamily. Whether LXRα and LXRβ subtypes have discrete roles in the regulation of cardiac physiology/pathology is unknown. We determine the role of each LXR subtype in myocardial ischemia/reperfusion (MI/R) injury. METHODS AND RESULTS Mice (wild type; those genetically depleted of LXRα, LXRβ, or both; and those overexpressing LXRα or LXRβ by in vivo intramyocardial adenoviral vector) were subjected to MI/R injury. Both LXRα and LXRβ were detected in wild-type mouse heart. LXRα, but not LXRβ, was significantly upregulated after MI/R. Dual activation of LXRα and LXRβ by natural and synthetic agonists reduced myocardial infarction and improved contractile function after MI/R. Mechanistically, LXR activation inhibited MI/R-induced oxidative stress and nitrative stress, attenuated endoplasmic reticulum stress and mitochondrial dysfunction, and reduced cardiomyocyte apoptosis in ischemic/reperfused myocardium. The aforementioned cardioprotective effects of LXR agonists were impaired in the setting of cardiac-specific gene silencing of LXRα, but not LXRβ subtype. Moreover, LXRα/β double-knockout and LXRα-knockout mice, but not LXRβ-knockout mice, increased MI/R injury, exacerbated MI/R-induced oxidative/nitrative stress, and aggravated endoplasmic reticulum stress and mitochondrial dysfunction. Furthermore, cardiac LXRα, not LXRβ, overexpression via adenoviral transfection suppressed MI/R injury. CONCLUSIONS Our study provides the first direct evidence that the LXRα, but not LXRβ, subtype is a novel endogenous cardiac protective receptor against MI/R injury. Drug development strategies specifically targeting LXRα may be beneficial in treating ischemic heart disease.
Collapse
Affiliation(s)
- Qing He
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Jun Pu
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.).
| | - Ancai Yuan
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Wayne Bond Lau
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Erhe Gao
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Walter J Koch
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Xin-Liang Ma
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.).
| | - Ben He
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.).
| |
Collapse
|
72
|
Ding L, Li J, Song B, Xiao X, Huang W, Zhang B, Tang X, Qi M, Yang Q, Yang Q, Yang L, Wang Z. Andrographolide prevents high-fat diet-induced obesity in C57BL/6 mice by suppressing the sterol regulatory element-binding protein pathway. J Pharmacol Exp Ther 2014; 351:474-83. [PMID: 25204338 DOI: 10.1124/jpet.114.217968] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sterol regulatory element-binding proteins (SREBPs) are major transcription factors regulating the expression of genes involved in biosynthesis of cholesterol, fatty acids, and triglycerides. We investigated the effect of the specific SREBP suppressor andrographolide, a natural compound isolated from Andrographis paniculata, on the regulation of SREBP signaling by use of Western blot, reporter gene assay, and quantitative real-time polymerase chain reaction analysis. In addition, the antiobesity effects of andrographolide were evaluated in C57BL/6 mice with high-fat diet (HFD)-induced obesity. Our results showed that andrographolide downregulated the expressions of SREBPs target genes and decreased cellular lipid accumulation in vitro. Further, andrographolide (100 mg/kg per day) attenuated HFD-induced body weight gain and fat accumulation in liver or adipose tissues, and improved serum lipid levels and insulin or glucose sensitivity in HFD-induced obese mice. Andrographolide effectively suppressed the respiratory quotient, energy expenditure, and oxygen consumption, which may have contributed to the decreased body-weight gain of the obese mice fed with a HFD. Consistently, andrographolide regulated SREBP target genes and metabolism-associated genes in liver or brown adipose tissue, which may have directly contributed to the lower lipid levels and enhanced insulin sensitivity. Taken together, our results indicated that andrographolide ameliorated lipid metabolism and improved glucose use in mice with HFD-induced obesity. Andrographolide has potential as a leading compound in the prevention or treatment of obesity and insulin resistance.
Collapse
Affiliation(s)
- Lili Ding
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Jinmei Li
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Baoliang Song
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Xu Xiao
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Wendong Huang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Binfeng Zhang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Xiaowen Tang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Meng Qi
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Qiming Yang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Qiaoling Yang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Li Yang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| | - Zhengtao Wang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, People's Republic of China (L.D., J.L., B.Z., X.T., M.Q., Qim.Y., Qia.Y., L.Y., Z.W.); State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China (B.S., X.X.); Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California (W.H.); and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, People's Republic of China (J.L.)
| |
Collapse
|
73
|
Svärd J, Blanco F, Nevin D, Fayne D, Mulcahy F, Hennessy M, Spiers JP. Differential interactions of antiretroviral agents with LXR, ER and GR nuclear receptors: potential contributing factors to adverse events. Br J Pharmacol 2014; 171:480-97. [PMID: 24372550 DOI: 10.1111/bph.12480] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 09/30/2013] [Accepted: 10/15/2013] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Antiretroviral (ARV) drugs activate pregnane X receptors and constitutive androstane receptors, increasing the risk of drug interactions due to altered drug metabolism and disposition. The closely related liver X receptors (LXRα/β), oestrogen receptors (ERα/β) and glucocorticoid receptor (GR) regulate many endogenous processes such as lipid/cholesterol homeostasis, cellular differentiation and inflammation. However, ARV drug activation of these nuclear receptors has not been thoroughly investigated. EXPERIMENTAL APPROACH The ability of an ARV drug library to activate LXRα/β, ERα/β and GR was assessed using a combined in silico and in vitro approach encompassing computational docking and molecular descriptor filtering, cell-free time-resolved fluorescence resonance energy transfer co-activator assays to assess direct binding to ligand-binding domains (LBDs), cell-based reporter assays and target gene expression. KEY RESULTS Direct LBD interactions with LXRα and/or LXRβ were predicted in silico and confirmed in vitro for darunavir, efavirenz, flavopiridol, maraviroc and tipranavir. Likewise, efavirenz was also predicted and confirmed as a ligand of ERα-LBD. Interestingly, atazanavir and ritonavir also activated LXRα/β in reporter assays, while tipranavir enhanced transcriptional activity of ERα. Effects on ER and LXR target gene expression were confirmed for efavirenz and tipranavir. CONCLUSIONS AND IMPLICATIONS There was good agreement between in silico predictions and in vitro results. However, some nuclear receptor interactions identified in vitro were probably due to allosteric effects or nuclear receptor cross-talk, rather than direct LBD binding. This study indicates that some of the adverse effects associated with ARV use may be mediated through 'off-target' effects involving nuclear receptor activation.
Collapse
Affiliation(s)
- J Svärd
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
74
|
Zheng F, Zhang S, Lu W, Wu F, Yin X, Yu D, Pan Q, Li H. Regulation of insulin resistance and adiponectin signaling in adipose tissue by liver X receptor activation highlights a cross-talk with PPARγ. PLoS One 2014; 9:e101269. [PMID: 24972069 PMCID: PMC4074121 DOI: 10.1371/journal.pone.0101269] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 06/05/2014] [Indexed: 01/15/2023] Open
Abstract
Liver X receptors (LXRs) have been recognized as a promising therapeutic target for atherosclerosis; however, their role in insulin sensitivity is controversial. Adiponectin plays a unique role in maintaining insulin sensitivity. Currently, no systematic experiments elucidating the role of LXR activation in insulin function based on adiponectin signaling have been reported. Here, we investigated the role of LXR activation in insulin resistance based on adiponectin signaling, and possible mechanisms. C57BL/6 mice maintained on a regular chow received the LXR agonist, T0901317 (30 mg/kg.d) for 3 weeks by intraperitoneal injection, and differentiated 3T3-L1 adipocytes were treated with T0901317 or GW3965. T0901317 treatment induced significant insulin resistance in C57BL/6 mice. It decreased adiponectin gene transcription in epididymal fat, as well as serum adiponectin levels. Activity of AMPK, a key mediator of adiponectin signaling, was also decreased, resulting in decreased Glut-4 membrane translocation in epididymal fat. In contrast, adiponectin activity was not changed in the liver of T0901317 treated mice. In vitro, both T0901317 and GW3965 decreased adiponectin expression in adipocytes in a dose-dependent manner, an effect which was diminished by LXRα silencing. ChIP-qPCR studies demonstrated that T0901317 decreased the binding of PPARγ to the PPAR-responsive element (PPRE) of the adiponectin promoter in a dose-dependent manner. Furthermore, T0901317 exerted an antagonistic effect on the expression of adiponectin in adipocytes co-treated with 3 µM Pioglitazone. In luciferase reporter gene assays, T0901317 dose-dependently inhibited PPRE-Luc activity in HEK293 cells co-transfected with LXRα and PPARγ. These results suggest that LXR activation induces insulin resistance with decreased adiponectin signaling in epididymal fat, probably due to negative regulation of PPARγ signaling. These findings indicate that the potential of LXR activation as a therapeutic target for atherosclerosis may be limited by the possibility of exacerbating insulin resistance-related disease.
Collapse
Affiliation(s)
- Fenping Zheng
- Department of Endocrinology, Sir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Saifei Zhang
- Department of Endocrinology, Sir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Weina Lu
- Department of Endocrinology, Sir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Fang Wu
- Department of Endocrinology, Sir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Xueyao Yin
- Department of Endocrinology, Sir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Dan Yu
- Department of Endocrinology, Sir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Qianqian Pan
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Hong Li
- Department of Endocrinology, Sir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, P. R. China
- * E-mail:
| |
Collapse
|
75
|
Lee B, Lee M, Lefevre M, Kim HR. Anthocyanins inhibit lipogenesis during adipocyte differentiation of 3T3-L1 preadipocytes. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2014; 69:137-141. [PMID: 24682657 DOI: 10.1007/s11130-014-0407-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Anthocyanins have been shown to suppress body weight and fat mass in animal studies. However, the effect of anthocyanins on the process of lipid accumulation during adipocyte differentiation is not fully understood and the lipogenic transcription factors regulated by anthocyanins have not been identified. We investigated the effects of anthocyanins on lipogenesis pathways during adipocyte differentiation in 3T3-L1 cells. Anthocyanins reduced triglyceride (TG) accumulation in a dose-dependent manner during adipocyte differentiation. Accumulation of TG was rapidly reversed by anthocyanin withdrawal. Anthocyanins markedly reduced gene and protein expression levels of lipogenic transcription factors such as liver X receptor α, sterol regulatory element-binding protein-1c, peroxisome proliferators-activated receptor-γ, and CCAAT enhancer-binding protein-α. In addition, the target gene and protein expression of these lipogenic transcription factors such as fatty acid synthase, stearoyl-CoA desaturase-1, and acetyl-CoA carboxylase α were markedly suppressed by anthocyanins. Thus, anthocyanins suppress lipid accumulation in adipocytes due to broad inhibition of the transcription factors regulating lipogenesis. This may partially explain the mechanism by which anthocyanins exert their anti-obesity effect.
Collapse
Affiliation(s)
- Bonggi Lee
- Department of Food science and Nutrition, Pukyong National University, 45 Yongso-ro, Nam-gu, Busan, 608-737, South Korea
| | | | | | | |
Collapse
|
76
|
Alam MA, Rahman MM. Mitochondrial dysfunction in obesity: potential benefit and mechanism of Co-enzyme Q10 supplementation in metabolic syndrome. J Diabetes Metab Disord 2014; 13:60. [PMID: 24932457 PMCID: PMC4057567 DOI: 10.1186/2251-6581-13-60] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 05/03/2014] [Indexed: 02/06/2023]
Abstract
Co-enzyme Q10 (Co-Q10) is an essential component of the mitochondrial electron transport chain. Most cells are sensitive to co-enzyme Q10 (Co-Q10) deficiency. This deficiency has been implicated in several clinical disorders such as heart failure, hypertension, Parkinson's disease and obesity. The lipid lowering drug statin inhibits conversion of HMG-CoA to mevalonate and lowers plasma Co-Q10 concentrations. However, supplementation with Co-Q10 improves the pathophysiological condition of statin therapy. Recent evidence suggests that Co-Q10 supplementation may be useful for the treatment of obesity, oxidative stress and the inflammatory process in metabolic syndrome. The anti-inflammatory response and lipid metabolizing effect of Co-Q10 is probably mediated by transcriptional regulation of inflammation and lipid metabolism. This paper reviews the evidence showing beneficial role of Co-Q10 supplementation and its potential mechanism of action on contributing factors of metabolic and cardiovascular complications.
Collapse
Affiliation(s)
- Md Ashraful Alam
- School of Biomedical Science, The University of Queensland, Brisbane, Australia
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Md Mahbubur Rahman
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| |
Collapse
|
77
|
Matsusue K, Aibara D, Hayafuchi R, Matsuo K, Takiguchi S, Gonzalez FJ, Yamano S. Hepatic PPARγ and LXRα independently regulate lipid accumulation in the livers of genetically obese mice. FEBS Lett 2014; 588:2277-81. [PMID: 24857376 DOI: 10.1016/j.febslet.2014.05.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/22/2014] [Accepted: 05/02/2014] [Indexed: 12/23/2022]
Abstract
The nuclear hormone receptors liver X receptor α (LXRα) and peroxisome proliferator-activated receptor γ (PPARγ) play key roles in the development of fatty liver. To determine the link between hepatic PPARγ and LXRα signaling and the development of fatty liver, a LXRα-specific ligand, T0901317, was administered to normal OB/OB and genetically obese (ob/ob) mice lacking hepatic PPARγ (Pparγ(ΔH)). In ob/ob-Pparγ(ΔH) and OB/OB-Pparγ(ΔH) mice, as well as ob/ob-Pparγ(WT) and OB/OB-Pparγ(WT) mice, the liver weights and hepatic triglyceride levels were markedly increased in response to T0901317 treatment. These results suggest that hepatic PPARγ and LXRα signals independently contribute to the development of fatty liver.
Collapse
Affiliation(s)
- Kimihiko Matsusue
- Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Daisuke Aibara
- Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Risa Hayafuchi
- Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Kohei Matsuo
- Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Soichi Takiguchi
- Institute for Clinical Research, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka 811-1395, Japan
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shigeru Yamano
- Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
78
|
Pereira-Fernandes A, Vanparys C, Vergauwen L, Knapen D, Jorens PG, Blust R. Toxicogenomics in the 3T3-L1 cell line, a new approach for screening of obesogenic compounds. Toxicol Sci 2014; 140:352-63. [PMID: 24848799 DOI: 10.1093/toxsci/kfu092] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The obesogen hypothesis states that together with an energy imbalance between calories consumed and calories expended, exposure to environmental compounds early in life or throughout lifetime might have an influence on obesity development. In this work, we propose a new approach for obesogen screening, i.e., the use of transcriptomics in the 3T3-L1 pre-adipocyte cell line. Based on the data from a previous study of our group using a lipid accumulation based adipocyte differentiation assay, several human-relevant obesogenic compounds were selected: reference obesogens (Rosiglitazone, Tributyltin), test obesogens (Butylbenzyl phthalate, butylparaben, propylparaben, Bisphenol A), and non-obesogens (Ethylene Brassylate, Bis (2-ethylhexyl)phthalate). The high stability and reproducibility of the 3T3-L1 gene transcription patterns over different experiments and cell batches is demonstrated by this study. Obesogens and non-obesogen gene transcription profiles were clearly distinguished using hierarchical clustering. Furthermore, a gradual distinction corresponding to differences in induction of lipid accumulation could be made between test and reference obesogens based on transcription patterns, indicating the potential use of this strategy for classification of obesogens. Marker genes that are able to distinguish between non, test, and reference obesogens were identified. Well-known genes involved in adipocyte differentiation as well as genes with unknown functions were selected, implying a potential adipocyte-related function of the latter. Cell-physiological lipid accumulation was well estimated based on transcription levels of the marker genes, indicating the biological relevance of omics data. In conclusion, this study shows the high relevance and reproducibility of this 3T3-L1 based in vitro toxicogenomics tool for classification of obesogens and biomarker discovery. Although the results presented here are promising, further confirmation of the predictive value of the set of candidate biomarkers identified as well as the validation of their clinical role will be needed.
Collapse
Affiliation(s)
- Anna Pereira-Fernandes
- Systemic Physiological and Ecotoxicological Research (SPHERE), Department of Biology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerpen, Belgium
| | - Caroline Vanparys
- Systemic Physiological and Ecotoxicological Research (SPHERE), Department of Biology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerpen, Belgium
| | - Lucia Vergauwen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Dries Knapen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Philippe Germaines Jorens
- Department of Clinical Pharmacology/Clinical Toxicology, University of Antwerp, Antwerp University Hospital, Universiteitsplein 1, 2610 Antwerpen, Belgium
| | - Ronny Blust
- Systemic Physiological and Ecotoxicological Research (SPHERE), Department of Biology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerpen, Belgium
| |
Collapse
|
79
|
A-Gonzalez N, Hidalgo A. Nuclear Receptors and Clearance of Apoptotic Cells: Stimulating the Macrophage's Appetite. Front Immunol 2014; 5:211. [PMID: 24860573 PMCID: PMC4026730 DOI: 10.3389/fimmu.2014.00211] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/28/2014] [Indexed: 01/04/2023] Open
Abstract
Clearance of apoptotic cells by macrophages occurs as a coordinated process to ensure tissue homeostasis. Macrophages play a dual role in this process; first, a rapid and efficient phagocytosis of the dying cells is needed to eliminate uncleared corpses that can promote inflammation. Second, after engulfment, macrophages exhibit an anti-inflammatory phenotype, to avoid unwanted immune reactions against cell components. Several nuclear receptors, including liver X receptor and proliferator-activated receptor, have been linked to these two important features of macrophages during apoptotic cell clearance. This review outlines the emerging implications of nuclear receptors in the response of macrophages to cell clearance. These include activation of genes implicated in metabolism, to process the additional cellular content provided by the engulfed cells, as well as inflammatory genes, to maintain apoptotic cell clearance as an “immunologically silent” process. Remarkably, genes encoding receptors for the so-called “eat-me” signals are also regulated by activated nuclear receptors after phagocytosis of apoptotic cells, thus enhancing the efficiency of macrophages to clear dead cells.
Collapse
Affiliation(s)
- Noelia A-Gonzalez
- Department of Epidemiology, Atherothrombosis and Imaging, Fundación Centro Nacional de Investigaciones Cardiovasculares , Madrid , Spain
| | - Andrés Hidalgo
- Department of Epidemiology, Atherothrombosis and Imaging, Fundación Centro Nacional de Investigaciones Cardiovasculares , Madrid , Spain
| |
Collapse
|
80
|
Tung EWY, Boudreau A, Wade MG, Atlas E. Induction of adipocyte differentiation by polybrominated diphenyl ethers (PBDEs) in 3T3-L1 cells. PLoS One 2014; 9:e94583. [PMID: 24722056 PMCID: PMC3983240 DOI: 10.1371/journal.pone.0094583] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/18/2014] [Indexed: 11/18/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are a class of brominated flame retardants that were extensively used in commercial products. PBDEs are ubiquitous environmental contaminants that are both lipophilic and bioaccumulative. Effects of PBDEs on adipogenesis were studied in the 3T3-L1 preadipocyte cell model in the presence and absence of a known adipogenic agent, dexamethasone (DEX). A PBDE mixture designed to mimic body burden of North Americans was tested, in addition to the technical mixture DE-71 and the individual congener BDE-47. The mixture, DE-71, and BDE-47 all induced adipocyte differentiation as assessed by markers for terminal differentiation [fatty acid binding protein 4 (aP2) and perilipin] and lipid accumulation. Characterization of the differentiation process in response to PBDEs indicated that adipogenesis induced by a minimally effective dose of DEX was enhanced by these PBDEs. Moreover, C/EBPα, PPARγ, and LXRα were induced late in the differentiation process. Taken together, these data indicate that adipocyte differentiation is induced by PBDEs; they act in the absence of glucocorticoid and enhance glucocorticoid-mediated adipogenesis.
Collapse
Affiliation(s)
- Emily W. Y. Tung
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Adèle Boudreau
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Michael G. Wade
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
81
|
Dib L, Bugge A, Collins S. LXRα fuels fatty acid-stimulated oxygen consumption in white adipocytes. J Lipid Res 2014; 55:247-57. [PMID: 24259533 PMCID: PMC3886663 DOI: 10.1194/jlr.m043422] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 10/28/2013] [Indexed: 02/06/2023] Open
Abstract
Liver X receptors (LXRs) are transcription factors known for their role in hepatic cholesterol and lipid metabolism. Though highly expressed in fat, the role of LXR in this tissue is not well characterized. We generated adipose tissue LXRα knockout (ATaKO) mice and showed that these mice gain more weight and fat mass on a high-fat diet compared with wild-type controls. White adipose tissue (WAT) accretion in ATaKO mice results from both a decrease in WAT lipolytic and oxidative capacities. This was demonstrated by decreased expression of the β2- and β3-adrenergic receptors, reduced level of phosphorylated hormone-sensitive lipase, and lower oxygen consumption rates (OCRs) in WAT of ATaKO mice. Furthermore, LXR activation in vivo and in vitro led to decreased adipocyte size in WAT and increased glycerol release from primary adipocytes, respectively, with a concomitant increase in OCR in both models. Our findings show that absence of LXRα in adipose tissue results in elevated adiposity through a decrease in WAT oxidation, secondary to attenuated FA availability.
Collapse
Affiliation(s)
- Lea Dib
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL
| | - Anne Bugge
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL
| | - Sheila Collins
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL
| |
Collapse
|
82
|
Escher BI, Allinson M, Altenburger R, Bain PA, Balaguer P, Busch W, Crago J, Denslow ND, Dopp E, Hilscherova K, Humpage AR, Kumar A, Grimaldi M, Jayasinghe BS, Jarosova B, Jia A, Makarov S, Maruya KA, Medvedev A, Mehinto AC, Mendez JE, Poulsen A, Prochazka E, Richard J, Schifferli A, Schlenk D, Scholz S, Shiraishi F, Snyder S, Su G, Tang JYM, van der Burg B, van der Linden SC, Werner I, Westerheide SD, Wong CKC, Yang M, Yeung BHY, Zhang X, Leusch FDL. Benchmarking organic micropollutants in wastewater, recycled water and drinking water with in vitro bioassays. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2014; 48:1940-56. [PMID: 24369993 DOI: 10.1021/es403899t] [Citation(s) in RCA: 312] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Thousands of organic micropollutants and their transformation products occur in water. Although often present at low concentrations, individual compounds contribute to mixture effects. Cell-based bioassays that target health-relevant biological endpoints may therefore complement chemical analysis for water quality assessment. The objective of this study was to evaluate cell-based bioassays for their suitability to benchmark water quality and to assess efficacy of water treatment processes. The selected bioassays cover relevant steps in the toxicity pathways including induction of xenobiotic metabolism, specific and reactive modes of toxic action, activation of adaptive stress response pathways and system responses. Twenty laboratories applied 103 unique in vitro bioassays to a common set of 10 water samples collected in Australia, including wastewater treatment plant effluent, two types of recycled water (reverse osmosis and ozonation/activated carbon filtration), stormwater, surface water, and drinking water. Sixty-five bioassays (63%) showed positive results in at least one sample, typically in wastewater treatment plant effluent, and only five (5%) were positive in the control (ultrapure water). Each water type had a characteristic bioanalytical profile with particular groups of toxicity pathways either consistently responsive or not responsive across test systems. The most responsive health-relevant endpoints were related to xenobiotic metabolism (pregnane X and aryl hydrocarbon receptors), hormone-mediated modes of action (mainly related to the estrogen, glucocorticoid, and antiandrogen activities), reactive modes of action (genotoxicity) and adaptive stress response pathway (oxidative stress response). This study has demonstrated that selected cell-based bioassays are suitable to benchmark water quality and it is recommended to use a purpose-tailored panel of bioassays for routine monitoring.
Collapse
Affiliation(s)
- Beate I Escher
- The University of Queensland , National Research Centre for Environmental Toxicology (Entox), 39 Kessels Rd, Brisbane, QLD 4108, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Sadasivuni MK, Reddy BM, Singh J, Anup MO, Sunil V, Lakshmi MN, Yogeshwari S, Chacko SK, Pooja TL, Dandu A, Harish C, Gopala AS, Pratibha S, Naveenkumar BS, Pallavi PM, Verma MK, Moolemath Y, Somesh BP, Venkataranganna MV, Jagannath MR. CNX-013-B2, a unique pan tissue acting rexinoid, modulates several nuclear receptors and controls multiple risk factors of the metabolic syndrome without risk of hypertriglyceridemia, hepatomegaly and body weight gain in animal models. Diabetol Metab Syndr 2014; 6:83. [PMID: 25143786 PMCID: PMC4138375 DOI: 10.1186/1758-5996-6-83] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 08/06/2014] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND In addition to their role in growth, cellular differentiation and homeostasis Retinoid X Receptors (RXR) regulate multiple physiological and metabolic pathways in various organs that have beneficial glucose and lipid (cholesterol) lowering, insulin sensitizing and anti-obesity effects. Rexinoids, compounds that specifically binds and activate RXR, are therefore considered as potential therapeutics for treating metabolic syndrome. Apparently many of the rexinoids developed in the past increased triglycerides, caused hepatomegaly and also suppressed the thyroid hormone axis. The aim of this study is to evaluate CNX-013-B2, a potent and highly selective rexinoid, for its potential to treat multiple risk factors of the metabolic syndrome. METHODS CNX-013-B2 was selected in a screening system designed to identify compounds that selectively activated only a chosen sub-set of heterodimer partners of RXR of importance to treat insulin resistance. Male C57BL/6j mice (n = 10) on high fat diet (HFD) and 16 week old ob/ob mice (n = 8) were treated orally with CNX-013-B2 (10 mg/kg twice daily) or vehicle for 10 weeks and 4 weeks respectively. Measurement of plasma glucose, triglyceride, cholesterol including LDL-C, glycerol, free fatty acids, feed intake, body weight, oral glucose tolerance and non-shivering thermogenesis were performed at selected time points. After study termination such measurements as organ weight, triglyceride content, mRNA levels, protein phosphorylation along with histological analysis were performed. RESULTS CNX-013-B2 selectively activates PPARs- α, β/δ and γ and modulates activity of LXR, THR and FXR. In ob/ob mice a significant reduction of 25% in fed glucose (p < 0.001 ), a 14% (p < 0.05) reduction in serum total cholesterol and 18% decrease (p < 0.01) in LDL-C and in DIO mice a reduction of 12% (p < 0.01 ) in fasting glucose, 20% in fed triglyceride (p < 0.01) and total cholesterol (p < 0.001) levels, coupled with enhanced insulin sensitivity, cold induced thermogenesis and 7% reduction in body weight were observed. CONCLUSION CNX-013-B2 is an orally bio available selective rexinoid that can be used as a novel therapeutic agent for management of multiple risk factors of the metabolic syndrome without the risk of side effects reported to be associated with rexinoids.
Collapse
|
84
|
Li J, Daly E, Campioli E, Wabitsch M, Papadopoulos V. De novo synthesis of steroids and oxysterols in adipocytes. J Biol Chem 2013; 289:747-64. [PMID: 24280213 DOI: 10.1074/jbc.m113.534172] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Local production and action of cholesterol metabolites such as steroids or oxysterols within endocrine tissues are currently recognized as an important principle in the cell type- and tissue-specific regulation of hormone effects. In adipocytes, one of the most abundant endocrine cells in the human body, the de novo production of steroids or oxysterols from cholesterol has not been examined. Here, we demonstrate that essential components of cholesterol transport and metabolism machinery in the initial steps of steroid and/or oxysterol biosynthesis pathways are present and active in adipocytes. The ability of adipocyte CYP11A1 in producing pregnenolone is demonstrated for the first time, rendering adipocyte a steroidogenic cell. The oxysterol 27-hydroxycholesterol (27HC), synthesized by the mitochondrial enzyme CYP27A1, was identified as one of the major de novo adipocyte products from cholesterol and its precursor mevalonate. Inhibition of CYP27A1 activity or knockdown and deletion of the Cyp27a1 gene induced adipocyte differentiation, suggesting a paracrine or autocrine biological significance for the adipocyte-derived 27HC. These findings suggest that the presence of the 27HC biosynthesis pathway in adipocytes may represent a defense mechanism to prevent the formation of new fat cells upon overfeeding with dietary cholesterol.
Collapse
Affiliation(s)
- Jiehan Li
- From the Research Institute, McGill University Health Centre, and
| | | | | | | | | |
Collapse
|
85
|
Chen YC, Zeng XY, He Y, Liu H, Wang B, Zhou H, Chen JW, Liu PQ, Gu LQ, Ye JM, Huang ZS. Rutaecarpine analogues reduce lipid accumulation in adipocytes via inhibiting adipogenesis/lipogenesis with AMPK activation and UPR suppression. ACS Chem Biol 2013; 8:2301-11. [PMID: 23962138 DOI: 10.1021/cb4003893] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Obesity is characterized by expansion of adipose tissue, which results from an increase in adipocyte number (adipogenesis) and adipocyte size (lipogenesis). A reversal of these processes has been suggested to be a potential antiobetic therapy. Rutaecarpine (Rut) and its novel analogues (R17 and R18) were identified to exert potent effect in reducing lipid accumulation during adipocyte differentiation in 3T3-L1 adipocytes with little cytotoxicity. All three compounds reduced lipid accumulation in a dose-dependent manner, while R17 and R18 exhibited much more potent inhibitory effects compared to that of Rut. Further studies showed that R17 suppressed both adipogenesis and lipogenesis during all stages of adipocyte differentiation as indicated by the reduced protein and mRNA levels of key regulators of adipogenesis/lipogenesis, including PPARγ, C/EBPα, SREBP-1c, ACC, FAS, and SCD-1. We next examined the effect of R17 on the UPR pathway and the results showed that the UPR markers (PERK, eIF2α, IRE1α, and spliced XBP1 mRNA) were all significantly reduced by R17. Further studies revealed that R17 persistently activated AMPK during differentiation, suggesting that the AMPK may be an upstream mechanism for the effect of R17 on adipogenesis and lipogenesis via the adipogenic/lipogenic markers and the UPR pathway. Finally, studies in fast/refeeding mice demonstrated that R17 administration was able to reduce epididymal fat mass and the levels of plasma TG and FFA in vivo. Our results suggest that rutaecarpine analogues may have therapeutic potential for obesity and related metabolic disorders. The mechanism involves the suppression of adipogenic/lipogenic proteins and the suppression of the UPR pathway possibly via the AMPK.
Collapse
Affiliation(s)
- Ying-Chun Chen
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Yi Zeng
- Molecular Pharmacology for Diabetes Group, Health Innovations Research Institute and School of Health Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Yan He
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Hong Liu
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Bin Wang
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Han Zhou
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Jian-Wen Chen
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Pei-Qing Liu
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Lian-Quan Gu
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Ji-Ming Ye
- Molecular Pharmacology for Diabetes Group, Health Innovations Research Institute and School of Health Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Zhi-Shu Huang
- School of Pharmaceutical Sciences and Institute of Medicinal Chemistry, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
86
|
Fan S, Guo L, Zhang Y, Sun Q, Yang B, Huang C. Okra polysaccharide improves metabolic disorders in high-fat diet-induced obese C57BL/6 mice. Mol Nutr Food Res 2013; 57:2075-8. [DOI: 10.1002/mnfr.201300054] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 04/03/2013] [Accepted: 05/11/2013] [Indexed: 01/01/2023]
Affiliation(s)
- Shengjie Fan
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai P. R. China
| | - Lu Guo
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai P. R. China
| | - Yu Zhang
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai P. R. China
| | - Qinhu Sun
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; Shanghai P. R. China
| | - Baican Yang
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai P. R. China
| | - Cheng Huang
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai P. R. China
| |
Collapse
|
87
|
Beaven SW, Matveyenko A, Wroblewski K, Chao L, Wilpitz D, Hsu TW, Lentz J, Drew B, Hevener AL, Tontonoz P. Reciprocal regulation of hepatic and adipose lipogenesis by liver X receptors in obesity and insulin resistance. Cell Metab 2013; 18:106-17. [PMID: 23823481 PMCID: PMC4089509 DOI: 10.1016/j.cmet.2013.04.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 03/28/2013] [Accepted: 04/26/2013] [Indexed: 12/25/2022]
Abstract
Liver X receptors (LXRs) regulate lipogenesis and inflammation, but their contribution to the metabolic syndrome is unclear. We show that LXRs modulate key aspects of the metabolic syndrome in mice. LXRαβ-deficient-ob/ob (LOKO) mice remain obese but show reduced hepatic steatosis and improved insulin sensitivity compared to ob/ob mice. Impaired hepatic lipogenesis in LOKO mice is accompanied by reciprocal increases in adipose lipid storage, reflecting tissue-selective effects on the SREBP, PPARγ, and ChREBP lipogenic pathways. LXRs are essential for obesity-driven SREBP-1c and ChREBP activity in liver, but not fat. Furthermore, loss of LXRs in obesity promotes adipose PPARγ and ChREBP-β activity, leading to improved insulin sensitivity. LOKO mice also exhibit defects in β cell mass and proliferation despite improved insulin sensitivity. Our data suggest that sterol sensing by LXRs in obesity is critically linked with lipid and glucose homeostasis and provide insight into the complex relationships between LXR and insulin signaling.
Collapse
Affiliation(s)
- Simon W Beaven
- Howard Hughes Medical Institute and Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Mizutani Y, Sun H, Ohno Y, Sassa T, Wakashima T, Obara M, Yuyama K, Kihara A, Igarashi Y. Cooperative Synthesis of Ultra Long-Chain Fatty Acid and Ceramide during Keratinocyte Differentiation. PLoS One 2013; 8:e67317. [PMID: 23826266 PMCID: PMC3694974 DOI: 10.1371/journal.pone.0067317] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/16/2013] [Indexed: 12/17/2022] Open
Abstract
The lipid lamellae in the stratum corneum is important for the epidermal permeability barrier. The lipid lamellae component ceramide (CER), comprising an ultra long-chain (ULC) fatty acid (FA) of ≥26 carbons (ULC CER), plays an essential role in barrier formation. ULC acyl-CoAs, produced by the FA elongase ELOVL4, are converted to ULC CERs by the CER synthase CERS3. In the presented study, we observed that ELOVL4 and CERS3 mRNAs increased during keratinocyte differentiation in vivo and in vitro. We also determined that peroxisome proliferator-activated receptor β/δ is involved in the up-regulation of the mRNAs. Knockdown of CERS3 caused a reduction in the elongase activities toward ULC acyl-CoAs, suggesting that CERS3 positively regulates ULCFA. Thus, we reveal that the two key players in ULC CER production in epidermis, CERS3 and ELOVL4, are coordinately regulated at both the transcriptional and enzymatic levels.
Collapse
Affiliation(s)
- Yukiko Mizutani
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Hui Sun
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Yusuke Ohno
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Takayuki Sassa
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Takeshi Wakashima
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Mari Obara
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Kohei Yuyama
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Akio Kihara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- * E-mail: (YI); (AK)
| | - Yasuyuki Igarashi
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
- * E-mail: (YI); (AK)
| |
Collapse
|
89
|
Antiadipogenic Effects of Aster glehni Extract: In Vivo and In Vitro Effects. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:859624. [PMID: 23864899 PMCID: PMC3705792 DOI: 10.1155/2013/859624] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 05/02/2013] [Accepted: 05/27/2013] [Indexed: 11/24/2022]
Abstract
Aster glehni (AG) is a Korean traditional herb that grows in Ulleungdo Island, Republic of Korea. None of the several reports on AG include a determination of the effect of AG on adipogenesis. The primary aim of this study was to determine whether AG attenuates adipogenesis in mouse 3T3-L1 cells and epididymal fat tissue. AG blocked the differentiation of 3T3-L1 preadipocytes in a concentration-dependent manner and suppressed the expression of adipogenesis-related genes such as PPARγ, C/EBPα, and SREBP1c, the master regulators of adipogenesis. Male C57BL/6J mice were divided randomly and equally into 4 diet groups: control diet (CON), high-fat diet (HFD), HFD with 1% AG extract added (AG1), and HFD with 5% AG extract added (AG5). The experimental animals were fed HFD and the 2 combinations for 10 weeks. Mice fed HFD with AG gained less body weight and visceral fat-pad weight than did the mice fed HFD alone. Moreover, AG inhibited the expression of important adipogenic genes such as PPARγ, C/EBPα, SREBP1c, LXR, and leptin in the epididymal adipose tissue of the mice treated with AG1 and AG5. These findings indicate antiadipogenic and antiobesity effects of AG and suggest its therapeutic potential in obesity and obesity-related diseases.
Collapse
|
90
|
Ramos-Nino ME. The role of chronic inflammation in obesity-associated cancers. ISRN ONCOLOGY 2013; 2013:697521. [PMID: 23819063 PMCID: PMC3683483 DOI: 10.1155/2013/697521] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 05/12/2013] [Indexed: 12/20/2022]
Abstract
There is a strong relationship between metabolism and immunity, which can become deleterious under conditions of metabolic stress. Obesity, considered a chronic inflammatory disease, is one example of this link. Chronic inflammation is increasingly being recognized as an etiology in several cancers, particularly those of epithelial origin, and therefore a potential link between obesity and cancer. In this review, the connection between the different factors that can lead to the chronic inflammatory state in the obese individual, as well as their effect in tumorigenesis, is addressed. Furthermore, the association between obesity, inflammation, and esophageal, liver, colon, postmenopausal breast, and endometrial cancers is discussed.
Collapse
Affiliation(s)
- Maria E. Ramos-Nino
- Department of Pathology and Department of Medical Laboratory Sciences, University of Vermont, Burlington, VT, USA
| |
Collapse
|
91
|
Pereira-Fernandes A, Vanparys C, Hectors TLM, Vergauwen L, Knapen D, Jorens PG, Blust R. Unraveling the mode of action of an obesogen: mechanistic analysis of the model obesogen tributyltin in the 3T3-L1 cell line. Mol Cell Endocrinol 2013; 370:52-64. [PMID: 23428407 DOI: 10.1016/j.mce.2013.02.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 02/12/2013] [Accepted: 02/12/2013] [Indexed: 12/30/2022]
Abstract
Obesogenic compounds are chemicals that have an influence on obesity development. This study was designed to unravel the molecular mechanisms of the model obesogen TBT, using microarray analysis in the 3T3-L1 in vitro system, and to evaluate the use of toxicogenomics for obesogen screening. The microarray results revealed enrichment of Gene Ontology terms involved in energy and fat metabolism after 10 days of TBT exposure. Pathway analysis unveiled PPAR signalling pathway as the sole pathway significantly enriched after 1 day and the most significantly enriched pathway after 10 days of exposure. To our knowledge, this is the first study delivering an in depth mechanistic outline of the mode of action of TBT as an obesogen, combining effects on both cell physiological and gene expression level. Furthermore, our results show that combining transcriptomics with 3T3-L1 cells is a promising tool for screening of potential obesogenic compounds.
Collapse
Affiliation(s)
- Anna Pereira-Fernandes
- Systemic Physiological and Ecotoxicological Research (SPHERE), Department of Biology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerpen, Belgium.
| | | | | | | | | | | | | |
Collapse
|
92
|
Molecular mechanisms underlying the anti-obesity potential of prunetin, an O-methylated isoflavone. Biochem Pharmacol 2013; 85:1525-33. [DOI: 10.1016/j.bcp.2013.02.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 02/13/2013] [Accepted: 02/14/2013] [Indexed: 01/21/2023]
|
93
|
Organotins: A review of their reproductive toxicity, biochemistry, and environmental fate. Reprod Toxicol 2013; 36:40-52. [DOI: 10.1016/j.reprotox.2012.11.008] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Revised: 11/05/2012] [Accepted: 11/23/2012] [Indexed: 01/10/2023]
|
94
|
Suzuki S, Suzuki M, Sembon S, Fuchimoto D, Onishi A. Characterization of actions of octanoate on porcine preadipocytes and adipocytes differentiated in vitro. Biochem Biophys Res Commun 2013; 432:92-8. [PMID: 23376076 DOI: 10.1016/j.bbrc.2013.01.080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
Abstract
Octanoate is used to induce adipogenic differentiation and/or lipid accumulation in preadipocytes of domestic animals. However, information on detailed actions of octanoate and the characteristics of octanoate-induced adipocytes is limited. The aim of this study was to examine these issues by comparing the outcomes of the effects of octanoate with those of rosiglitazone, which is a well-defined activator of peroxisome proliferator-activated receptor (PPAR)-γ. The adipocytes that were differentiated with 5mM of octanoate had dispersed and diversely sized lipid droplets compared to those that were differentiated with 1 μM of rosiglitazone. The gene expression levels of adiponectin, glycerol-3-phosphate dehydrogenase, perilipin 1, and perilipin 4 were much higher in the adipocytes that were differentiated with rosiglitazone than in those differentiated with octanoate, while the gene expression levels of lipoprotein lipase and perilipin 2 were decreased in rosiglitazone-differentiated adipocytes compared to octanoate-differentiated adipocytes. However, the expressions of aP2 and CD36 genes were comparably induced. Luciferase reporter assays revealed that PPAR and liver-X-receptor activities were upregulated by octanoate more effectively than by rosiglitazone. Overall, these results suggested that the action of octanoate was complicated and may be dependent on the targeted genes and cellular status.
Collapse
Affiliation(s)
- Shunichi Suzuki
- Transgenic Pig Research Unit, National Institute of Agrobiological Sciences, 2 Ikenodai, Tsukuba, Ibaraki 305-0901, Japan.
| | | | | | | | | |
Collapse
|
95
|
Bonofiglio D, Santoro A, Martello E, Vizza D, Rovito D, Cappello AR, Barone I, Giordano C, Panza S, Catalano S, Iacobazzi V, Dolce V, Andò S. Mechanisms of divergent effects of activated peroxisome proliferator-activated receptor-γ on mitochondrial citrate carrier expression in 3T3-L1 fibroblasts and mature adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1027-36. [PMID: 23370576 DOI: 10.1016/j.bbalip.2013.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 12/14/2022]
Abstract
The citrate carrier (CIC), a nuclear-encoded protein located in the mitochondrial inner membrane, plays an important metabolic role in the transport of acetyl-CoA from the mitochondrion to the cytosol in the form of citrate for fatty acid and cholesterol synthesis. Citrate has been reported to be essential for fibroblast differentiation into fat cells. Because peroxisome proliferator-activated receptor-gamma (PPARγ) is known to be one of the master regulators of adipogenesis, we aimed to study the regulation of CIC by the PPARγ ligand rosiglitazone (BRL) in 3T3-L1 fibroblasts and in adipocytes. We demonstrated that BRL up-regulated CIC mRNA and protein levels in fibroblasts, while it did not elicit any effects in mature adipocytes. The enhancement of CIC levels upon BRL treatment was reversed using the PPARγ antagonist GW9662, addressing how this effect was mediated by PPARγ. Functional experiments using a reporter gene containing rat CIC promoter showed that BRL enhanced CIC promoter activity. Mutagenesis studies, electrophoretic-mobility-shift assay and chromatin-immunoprecipitation analysis revealed that upon BRL treatment, PPARγ and Sp1 are recruited on the Sp1-containing region within the CIC promoter, leading to an increase in CIC expression. In addition, mithramycin, a specific inhibitor for Sp1-DNA binding activity, abolished the PPARγ-mediated up-regulation of CIC in fibroblasts. The stimulatory effects of BRL disappeared in mature adipocytes in which PPARγ/Sp1 complex recruited SMRT corepressor to the Sp1 site of the CIC promoter. Taken together, our results contribute to clarify the molecular mechanisms by which PPARγ regulates CIC expression during the differentiation stages of fibroblasts into mature adipocytes.
Collapse
Affiliation(s)
- Daniela Bonofiglio
- Dept. Pharmacy, Health Sciences and Nutritional, University of Calabria, Cosenza, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Goto T, Kim YI, Takahashi N, Kawada T. Natural compounds regulate energy metabolism by the modulating the activity of lipid-sensing nuclear receptors. Mol Nutr Food Res 2012. [DOI: 10.1002/mnfr.201200522] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology; Graduate School of Agriculture, Kyoto University; Uji Japan
- Research Unit for Physiological Chemistry; the Center for the Promotion of Interdisciplinary Education and Research; Kyoto University; Kyoto Japan
| | - Young-Il Kim
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology; Graduate School of Agriculture, Kyoto University; Uji Japan
| | - Nobuyuki Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology; Graduate School of Agriculture, Kyoto University; Uji Japan
- Research Unit for Physiological Chemistry; the Center for the Promotion of Interdisciplinary Education and Research; Kyoto University; Kyoto Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology; Graduate School of Agriculture, Kyoto University; Uji Japan
- Research Unit for Physiological Chemistry; the Center for the Promotion of Interdisciplinary Education and Research; Kyoto University; Kyoto Japan
| |
Collapse
|
97
|
Mechanisms underlying the rapid peroxisome proliferator-activated receptor-γ-mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimer's disease. J Neurosci 2012; 32:10117-28. [PMID: 22836247 DOI: 10.1523/jneurosci.5268-11.2012] [Citation(s) in RCA: 283] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease is associated with a disruption of amyloid β (Aβ) homeostasis, resulting in the accumulation and subsequent deposition of Aβ peptides within the brain. The peroxisome proliferator-activated receptor-γ (PPARγ) is a ligand-activated nuclear receptor that acts in a coupled metabolic cycle with Liver X Receptors (LXRs) to increase brain apolipoprotein E (apoE) levels. apoE functions to promote the proteolytic clearance of soluble forms of Aβ, and we found that the synthetic PPARγ agonist, pioglitazone, stimulated Aβ degradation by both microglia and astrocytes in an LXR and apoE-dependent manner. Remarkably, a brief 9 d oral treatment of APPswe/PS1Δe9 mice with pioglitazone resulted in dramatic reductions in brain levels of soluble and insoluble Aβ levels which correlated with the loss of both diffuse and dense-core plaques within the cortex. The removal of preexisting amyloid deposits was associated with the appearance of abundant Aβ-laden microglia and astrocytes. Pioglitazone treatment resulted in the phenotypic polarization of microglial cells from a proinflammatory M1 state, into an anti-inflammatory M2 state that was associated with enhanced phagocytosis of deposited forms of amyloid. The reduction in amyloid levels was associated with a reversal of contextual memory deficits in the drug-treated mice. These data provide a mechanistic explanation for how PPARγ activation facilitates amyloid clearance and supports the therapeutic utility of PPARγ agonists for the treatment of Alzheimer's disease.
Collapse
|
98
|
Baek K, Cho JY, Hwang HR, Kwon A, Lee HL, Park HJ, Qadir AS, Ryoo HM, Woo KM, Baek JH. Myeloid Elf-1-like factor stimulates adipogenic differentiation through the induction of peroxisome proliferator-activated receptor γ expression in bone marrow. J Cell Physiol 2012; 227:3603-12. [PMID: 22307523 DOI: 10.1002/jcp.24064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Myeloid Elf-1 like factor (MEF) is one of the Ets transcription factors known to regulate cell proliferation and differentiation. A previous report has shown that osteoblast-specific MEF transgenic mice (Col1a1-MEF TG mice) have low bone mass but high bone marrow adiposity. In the present study, we explored a previously unappreciated mechanism whereby MEF promotes adipogenesis in bone marrow. An adipogenic colony-forming unit assay showed that bone marrow cells derived from Col1a1-MEF TG mice had a higher adipogenic differentiation potential compared to those from wild-type. The levels of adipogenic marker genes expression in 3T3L1 cells were higher when co-cultured with Col1a1-MEF TG bone marrow cells than with wild-type cells. MC3T3-E1 preosteoblasts transfected with MEF secreted higher levels of 15-deoxy-delta (12, 14)-prostaglandin J(2), a potent endogenous ligand of peroxisome proliferator-activated receptor γ (PPARγ), under adipogenic conditions. MEF overexpression increased the adipogenic marker genes expression including PPARγ and lipid droplet accumulation in MC3T3-E1 preosteoblasts and 3T3L1 preadipocytes. Endogenous MEF expression levels increased as adipocyte differentiation proceeded. Knockdown of MEF by siRNA suppressed expression levels of adipogenic marker genes including PPARγ. MEF directly bound to the MEF binding element on the mouse PPARγ promoter, transactivating promoter activity. Immunohistochemical staining of tibia sections demonstrated that bone lining cells and bone marrow cells express higher levels of PPARγ protein in Col1a1-MEF TG mice than in wild-type mice. These results suggest that MEF transactivates PPARγ expression, which, in turn, enhances adipogenic differentiation. Furthermore, MEF overexpressing osteoblasts secrete higher levels of adipogenic factors, creating a marrow microenvironment that favors adipogenesis.
Collapse
Affiliation(s)
- Kyunghwa Baek
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Lyahyai J, Mediano DR, Ranera B, Sanz A, Remacha AR, Bolea R, Zaragoza P, Rodellar C, Martín-Burriel I. Isolation and characterization of ovine mesenchymal stem cells derived from peripheral blood. BMC Vet Res 2012; 8:169. [PMID: 22999337 PMCID: PMC3514285 DOI: 10.1186/1746-6148-8-169] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 09/17/2012] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stem cells with capacity to differentiate into several mesenchymal lineages. This quality makes MSCs good candidates for use in cell therapy. MSCs can be isolated from a variety of tissues including bone marrow and adipose tissue, which are the most common sources of these cells. However, MSCs can also be isolated from peripheral blood. Sheep has been proposed as an ideal model for biomedical studies including those of orthopaedics and transmissible spongiform encephalopathies (TSEs). The aim of this work was to advance these studies by investigating the possibility of MSC isolation from ovine peripheral blood (oPB-MSCs) and by subsequently characterizing there in vitro properties. RESULTS Plastic-adherent fibroblast-like cells were obtained from the mononuclear fraction of blood samples. These cells were analysed for their proliferative and differentiation potential into adipocytes, osteoblasts and chondrocytes, as well as for the gene expression of cell surface markers. The isolated cells expressed transcripts for markers CD29, CD73 and CD90, but failed to express the haematopoietic marker CD45 and expressed only low levels of CD105. The expression of CD34 was variable. The differentiation potential of this cell population was evaluated using specific differentiation media. Although the ability of the cultures derived from different animals to differentiate into adipocytes, osteoblasts and chondrocytes was heterogeneous, we confirmed this feature using specific staining and analysing the gene expression of differentiation markers. Finally, we tested the ability of oPB-MSCs to transdifferentiate into neuronal-like cells. Morphological changes were observed after 24-hour culture in neurogenic media, and the transcript levels of the neurogenic markers increased during the prolonged induction period. Moreover, oPB-MSCs expressed the cellular prion protein gene (PRNP), which was up-regulated during neurogenesis. CONCLUSIONS This study describes for the first time the isolation and characterization of oPB-MSCs. Albeit some variability was observed between animals, these cells retained their capacity to differentiate into mesenchymal lineages and to transdifferentiate into neuron-like cells in vitro. Therefore, oPB-MSCs could serve as a valuable tool for biomedical research in fields including orthopaedics or prion diseases.
Collapse
Affiliation(s)
- Jaber Lyahyai
- Laboratorio de Genética Bioquímica (LAGENBIO), Facultad de Veterinaria, Universidad de Zaragoza, Miguel Servet 177, Zaragoza, 50013, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Meng ZX, Yin Y, Lv JH, Sha M, Lin Y, Gao L, Zhu YX, Sun YJ, Han X. Aberrant activation of liver X receptors impairs pancreatic beta cell function through upregulation of sterol regulatory element-binding protein 1c in mouse islets and rodent cell lines. Diabetologia 2012; 55:1733-44. [PMID: 22415588 DOI: 10.1007/s00125-012-2516-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 02/10/2012] [Indexed: 10/28/2022]
Abstract
AIMS/HYPOTHESIS Liver X receptors (LXR) are important transcriptional regulators of lipid and glucose metabolism. Our previous report demonstrated that LXR activation inhibited pancreatic beta cell proliferation through cell cycle arrest. Here we explore the role of LXR activation in beta cell insulin secretion and the underlying mechanism that might be involved. METHODS Mouse pancreatic islets or insulin-secreting MIN6 cells were exposed to the LXR agonist, T0901317, and insulin secretion, glucose and fatty acid oxidation, and lipogenic gene expression were assessed. The unsaturated fatty acid eicosapentaenoic acid and the dominant negative sterol regulatory element binding protein 1c (SREBP1c) were used to inhibit endogenous SREBP1c and evaluate the involvement of SREBP1c in beta cell dysfunction induced by LXR activation. RESULTS Treatment with the LXR agonist decreased beta cell glucose sensitivity and impaired glucose-stimulated insulin secretion in vivo and in vitro. This was accompanied by derangements of beta cell glucose oxygen consumption, glucose oxidation, ATP production and intracellular voltage-gated calcium channel flux. LXR activation also regulated the expression of lipid metabolism-related genes such as Fas, Acc (also known as Acaca) and Cpt1a, and led to intracellular lipid accumulation. Further studies revealed that inhibition of SREBP1c abolished LXR activation-induced lipid accumulation and improved beta cell glucose metabolism, ATP production and insulin secretion. CONCLUSIONS/INTERPRETATION Our data reveal that aberrant activation of LXR reproduced the phenomenon of beta cell dysfunction in the development of type 2 diabetes in vitro and in vivo. Upregulation of SREBP1c production and the lipotoxicity mediated by it played a central role in this process.
Collapse
Affiliation(s)
- Z X Meng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|