1
|
Jazieh C, Arabi TZ, Asim Z, Sabbah BN, Alsaud AW, Alkattan K, Yaqinuddin A. Unraveling the epigenetic fabric of type 2 diabetes mellitus: pathogenic mechanisms and therapeutic implications. Front Endocrinol (Lausanne) 2024; 15:1295967. [PMID: 38323108 PMCID: PMC10845351 DOI: 10.3389/fendo.2024.1295967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a rapidly escalating global health concern, with its prevalence projected to increase significantly in the near future. This review delves into the intricate role of epigenetic modifications - including DNA methylation, histone acetylation, and micro-ribonucleic acid (miRNA) expression - in the pathogenesis and progression of T2DM. We critically examine how these epigenetic changes contribute to the onset and exacerbation of T2DM by influencing key pathogenic processes such as obesity, insulin resistance, β-cell dysfunction, cellular senescence, and mitochondrial dysfunction. Furthermore, we explore the involvement of epigenetic dysregulation in T2DM-associated complications, including diabetic retinopathy, atherosclerosis, neuropathy, and cardiomyopathy. This review highlights recent studies that underscore the diagnostic and therapeutic potential of targeting epigenetic modifications in T2DM. We also provide an overview of the impact of lifestyle factors such as exercise and diet on the epigenetic landscape of T2DM, underscoring their relevance in disease management. Our synthesis of the current literature aims to illuminate the complex epigenetic underpinnings of T2DM, offering insights into novel preventative and therapeutic strategies that could revolutionize its management.
Collapse
|
2
|
Bian C, Zhang H, Gao J, Wang Y, Li J, Guo D, Wang W, Song Y, Weng Y, Ren H. SIRT6 regulates SREBP1c-induced glucolipid metabolism in liver and pancreas via the AMPKα-mTORC1 pathway. J Transl Med 2022; 102:474-484. [PMID: 34923569 DOI: 10.1038/s41374-021-00715-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to determine the mechanism by which SIRT6 regulates glucolipid metabolism disorders. We detected histological and molecular changes in Sprague-Dawley rats as well as in BRL 3A and INS-1 cell lines subjected to overnutrition and starvation. SIRT6, SREBP1c, and glucolipid metabolism biomarkers were identified by fluorescence co-localization, real-time PCR, and western blotting. Gene silencing studies were performed. Recombinant SIRT6, AMPK agonist (AICAR), mTOR inhibitor (rapamycin), and liver X receptor (LXR) agonist (T0901317) were used to pre-treated in BRL 3A and INS-1 cells. Real-time PCR and western blotting were used to detect related proteins, and cell counting was utilized to detect proliferation. We obtained conflicting results; SIRT6 and SREBP1c appeared in both the liver and pancreas of high-fat and hungry rats. Recombinant SIRT6 alleviated the decrease in AMPKα and increase in mTORC1 (complex of mTOR, Raptor, and Rheb) caused by overnutrition. SIRT6 siRNA reversed the glucolipid metabolic disorders caused by the AMPK agonist and mTOR inhibitor but not by the LXR agonist. Taken together, our results demonstrate that SIRT6 regulates glycolipid metabolism through AMPKα-mTORC1 regulating SREBP1c in the liver and pancreas induced by overnutrition and starvation, independent of LXR.
Collapse
Affiliation(s)
- Che Bian
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Haibo Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Jing Gao
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuxia Wang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jia Li
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Dan Guo
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Wang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuling Song
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Weng
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
3
|
Min J, Ma F, Seyran B, Pellegrini M, Greeff O, Moncada S, Tudzarova S. β-cell-specific deletion of PFKFB3 restores cell fitness competition and physiological replication under diabetogenic stress. Commun Biol 2022; 5:248. [PMID: 35318430 PMCID: PMC8941137 DOI: 10.1038/s42003-022-03209-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
HIF1α and PFKFB3 play a critical role in the survival of damaged β-cells in type–2 diabetes while rendering β-cells non-responsive to glucose stimulation. To discriminate the role of PFKFB3 from HIF1α in vivo, we generated mice with conditional β-cell specific disruption of the Pfkfb3 gene on a human islet pancreatic polypeptide (hIAPP+/−) background and a high-fat diet (HFD) [PFKFB3βKO + diabetogenic stress (DS)]. PFKFB3 disruption in β-cells under DS led to selective purging of hIAPP-damaged β-cells and the disappearance of insulin- and glucagon positive bihormonal cells. PFKFB3 disruption induced a three-fold increase in β-cell replication as evidenced by minichromosome maintenance 2 protein (MCM2) expression. Unlike high-, lower DS or switch to restricted chow diet abolished HIF1α levels and reversed glucose intolerance of PFKFB3βKO DS mice. Our data suggest that replication and functional recovery of β-cells under DS depend on β-cell competitive and selective purification of HIF1α and PFKFB3-positive β-cells. β-cell specific deletion of PFKFB3 results in removal of bihormonal cells and increase in β-cell replication, suggesting that this could lead to β-cell replenishment in type–2 diabetes.
Collapse
Affiliation(s)
- Jie Min
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Department of Endocrinology, Union Hospital of Tongji Medical College Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Feiyang Ma
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Berfin Seyran
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Oppel Greeff
- Department of Pharmacology, University of Pretoria, Pretoria, South Africa
| | | | - Slavica Tudzarova
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Gresch A, Noguera Hurtado H, Wörmeyer L, De Luca V, Wiggers R, Seebohm G, Wünsch B, Düfer M. Selective Inhibition of NMDA receptors with GluN2B subunit protects beta cells against stress-induced apoptotic cell death. J Pharmacol Exp Ther 2021; 379:235-244. [PMID: 34593560 DOI: 10.1124/jpet.121.000807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/28/2021] [Indexed: 11/22/2022] Open
Abstract
Participation of NMDA receptors (NMDARs) in the failure of pancreatic beta cells during development of type 2 diabetes mellitus is discussed. Our study investigates whether beta cell mass and function can be preserved by selectively addressing the GluN2B subunit of the NMDAR. NMDAR activation by NMDA and its co-agonist glycine moderately influenced electrical activity and Ca2+ handling in islet cells at a threshold glucose concentration (4-5 mM) without affecting glucose-mediated insulin secretion. Exposure of islet cells to NMDA/glycine or a glucolipotoxic milieu increased apoptosis by 5 and 8 %, respectively. The GluN2B-specific NMDAR antagonist WMS-1410 (0.1 and 1 µM) partly protected against this. In addition, WMS-1410 completely prevented the decrease in insulin secretion of about 32 % provoked by a 24-h-treatment with NMDA/glycine. WMS-1410 eliminated NMDA-induced changes in the oxidation status of the islet cells and elevated the sensitivity of intracellular calcium to 15 mM glucose. By contrast, WMS-1410 did not prevent the decline in glucose-stimulated insulin secretion occurring after glucolipotoxic culture. This lack of effect was due to a decrease in insulin content to 18 % that obviously could not be compensated by the preservation of cell mass or the higher percentage of insulin release in relation to insulin content. In conclusion, the negative effects of permanent NMDAR activation were effectively counteracted by WMS-1410 as well as the apoptotic cell death induced by high glucose and lipid concentrations. Modulation of NMDARs containing the GluN2B subunit is suggested to preserve beta cell mass during development of type 2 diabetes mellitus. Significance Statement Addressing NMDA receptors containing the GluN2B subunit in pancreatic islet cells has the potential to protect the beta cell mass that progressively declines during the development of type 2 diabetes. Furthermore, this study shows that harmful effects of permanent NMDAR activation can be effectively counteracted by the compound WMS-1410, a selective modulator for NMDARs containing the GluN2B subunit.
Collapse
Affiliation(s)
- Anne Gresch
- Pharmacology, Institute for Pharmaceutical and Medical Chemistry, Germany
| | | | - Laura Wörmeyer
- Pharmaceutical and Medicinal Chemistry, University of Münster, Germany
| | - Vivien De Luca
- Pharmaceutical and Medicinal Chemistry, University of Münster, Germany
| | - Rebekka Wiggers
- Pharmaceutical and Medicinal Chemistry, University of Münster, Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases, University Hospital Münster, Germany
| | - Bernhard Wünsch
- Fachbereich Chemie und Pharmazie, Institut für Pharmazeutische und Medizinische Chemie, Germany
| | - Martina Düfer
- Pharmaceutical and Medicinal Chemistry, University of Münster, Germany
| |
Collapse
|
5
|
Kim JK, Cho IJ, Kim EO, Lee DG, Jung DH, Ki SH, Ku SK, Kim SC. Hemistepsin A inhibits T0901317-induced lipogenesis in the liver. BMB Rep 2021. [PMID: 32843130 PMCID: PMC7907741 DOI: 10.5483/bmbrep.2021.54.2.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Jae Kwang Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Korea
| | - Il Je Cho
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Eun Ok Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Dae Geon Lee
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Dae Hwa Jung
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
- Hani Bio Co., Ltd, Daegu 41059, Korea
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju 61452, Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Sang Chan Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| |
Collapse
|
6
|
Sadeghi MB, Nakhaee A, Saravani R, Sargazi S. Significant association of LXRβ (NR1H2) polymorphisms (rs28514894, rs2303044) with type 2 diabetes mellitus and laboratory characteristics. J Diabetes Metab Disord 2021; 20:261-270. [PMID: 34178836 DOI: 10.1007/s40200-021-00740-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/10/2021] [Indexed: 02/07/2023]
Abstract
Purpose To investigate if single-nucleotide polymorphisms (SNPs) in the NR1H2 gene encoding LXRβ contribute to the development of type-2 diabetes mellitus (T2DM) and whether genotypes of two NR1H2 polymorphisms, rs28514894 and rs2303044, are associated with laboratory characteristics of T2DM patients. Method A total of 900 subjects (450 T2DM cases and 450 healthy subjects) of Iranian ancestry were genotyped for NR1H2 polymorphisms via ARMS-PCR and PCR-RFLP techniques. Result Our findings showed a significant correlation between both polymorphisms and increased risk of T2DM. The haplotype analysis showed an association between the C A haplotype with enhanced risk of T2DM. In T2DM patients, the mean level of HbA1C and BUN significantly differed among carriers of CC and TT genotypes of the rs28514894 polymorphism (P = 0.05 and P < 0.0001, respectively); while in the control group, no significant difference was noticed between subjects with these genotypes. The mean BUN levels also significantly differed among carriers of TC and TT genotypes of this variant in T2DM patients (P = 0.01) and controls (P = 0.04). As for rs2303044 polymorphism, only the mean BUN level significantly differed between GA and GG carriers in T2DM patients (P = 0.006). Compared with CT and TT genotypes, the CC genotype of rs28514894 polymorphism was more frequent in overweight T2DM patients ( 25 < body mass index < 30). Conclusions The present research provided the first documents of the correlation of NR1H2 rs28514894 and rs2303044 polymorphisms with susceptibility to T2DM. Replicated case-control studies on larger populations are needed to validate these findings.
Collapse
Affiliation(s)
- Mohammad Bagher Sadeghi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.,Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Alireza Nakhaee
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.,Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin Saravani
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.,Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
7
|
Zhang Z, Tang S, Gui W, Lin X, Zheng F, Wu F, Li H. Liver X receptor activation induces podocyte injury via inhibiting autophagic activity. J Physiol Biochem 2020; 76:317-328. [PMID: 32328877 DOI: 10.1007/s13105-020-00737-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/17/2020] [Indexed: 12/27/2022]
Abstract
Podocyte injury plays a key role in the occurrence and development of kidney diseases. Decreased autophagic activity in podocyte is closely related to its injury and the occurrence of proteinuria. Liver X receptors (LXRs), as metabolic nuclear receptors, participate in multiple pathophysiological processes and express in several tissues, including podocytes. Although the functional roles of LXRs in the liver, adipose tissue and intestine are well established; however, the effect of LXRs on podocytes function remains unclear. In this study, we used mouse podocytes cell line to investigate the effects of LXR activation on podocytes autophagy level and related signaling pathway by performing Western blotting, RT-PCR, GFP-mRFP-LC3 transfection, and immunofluorescence staining. Then, we tested this effect in STZ-induced diabetic mice. Transmission electron microscopy and immunohistochemistry were employed to explore the effects of LXR activation on podocytes function and autophagic activity. We found that LXR activation could inhibit autophagic flux through blocking the formation of autophagosome in podocytes in vitro which was possibly achieved by affecting AMPK, mTOR, and SIRT1 signaling pathways. Furthermore, LXR activation in vivo induced autophagy suppression in glomeruli, leading to aggravated podocyte injury. In summary, our findings indicated that activation of LXRs induced autophagy suppression, which in turn contributed to the podocyte injury.
Collapse
Affiliation(s)
- Ziyi Zhang
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Road, Hangzhou, 310016, China
| | - Shengjie Tang
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Road, Hangzhou, 310016, China
| | - Weiwei Gui
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Road, Hangzhou, 310016, China
| | - Xihua Lin
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Road, Hangzhou, 310016, China
| | - Fenping Zheng
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Road, Hangzhou, 310016, China
| | - Fang Wu
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Road, Hangzhou, 310016, China
| | - Hong Li
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Road, Hangzhou, 310016, China.
| |
Collapse
|
8
|
Jalil A, Bourgeois T, Ménégaut L, Lagrost L, Thomas C, Masson D. Revisiting the Role of LXRs in PUFA Metabolism and Phospholipid Homeostasis. Int J Mol Sci 2019; 20:ijms20153787. [PMID: 31382500 PMCID: PMC6696407 DOI: 10.3390/ijms20153787] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/19/2023] Open
Abstract
Liver X receptors (LXRs) play a pivotal role in fatty acid (FA) metabolism. So far, the lipogenic consequences of in vivo LXR activation, as characterized by a major hepatic steatosis, has constituted a limitation to the clinical development of pharmacological LXR agonists. However, recent studies provided a different perspective. Beyond the quantitative accumulation of FA, it appears that LXRs induce qualitative changes in the FA profile and in the distribution of FAs among cellular lipid species. Thus, LXRs activate the production of polyunsaturated fatty acids (PUFAs) and their distribution into phospholipids via the control of FA desaturases, FA elongases, lysophosphatidylcholine acyltransferase (LPCAT3), and phospholipid transfer protein (PLTP). Therefore, LXRs control, in a dynamic manner, the PUFA composition and the physicochemical properties of cell membranes as well as the release of PUFA-derived lipid mediators. Recent studies suggest that modulation of PUFA and phospholipid metabolism by LXRs are involved in the control of lipogenesis and lipoprotein secretion by the liver. In myeloid cells, the interplay between LXR and PUFA metabolism affects the inflammatory response. Revisiting the complex role of LXRs in FA metabolism may open new opportunities for the development of LXR modulators in the field of cardiometabolic diseases.
Collapse
Affiliation(s)
- Antoine Jalil
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Thibaut Bourgeois
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Louise Ménégaut
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Laurent Lagrost
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Charles Thomas
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - David Masson
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France.
- INSERM, LNC UMR 1231, F-21000 Dijon, France.
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France.
| |
Collapse
|
9
|
Lee G, Jang H, Kim YY, Choe SS, Kong J, Hwang I, Park J, Im SS, Kim JB. SREBP1c-PAX4 Axis Mediates Pancreatic β-Cell Compensatory Responses Upon Metabolic Stress. Diabetes 2019; 68:81-94. [PMID: 30352876 DOI: 10.2337/db18-0556] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/03/2018] [Indexed: 11/13/2022]
Abstract
SREBP1c is a key transcription factor for de novo lipogenesis. Although SREBP1c is expressed in pancreatic islets, its physiological roles in pancreatic β-cells are largely unknown. In this study, we demonstrate that SREBP1c regulates β-cell compensation under metabolic stress. SREBP1c expression level was augmented in pancreatic islets from obese and diabetic animals. In pancreatic β-cells, SREBP1c activation promoted the expression of cell cycle genes and stimulated β-cell proliferation through its novel target gene, PAX4 Compared with SREBP1c+/+ mice, SREBP1c-/- mice showed glucose intolerance with low insulin levels. Moreover, β-cells from SREBP1c-/- mice exhibited reduced capacity to proliferate and secrete insulin. Conversely, transplantation of SREBP1c-overexpressing islets restored insulin levels and relieved hyperglycemia in streptozotocin-induced diabetic animals. Collectively, these data suggest that pancreatic SREBP1c is a key player in mediating β-cell compensatory responses in obesity.
Collapse
Affiliation(s)
- Gung Lee
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Hagoon Jang
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Ye Young Kim
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sung Sik Choe
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jinuk Kong
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Injae Hwang
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jeu Park
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Seung-Soon Im
- Department of Physiology and Medical Research Center, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jae Bum Kim
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
10
|
Maczewsky J, Sikimic J, Bauer C, Krippeit-Drews P, Wolke C, Lendeckel U, Barthlen W, Drews G. The LXR Ligand T0901317 Acutely Inhibits Insulin Secretion by Affecting Mitochondrial Metabolism. Endocrinology 2017; 158:2145-2154. [PMID: 28449117 DOI: 10.1210/en.2016-1941] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/20/2017] [Indexed: 12/15/2022]
Abstract
The role of liver X receptor (LXR) in pancreatic β-cell physiology and pathophysiology is still unclear. It has been postulated that chronic LXR activation in β-cells induces lipotoxicity, a key step in the development of β-cell dysfunction, which accompanies type 2 diabetes mellitus. In most of these studies, the LXR ligand T0901317 has been administered chronically in the micromolar range to study the significance of LXR activation. In the current study, we have evaluated acute effects of T0901317 on stimulus-secretion coupling of β-cells. We found that 10 µM T0901317 completely suppressed oscillations of the cytosolic Ca2+ concentration induced by 15 mM glucose. Obviously, this effect was due to inhibition of mitochondrial metabolism. T0901317 markedly depolarized the mitochondrial membrane potential, thus inhibiting adenosine triphosphate (ATP) production and reducing the cytosolic ATP concentration. This led in turn to a huge increase in KATP current and hyperpolarization of the cell membrane potential. Eventually, T0901317 inhibited glucose-induced insulin secretion. These effects were rapid in on-set and not compatible with the activation of a nuclear receptor. In vivo, T0901317 acutely increased the blood glucose concentration after intraperitoneal application. In summary, these data clearly demonstrate that T0901317 exerts acute effects on stimulus-secretion coupling. This observation questions the chronic use of T0901317 and limits the interpretation of results obtained under these experimental conditions.
Collapse
Affiliation(s)
- Jonas Maczewsky
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Jelena Sikimic
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Cita Bauer
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Peter Krippeit-Drews
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Carmen Wolke
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Winfried Barthlen
- Department of Pediatric Surgery, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Gisela Drews
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
11
|
Autophagy protects against cholesterol-induced apoptosis in pancreatic β-cells. Biochem Biophys Res Commun 2017; 482:678-685. [DOI: 10.1016/j.bbrc.2016.11.093] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 11/15/2016] [Indexed: 01/04/2023]
|
12
|
Athanason MG, Ratliff WA, Chaput D, MarElia CB, Kuehl MN, Stevens SM, Burkhardt BR. Quantitative proteomic profiling reveals hepatic lipogenesis and liver X receptor activation in the PANDER transgenic model. Mol Cell Endocrinol 2016; 436:41-9. [PMID: 27394190 PMCID: PMC5789791 DOI: 10.1016/j.mce.2016.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/06/2016] [Accepted: 07/05/2016] [Indexed: 12/21/2022]
Abstract
PANcreatic-DERived factor (PANDER) is a member of a superfamily of FAM3 proteins modulating glycemic levels by metabolic regulation of the liver and pancreas. The precise PANDER-induced hepatic signaling mechanism is still being elucidated and has been very complex due to the pleiotropic nature of this novel hormone. Our PANDER transgenic (PANTG) mouse displays a selective hepatic insulin resistant (SHIR) phenotype whereby insulin signaling is blunted yet lipogenesis is increased, a phenomena observed in type 2 diabetes. To examine the complex PANDER-induced mechanism of SHIR, we utilized quantitative mass spectrometry-based proteomic analysis using Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) to reveal the global hepatic proteome differences within the PANTG under the metabolic states of fasting, fed and insulin-stimulated conditions. Proteomic analysis identified lipid metabolism as one of the top cellular functions differentially altered in all metabolic states. Differentially expressed proteins within the PANTG having a lipid metabolic role included ACC, ACLY, CD36, CYP7A1, FASN and SCD1. Central to the differentially expressed proteins involved in lipid metabolism was the predicted activation of the liver X receptor (LXR) pathway. Western analysis validated the increased hepatic expression of LXRα along with LXR-directed targets such as FASN and CYP7A1 within the PANTG liver. Furthermore, recombinant PANDER was capable of inducing LXR promoter activity in-vitro as determined by luciferase reporter assays. Taken together, PANDER strongly impacts hepatic lipid metabolism across metabolic states and may induce a SHIR phenotype via the LXR pathway.
Collapse
Affiliation(s)
- Mark G Athanason
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Whitney A Ratliff
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Catherine B MarElia
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Melanie N Kuehl
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Stanley M Stevens
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Brant R Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA.
| |
Collapse
|
13
|
Abstract
The mechanisms or causes of pancreatic β-cell death as well as impaired insulin secretion, which are the principal events of diabetic etiopathology, are largely unknown. Diabetic complications are known to be associated with abnormal plasma lipid profile, mainly elevated level of cholesterol and free fatty acids. However, in recent years, elevated plasma cholesterol has been implicated as a primary modulator of pancreatic β-cell functions as well as death. High-cholesterol diet in animal models or excess cholesterol in pancreatic β-cell causes transporter desensitization and results in morphometric changes in insulin granules. Moreover, cholesterol is also held responsible to cause oxidative stress, mitochondrial dysfunction, and activation of proapoptotic markers leading to β-cell death. The present review focuses on the pathways and molecularevents that occur in the β-cell under the influence of excess cholesterol that hampers the basal physiology of the cell leading to the progression of diabetes.
Collapse
|
14
|
Hevener AL, Clegg DJ, Mauvais-Jarvis F. Impaired estrogen receptor action in the pathogenesis of the metabolic syndrome. Mol Cell Endocrinol 2015; 418 Pt 3:306-21. [PMID: 26033249 PMCID: PMC5965692 DOI: 10.1016/j.mce.2015.05.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 12/13/2022]
Abstract
Considering the current trends in life expectancy, women in the modern era are challenged with facing menopausal symptoms as well as heightened disease risk associated with increasing adiposity and metabolic dysfunction for up to three decades of life. Treatment strategies to combat metabolic dysfunction and associated pathologies have been hampered by our lack of understanding regarding the biological underpinnings of these clinical conditions and our incomplete understanding of the effects of estrogens and the tissue-specific functions and molecular actions of its receptors. In this review we provide evidence supporting a critical and protective role for the estrogen receptor α specific form in the maintenance of metabolic homeostasis and insulin sensitivity. Studies identifying the ER-regulated pathways required for disease prevention will lay the important foundation for the rational design of targeted therapeutics to improve women's health while limiting complications that have plagued traditional hormone replacement interventions.
Collapse
Affiliation(s)
- Andrea L Hevener
- Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, Iris Cantor-UCLA Women's Health Center, University of California, Los Angeles, CA 90095, USA.
| | - Deborah J Clegg
- Department of Biomedical Sciences, Diabetes and Obesity Research Institute Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Franck Mauvais-Jarvis
- Section of Endocrinology, Department of Medicine Tulane University, Health Science Center New Orleans, New Orleans, LA 70112, USA
| |
Collapse
|
15
|
Emerging role of liver X receptors in cardiac pathophysiology and heart failure. Basic Res Cardiol 2015; 111:3. [PMID: 26611207 PMCID: PMC4661180 DOI: 10.1007/s00395-015-0520-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/03/2015] [Indexed: 01/09/2023]
Abstract
Liver X receptors (LXRs) are master regulators of metabolism and have been studied for their pharmacological potential in vascular and metabolic disease. Besides their established role in metabolic homeostasis and disease, there is mounting evidence to suggest that LXRs may exert direct beneficial effects in the heart. Here, we aim to provide a conceptual framework to explain the broad mode of action of LXRs and how LXR signaling may be an important local and systemic target for the treatment of heart failure. We discuss the potential role of LXRs in systemic conditions associated with heart failure, such as hypertension, diabetes, and renal and vascular disease. Further, we expound on recent data that implicate a direct role for LXR activation in the heart, for its impact on cardiomyocyte damage and loss due to ischemia, and effects on cardiac hypertrophy, fibrosis, and myocardial metabolism. Taken together, the accumulating evidence supports the notion that LXRs may represent a novel therapeutic target for the treatment of heart failure.
Collapse
|
16
|
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: 4.9] [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
|
17
|
Choe SS, Shin KC, Ka S, Lee YK, Chun JS, Kim JB. Macrophage HIF-2α ameliorates adipose tissue inflammation and insulin resistance in obesity. Diabetes 2014; 63:3359-71. [PMID: 24947359 DOI: 10.2337/db13-1965] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In obesity, adipose tissue macrophages (ATMs) play a key role in mediating proinflammatory responses in the adipose tissue, which are associated with obesity-related metabolic complications. Recently, adipose tissue hypoxia has been implicated in the regulation of ATMs in obesity. However, the role of hypoxia-inducible factor (HIF)-2α, one of the major transcription factors induced by hypoxia, has not been fully elucidated in ATMs. In this study, we demonstrate that elevation of macrophage HIF-2α would attenuate adipose tissue inflammation and improve insulin resistance in obesity. In macrophages, overexpression of HIF-2α decreased nitric oxide production and suppressed expression of proinflammatory cytokines through induction of arginase 1. HIF-2α-overexpressing macrophages alleviated proinflammatory responses and improved insulin resistance in adipocytes. In contrast, knockdown of macrophage HIF-2α augmented palmitate-induced proinflammatory gene expression in adipocytes. Furthermore, compared with wild-type mice, Hif-2α heterozygous-null mice aggravated insulin resistance and adipose tissue inflammation with more M1-like ATMs upon high-fat diet (HFD). Moreover, glucose intolerance in HFD-fed Hif-2α heterozygous-null mice was relieved by macrophage depletion with clodronate treatment, implying that increase of proinflammatory ATMs is responsible for insulin resistance by haplodeficiency of Hif-2α upon HFD. Taken together, these data suggest that macrophage HIF-2α would counteract the proinflammatory responses to relieve obesity-induced insulin resistance in adipose tissue.
Collapse
Affiliation(s)
- Sung Sik Choe
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Kyung Cheul Shin
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Sojeong Ka
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Yun Kyung Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Jang-Soo Chun
- Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jae Bum Kim
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| |
Collapse
|
18
|
Li Y, Zhu S, Li B, Shao X, Liu X, Liu A, Wu B, Zhang Y, Wang H, Wang X, Deng K, Liu Q, Huang M, Liu H, Holthöfer H, Zou H. Association between non-alcoholic fatty liver disease and chronic kidney disease in population with prediabetes or diabetes. Int Urol Nephrol 2014; 46:1785-91. [DOI: 10.1007/s11255-014-0796-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 07/21/2014] [Indexed: 02/06/2023]
|
19
|
Kruse MS, Vega MC, Rey M, Coirini H. Sex differences in LXR expression in normal offspring and in rats born to diabetic dams. J Endocrinol 2014; 222:53-60. [PMID: 24824431 DOI: 10.1530/joe-14-0054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Gestational diabetes (GD) alters normal fetal development and is related to a diabetogenic effect in the progeny. Liver X receptors (LXRs) are considered to be potential drug targets for the regulation, treatment, or prevention of diabetes. The aim of this study was to evaluate early and late changes of LXR in the hippocampus and hypothalamus of the male and female offspring of control (CO) and diabetic (DO) mothers. We used an experimental model of streptozotocin-induced GD to assess the protein expression of LXRα (NR1H3) and LXRβ (NR1H2) by western blotting. The tissues were obtained from CO and DO animals at postnatal day 1 (1D), day 10 (10D), and day 35 (35D) and 9 months (9M). In CO, the LXR expression showed significant differences among the groups, which were tissue- and receptor-specific (P<0.05). Sex differences in CO were found only in the hypothalamus for LXRβ expression at 35D and 9M (P<0.05). When CO and DO were compared, differences between them were observed in the majority of the studied groups at 1D (male hippocampus, LXRα 31% and LXRβ 161%; female hippocampus, LXRβ 165%; male hypothalamus, LXRβ 182%; and female hypothalamus, LXRα 85%; P<0.05). However, these differences disappeared later with the exception of LXRβ expression in the male hypothalamus (P<0.05). The area under the curve during the glucose tolerance test correlated negatively with LXRβ in CO but not in DO animals. Moreover, in a male DO subpopulation this correlation was positive as it occurs in intolerant animals. These results indicate that GD affects hypothalamic LXR expression differently in male and female offspring.
Collapse
Affiliation(s)
- María Sol Kruse
- Laboratorio de NeurobiologíaInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN Ciudad Autónoma de Buenos Aires, ArgentinaDepartamento de Bioquímica HumanaFacultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 5to Piso, C1121ABG Ciudad Autónoma de Buenos Aires, ArgentinaFacultad de Ciencias MedicasUniversidad Católica de Cuyo, Rivadavia, Provincia de San Juan, Argentina
| | - María Cristina Vega
- Laboratorio de NeurobiologíaInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN Ciudad Autónoma de Buenos Aires, ArgentinaDepartamento de Bioquímica HumanaFacultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 5to Piso, C1121ABG Ciudad Autónoma de Buenos Aires, ArgentinaFacultad de Ciencias MedicasUniversidad Católica de Cuyo, Rivadavia, Provincia de San Juan, Argentina
| | - Mariana Rey
- Laboratorio de NeurobiologíaInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN Ciudad Autónoma de Buenos Aires, ArgentinaDepartamento de Bioquímica HumanaFacultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 5to Piso, C1121ABG Ciudad Autónoma de Buenos Aires, ArgentinaFacultad de Ciencias MedicasUniversidad Católica de Cuyo, Rivadavia, Provincia de San Juan, Argentina
| | - Héctor Coirini
- Laboratorio de NeurobiologíaInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN Ciudad Autónoma de Buenos Aires, ArgentinaDepartamento de Bioquímica HumanaFacultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 5to Piso, C1121ABG Ciudad Autónoma de Buenos Aires, ArgentinaFacultad de Ciencias MedicasUniversidad Católica de Cuyo, Rivadavia, Provincia de San Juan, ArgentinaLaboratorio de NeurobiologíaInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN Ciudad Autónoma de Buenos Aires, ArgentinaDepartamento de Bioquímica HumanaFacultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 5to Piso, C1121ABG Ciudad Autónoma de Buenos Aires, ArgentinaFacultad de Ciencias MedicasUniversidad Católica de Cuyo, Rivadavia, Provincia de San Juan, ArgentinaLaboratorio de NeurobiologíaInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN Ciudad Autónoma de Buenos Aires, ArgentinaDepartamento de Bioquímica HumanaFacultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 5to Piso, C1121ABG Ciudad Autónoma de Buenos Aires, ArgentinaFacultad de Ciencias MedicasUniversidad Católica de Cuyo, Rivadavia, Provincia de San Juan, Argentina
| |
Collapse
|
20
|
Lee JH, Jung IR, Choi SE, Lee SM, Lee SJ, Han SJ, Kim HJ, Kim DJ, Lee KW, Kang Y. Toxicity generated through inhibition of pyruvate carboxylase and carnitine palmitoyl transferase-1 is similar to high glucose/palmitate-induced glucolipotoxicity in INS-1 beta cells. Mol Cell Endocrinol 2014; 383:48-59. [PMID: 24333689 DOI: 10.1016/j.mce.2013.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 11/14/2013] [Accepted: 12/01/2013] [Indexed: 01/22/2023]
Abstract
This work was initiated to determine whether toxicity generated through inhibition of mitochondrial fuel metabolism is similar to high glucose/palmitate (HG/PA)-induced glucolipotoxicity. Influx of glucose and free fatty acids into the tricarboxylic acid (TCA) cycle was inhibited by treatment with the pyruvate carboxylase (PC) inhibitor phenylacetic acid (PAA) and carnitine palmitoyl transferase-1 (CPT-1) inhibitor etomoxir (Eto), or knockdown of PC and CPT-1. Treatment of PAA/Eto or knockdown of PC/CPT-1 induced apoptotic death in INS-1 beta cells. Similar to HG/PA treatment, PAA/Eto increased endoplasmic reticulum stress responses but decreased the Akt signal. JNK inhibitor or chemical chaperone was protective against both PAA/Eto- and HG/PA-induced cell death. All attempts to reduce [Ca²⁺](i), stimulate lipid metabolism, and increase the TCA cycle intermediate pool protected PAA/Eto-induced death as well as HG/PA-induced death. These data suggest that signals induced from impaired mitochondrial fuel metabolism play a critical role in HG/PA-induced glucolipotoxicity.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Life Science, Korea University Seoul 136-701, Republic of Korea
| | - Ik-Rak Jung
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sung-E Choi
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sung-Mi Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Soo-Jin Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Kwan-Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Yup Kang
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea.
| |
Collapse
|
21
|
Ding L, Pang S, Sun Y, Tian Y, Yu L, Dang N. Coordinated Actions of FXR and LXR in Metabolism: From Pathogenesis to Pharmacological Targets for Type 2 Diabetes. Int J Endocrinol 2014; 2014:751859. [PMID: 24872814 PMCID: PMC4020365 DOI: 10.1155/2014/751859] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 04/09/2014] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes (T2D) is the most prevalent metabolic disease, and many people are suffering from its complications driven by hyperglycaemia and dyslipidaemia. Nuclear receptors (NRs) are ligand-inducible transcription factors that mediate changes to metabolic pathways within the body. As metabolic regulators, the farnesoid X receptor (FXR) and the liver X receptor (LXR) play key roles in the pathogenesis of T2D, which remains to be clarified in detail. Here we review the recent progress concerning the physiological and pathophysiological roles of FXRs and LXRs in the regulation of bile acid, lipid and glucose metabolism and the implications in T2D, taking into account that these two nuclear receptors are potential pharmaceutical targets for the treatment of T2D and its complications.
Collapse
Affiliation(s)
- Lin Ding
- Endocrinology Department, Jinan Central Hospital Affiliated to Shandong University, No. 105 Jiefang Road, Jinan, Shandong 250013, China
| | - Shuguang Pang
- Endocrinology Department, Jinan Central Hospital Affiliated to Shandong University, No. 105 Jiefang Road, Jinan, Shandong 250013, China
- *Shuguang Pang:
| | - Yongmei Sun
- Endocrinology Department, Jinan Central Hospital Affiliated to Shandong University, No. 105 Jiefang Road, Jinan, Shandong 250013, China
| | - Yuling Tian
- Endocrinology Department, Jinan Central Hospital Affiliated to Shandong University, No. 105 Jiefang Road, Jinan, Shandong 250013, China
| | - Li Yu
- Endocrinology Department, Jinan Central Hospital Affiliated to Shandong University, No. 105 Jiefang Road, Jinan, Shandong 250013, China
| | - Ningning Dang
- Endocrinology Department, Jinan Central Hospital Affiliated to Shandong University, No. 105 Jiefang Road, Jinan, Shandong 250013, China
| |
Collapse
|
22
|
Cermenati G, Brioschi E, Abbiati F, Melcangi RC, Caruso D, Mitro N. Liver X receptors, nervous system, and lipid metabolism. J Endocrinol Invest 2013; 36:435-43. [PMID: 23609963 DOI: 10.3275/8941] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Lipids in the nervous system are represented by cholesterol and phospholipids as constituents of cell membranes and, in particular, of myelin. Therefore, lipids are finely regulated to guarantee physiological functions. In the central nervous system, cholesterol is locally synthesized due to the presence of the blood brain barrier. In the peripheral nervous system cholesterol is either up-taken by lipoproteins and/or produced by de novo biosynthesis. Defects in lipid homeostasis in these tissues lead to structural and functional changes that often result in different pathological conditions depending on the affected pathways (i.e. cholesterol biosynthesis, cholesterol efflux, fatty acid biosynthesis etc.). Alterations in cholesterol metabolism in the central nervous system are linked to several disorders such as Alzheimer's disease, Huntington disease, Parkinson disease, Multiple sclerosis, Smith-Lemli-Opitz syndrome, Niemann-Pick type C disease, and glioblastoma. In the peripheral nervous system changes in lipid metabolism are associated with the development of peripheral neuropathy that may be caused by metabolic disorders, injuries, therapeutics, and autoimmune diseases. Transcription factors, such as the Liver X receptors (LXR), regulate both cholesterol and fatty acid metabolism in several tissues including the nervous system. In the last few years several studies elucidated the biology of LXR in the nervous system due to the availability of knock-out mice and the development of synthetic ligands. Here, we review a survey of the literature focused on the central and peripheral nervous system and in physiological and pathological settings with particular attention to the roles played by LXR in both districts.
Collapse
Affiliation(s)
- G Cermenati
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, Italy
| | | | | | | | | | | |
Collapse
|
23
|
Mauvais-Jarvis F, Clegg DJ, Hevener AL. The role of estrogens in control of energy balance and glucose homeostasis. Endocr Rev 2013; 34:309-38. [PMID: 23460719 PMCID: PMC3660717 DOI: 10.1210/er.2012-1055] [Citation(s) in RCA: 823] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Estrogens play a fundamental role in the physiology of the reproductive, cardiovascular, skeletal, and central nervous systems. In this report, we review the literature in both rodents and humans on the role of estrogens and their receptors in the control of energy homeostasis and glucose metabolism in health and metabolic diseases. Estrogen actions in hypothalamic nuclei differentially control food intake, energy expenditure, and white adipose tissue distribution. Estrogen actions in skeletal muscle, liver, adipose tissue, and immune cells are involved in insulin sensitivity as well as prevention of lipid accumulation and inflammation. Estrogen actions in pancreatic islet β-cells also regulate insulin secretion, nutrient homeostasis, and survival. Estrogen deficiency promotes metabolic dysfunction predisposing to obesity, the metabolic syndrome, and type 2 diabetes. We also discuss the effect of selective estrogen receptor modulators on metabolic disorders.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA.
| | | | | |
Collapse
|
24
|
Kim HJ, Yoon KA, Yoon HJ, Hong JM, Lee MJ, Lee IK, Kim SY. Liver X receptor activation inhibits osteoclastogenesis by suppressing NF-κB activity and c-Fos induction and prevents inflammatory bone loss in mice. J Leukoc Biol 2013; 94:99-107. [PMID: 23657115 DOI: 10.1189/jlb.1112601] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
LXRs are nuclear receptors that function as important regulators of lipid homeostasis and inflammatory responses. LXR activation has been shown to suppress RANKL-induced osteoclast differentiation, but its underlying mechanisms and its influence on inflammatory bone destruction remain unclear. In this study, we report that the LXR agonists T0901317 and GW3965 inhibit osteoclastogenesis from primary BMMs in a dose-dependent manner. LXR activation suppressed RANKL-induced transcriptional activity of NF-κB without affecting IκBα degradation and the phosphorylation of p38. LXR agonists significantly suppressed RANKL-induced expression of c-Fos and NFATc1, which are crucial transcription factors for osteoclastogenesis. The activation of LXRs also inhibited RANKL-mediated AP-1 transcriptional activity. Furthermore, LXR activation attenuated PPARγ ligand-induced c-Fos expression, and LXR suppressed AP-1 promoter activity by PPARγ. The inhibitory effect of LXR activation on osteoclastogenesis was reversed by overexpression of c-Fos, suggesting that c-Fos is a downstream target of the antiosteoclastogenic action of LXRs. In addition to osteoclast differentiation, LXR activation accelerated apoptosis in mature osteoclasts by the induction of caspase-3 and -9 activity and Bim expression. Consistent with the in vitro effects we observed, the administration of a LXR agonist protected from bone loss induced by LPS in vivo. Together, our data provide evidence that LXRs may have potential as therapeutic targets for bone resorption-associated diseases.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Kyungpook National University Hospital, 44-2 Samduk 2-ga, Jung-gu, Daegu 700-412, South Korea.
| | | | | | | | | | | | | |
Collapse
|
25
|
Jin SH, Yang JH, Shin BY, Seo K, Shin SM, Cho IJ, Ki SH. Resveratrol inhibits LXRα-dependent hepatic lipogenesis through novel antioxidant Sestrin2 gene induction. Toxicol Appl Pharmacol 2013; 271:95-105. [PMID: 23651738 DOI: 10.1016/j.taap.2013.04.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/06/2013] [Accepted: 04/25/2013] [Indexed: 01/04/2023]
Abstract
Liver X receptor-α (LXRα), a member of the nuclear receptor superfamily of ligand-activated transcription factors, regulates de novo fatty acid synthesis that leads to stimulate hepatic steatosis. Although, resveratrol has beneficial effects on metabolic disease, it is not known whether resveratrol affects LXRα-dependent lipogenic gene expression. This study investigated the effect of resveratrol in LXRα-mediated lipogenesis and the underlying molecular mechanism. Resveratrol inhibited the ability of LXRα to activate sterol regulatory element binding protein-1c (SREBP-1c) and thereby inhibited target gene expression in hepatocytes. Moreover, resveratrol decreased LXRα-RXRα DNA binding activity and LXRE-luciferase transactivation. Resveratrol is known to activate Sirtuin 1 (Sirt1) and AMP-activated protein kinase (AMPK), although its precise mechanism of action remains controversial. We found that the ability of resveratrol to repress T0901317-induced SREBP-1c expression was not dependent on AMPK and Sirt1. It is well established that hepatic steatosis is associated with antioxidant and redox signaling. Our data showing that expression of Sestrin2 (Sesn2), which is a novel antioxidant gene, was significantly down-regulated in the livers of high-fat diet-fed mice. Moreover, resveratrol up-regulated Sesn2 expression, but not Sesn1 and Sesn3. Sesn2 overexpression repressed LXRα-activated SREBP-1c expression and LXRE-luciferase activity. Finally, Sesn2 knockdown using siRNA abolished the effect of resveratrol in LXRα-induced FAS luciferase gene transactivation. We conclude that resveratrol affects Sesn2 gene induction and contributes to the inhibition of LXRα-mediated hepatic lipogenesis.
Collapse
Affiliation(s)
- So Hee Jin
- College of Pharmacy, Chosun University, Gwangju 501-759, South Korea
| | | | | | | | | | | | | |
Collapse
|
26
|
Gao M, Liu D. The liver X receptor agonist T0901317 protects mice from high fat diet-induced obesity and insulin resistance. AAPS JOURNAL 2012. [PMID: 23180161 DOI: 10.1208/s12248-012-9429-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The effect of activation of liver X receptor by N-(2,2,2-trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1(trifluoromethyl)ethyl]phenyl] benzenesulfonamide (T0901317) on high fat diet (HFD)-induced obesity and insulin resistance was examined in C57BL/6 mice. When on HFD continuously for 10 weeks, C57BL/6 mice became obese with an average body weight of 42 g, insulin resistant, and glucose intolerant. Twice weekly intraperitoneal injections of T0901317 at 50 mg/kg in animals on the same diet completely blocked obesity development, obesity-associated insulin resistance, and glucose intolerance. Quantitative real-time PCR analysis showed that T0901317-treated animals had significantly higher mRNA levels of genes involved in energy metabolism, including Ucp-1, Pgc1a, Pgc1b, Cpt1a, Cpt1b, Acadm, Acadl, Aox, and Ehhadh. Transcription activation of Cyp7a1, Srebp-1c, Fas, Scd-1, and Acc-1 genes was also seen in T0901317-treated animals. T0901317 treatment induced reversible aggregation of lipids in the liver. These results suggest that liver X receptor could be a potential target for prevention of obesity and obesity-associated insulin resistance.
Collapse
Affiliation(s)
- Mingming Gao
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA
| | | |
Collapse
|
27
|
Jang H, Lee G, Kong J, Choi G, Park YJ, Kim JB. Feeding period restriction alters the expression of peripheral circadian rhythm genes without changing body weight in mice. PLoS One 2012; 7:e49993. [PMID: 23166806 PMCID: PMC3499481 DOI: 10.1371/journal.pone.0049993] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 10/15/2012] [Indexed: 01/10/2023] Open
Abstract
Accumulating evidence suggests that the circadian clock is closely associated with metabolic regulation. However, whether an impaired circadian clock is a direct cause of metabolic dysregulation such as body weight gain is not clearly understood. In this study, we demonstrate that body weight gain in mice is not significantly changed by restricting feeding period to daytime or nighttime. The expression of peripheral circadian clock genes was altered by feeding period restriction, while the expression of light-regulated hypothalamic circadian clock genes was unaffected by either a normal chow diet (NCD) or a high-fat diet (HFD). In the liver, the expression pattern of circadian clock genes, including Bmal1, Clock, and Per2, was changed by different feeding period restrictions. Moreover, the expression of lipogenic genes, gluconeogenic genes, and fatty acid oxidation-related genes in the liver was also altered by feeding period restriction. Given that feeding period restriction does not affect body weight gain with a NCD or HFD, it is likely that the amount of food consumed might be a crucial factor in determining body weight. Collectively, these data suggest that feeding period restriction modulates the expression of peripheral circadian clock genes, which is uncoupled from light-sensitive hypothalamic circadian clock genes.
Collapse
Affiliation(s)
- Hagoon Jang
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Gung Lee
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Jinuk Kong
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Goun Choi
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Yoon Jeong Park
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, Korea
| | - Jae Bum Kim
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, Korea
- * E-mail:
| |
Collapse
|
28
|
Kumar R, Parsad D, Kanwar A, Kaul D. Altered levels of LXR-α: crucial implications in the pathogenesis of vitiligo. Exp Dermatol 2012; 21:853-8. [DOI: 10.1111/exd.12017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2012] [Indexed: 01/13/2023]
Affiliation(s)
- Ravinder Kumar
- Department of Dermatology; Postgraduate Institute of Medical Education and Research; Chandigarh; India
| | - Davinder Parsad
- Department of Dermatology; Postgraduate Institute of Medical Education and Research; Chandigarh; India
| | - Amrinderjit Kanwar
- Department of Dermatology; Postgraduate Institute of Medical Education and Research; Chandigarh; India
| | - Deepak Kaul
- Department of Experimental Medicine and Biotechnology; Postgraduate Institute of Medical Education and Research; Chandigarh; India
| |
Collapse
|
29
|
Kruse MS, Rey M, Vega MC, Coirini H. Alterations of LXRα and LXRβ expression in the hypothalamus of glucose-intolerant rats. J Endocrinol 2012; 215:51-8. [PMID: 22836489 DOI: 10.1530/joe-12-0088] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Liver X receptor (LXR) α and β are nuclear receptors that are crucial for the regulation of carbohydrate and lipid metabolism. Activation of LXRs in the brain facilitates cholesterol clearance and improves cognitive deficits, thus they are considered as promising drug targets to treat diseases such as atherosclerosis and Alzheimer's disease. Nevertheless, little is known about the function and localization of LXRs in the brain. Here, we studied the expression of LXR in the brains of rats that received free access to 10% (w/v) fructose group (FG) in their beverages or water control drinks (control group (CG)). After 6 weeks rats in the FG presented with hypertriglyceridemia, hyperinsulinemia, and became glucose intolerant, suggesting a progression toward type 2 diabetes. We found that hypothalamic LXR expression was altered in fructose-fed rats. Rats in the FG presented with a decrease in LXRβ levels while showing an increase in LXRα expression in the hypothalamus but not in the hippocampus, cerebellum, or neocortex. Moreover, both LXRα and β expression correlated negatively with insulin and triglyceride levels. Interestingly, LXRβ showed a negative correlation with the area under the curve during the glucose tolerance test in the CG and a positive correlation in the FG. Immunocytochemistry revealed that the paraventricular and ventromedial nuclei express mainly LXRα whereas the arcuate nucleus expresses LXRβ. Both LXR immunosignals were found in the median preoptic area. This is the first study showing a relationship between glucose and lipid homeostasis and the expression of LXRs in the hypothalamus, suggesting that LXRs may trigger neurochemical and neurophysiological responses for the control of food intake and energy expenditure through these receptors.
Collapse
Affiliation(s)
- María Sol Kruse
- Laboratorio de Neurobiología, Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
30
|
Yoon CH, Kwon YJ, Lee SW, Park YB, Lee SK, Park MC. Activation of Liver X Receptors Suppresses Inflammatory Gene Expressions and Transcriptional Corepressor Clearance in Rheumatoid Arthritis Fibroblast Like Synoviocytes. J Clin Immunol 2012; 33:190-9. [DOI: 10.1007/s10875-012-9799-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 09/10/2012] [Indexed: 12/20/2022]
|
31
|
Zhang X, Bai Q, Xu L, Kakiyama G, Pandak WM, Zhang Z, Ren S. Cytosolic sulfotransferase 2B1b promotes hepatocyte proliferation gene expression in vivo and in vitro. Am J Physiol Gastrointest Liver Physiol 2012; 303:G344-55. [PMID: 22679001 PMCID: PMC3423104 DOI: 10.1152/ajpgi.00403.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cytosolic sulfotransferase 2B1b (SULT2B1b) catalyzes the sulfation of 3β-hydroxysteroids and functions as a selective cholesterol and oxysterol sulfotransferase. Activation of liver X receptors (LXRs) by oxysterols has been known to be an antiproliferative factor. Overexpression of SULT2B1b impairs LXR's response to oxysterols, by which it regulates lipid metabolism. The aim of this study was to investigate in vivo and in vitro effects of SULT2B1b on liver proliferation and the underlying mechanisms. Primary rat hepatocytes and C57BL/6 mice were infected with adenovirus encoding SULT2B1b. Liver proliferation was determined by measuring the proliferating cell nuclear antigen (PCNA) immunostaining labeling index. The correlation between SULT2B1b and PCNA expression in mouse liver tissues was determined by double immunofluorescence. Gene expressions were evaluated by quantitative real-time PCR and Western blot analysis. SULT2B1b overexpression in mouse liver tissues increased PCNA-positive cells in a dose- and time-dependent manner. The increased expression of PCNA in mouse liver tissues was only observed in the SULT2B1b transgenic cells. Small interference RNA SULT2B1b significantly inhibited cell cycle regulatory gene expressions in primary rat hepatocytes. LXR activation by T0901317 effectively suppressed SULT2B1b-induced gene expression in vivo and in vitro. SULT2B1b may promote hepatocyte proliferation by inactivating oxysterol/LXR signaling.
Collapse
Affiliation(s)
- Xin Zhang
- 1Department of Pathology, Fudan University Shanghai Medical College, Shanghai, China; and ,2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - Qianming Bai
- 1Department of Pathology, Fudan University Shanghai Medical College, Shanghai, China; and ,2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - Leyuan Xu
- 2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - Genta Kakiyama
- 2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - William M. Pandak
- 2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - Zhigang Zhang
- 1Department of Pathology, Fudan University Shanghai Medical College, Shanghai, China; and
| | - Shunlin Ren
- 2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| |
Collapse
|
32
|
Tiano JP, Mauvais-Jarvis F. Molecular mechanisms of estrogen receptors' suppression of lipogenesis in pancreatic β-cells. Endocrinology 2012; 153:2997-3005. [PMID: 22564979 PMCID: PMC3380304 DOI: 10.1210/en.2011-1980] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The gonadal steroid, 17β-estradiol (E2), suppresses pancreatic islet fatty acid and glycerolipid synthesis and prevents β-cell failure in rodent models of type 2 diabetes. β-Cell estrogen receptors (ER) mediate these actions by suppressing the expression and enzymatic activity of fatty acid synthase (FAS). Here, we explored the mechanism of FAS suppression. We show that E2, and pharmacological agonists for ERα, ERβ, and the G protein-coupled ER, suppress mRNA and protein expression of the transcriptional regulators of FAS, namely, sterol regulatory element-binding protein 1c (SREBP1c) and carbohydrate response element binding protein (ChREBP) in insulin-secreting INS-1 cells. ER suppress SREBP1c and ChREBP mRNA and protein expression via an extranuclear localization. Using two mouse lines with pancreas-specific null deletion of either ERα or the signal transducer and activator of transcription 3 (STAT3), we show that ERα activation in vivo reduces SREBP1c and ChREBP mRNA expression via a direct islet action involving STAT3 activation. The master regulators of lipogenesis, liver X receptor (LXR) α and β, transcriptionally up-regulate SREBP1c and ChREBP. We find that activation of ERα, ERβ, and G protein-coupled ER suppresses LXR's mRNA expression in INS-1 cells. We also observe that activation of ERα in mouse islets in vivo suppresses LXR mRNA in a STAT3-dependent manner. Finally, we show that E2 also activates and uses AMP-activated protein kinase in INS-1 cells to suppress SREBP1c protein expression. This study identifies extranuclear ER pathways involving STAT3 and AMP-activated protein kinase in the genetic control of lipogenesis with therapeutic implications to protect β-cells in type 2 diabetes.
Collapse
Affiliation(s)
- Joseph P Tiano
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, Illinois 60611, USA
| | | |
Collapse
|
33
|
Elevated expression of liver X receptor alpha (LXRα) in myocardium of streptozotocin-induced diabetic rats. Inflammation 2012; 34:698-706. [PMID: 21136146 DOI: 10.1007/s10753-010-9281-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The present study was designed to investigate the myocardial expression of liver X receptor alpha (LXRα) in a streptozotocin (STZ)-induced diabetic rat model. Immunohistochemical staining, quantitative real-time RT-PCR, and Western blot analysis were used to determine the expression of LXRα in the myocardium of STZ-induced diabetic rats. The myocardial expression of LXRα target genes, long-chain acyl-CoA synthetase 3 (ACSL3), fatty acid transporter protein (FAT/CD36), ATP-binding cassette transporter A1 (ABCA1), and ABCG1 were also detected. Bisulfite sequencing analysis was employed to examine the methylation status of the CpG island at the LXRα promoter region in the myocardium of STZ-induced diabetic rats. We found that LXRα mRNA and protein expression in the left ventricles, right ventricles, and atria of diabetic rats were gradually increased during the progression of diabetic cardiomyopathy (DCM). The mRNA expression levels of ACSL3 and FAT/CD36 and the protein expression levels of ABCA1 and ABCG1 were also markedly increased in different heart chambers of diabetic rats. Moreover, there was a significant difference in the methylation status of LXRα gene between the ventricles of control and diabetic rats (P < 0.05). Our findings suggest that elevated expression of LXRα may be involved in the progression of DCM, and demethylation of LXRα is likely to be responsible for its increased expression in myocardial tissues.
Collapse
|
34
|
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: 1.8] [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
|
35
|
Lee JW, Choe SS, Jang H, Kim J, Jeong HW, Jo H, Jeong KH, Tadi S, Park MG, Kwak TH, Man Kim J, Hyun DH, Kim JB. AMPK activation with glabridin ameliorates adiposity and lipid dysregulation in obesity. J Lipid Res 2012; 53:1277-86. [PMID: 22493094 DOI: 10.1194/jlr.m022897] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we demonstrate that activation of AMP-activated protein kinase (AMPK) with glabridin alleviates adiposity and hyperlipidemia in obesity. In several obese rodent models, glabridin decreased body weight and adiposity with a concomitant reduction in fat cell size. Further, glabridin ameliorated fatty liver and plasma levels of triglyceride and cholesterol. In accordance with these findings, glabridin suppressed the expression of lipogenic genes such as sterol regulatory element binding transcription factor (SREBP)-1c, fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and stearoyl-CoA desaturase (SCD)-1 in white adipose tissues and liver, whereas it elevated the expression of fatty acid oxidation genes such as carnitine palmitoyl transferase (CPT)1, acyl-CoA oxidase (ACO), and peroxisome proliferator-activated receptor (PPAR)α in muscle. Moreover, glabridin enhanced phosphorylation of AMPK in muscle and liver and promoted fatty acid oxidation by modulating mitochondrial activity. Together, these data suggest that glabridin is a novel AMPK activator that would exert therapeutic effects in obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Joo-Won Lee
- Department of Biophysics and Chemical Biology, School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Pehkonen P, Welter-Stahl L, Diwo J, Ryynänen J, Wienecke-Baldacchino A, Heikkinen S, Treuter E, Steffensen KR, Carlberg C. Genome-wide landscape of liver X receptor chromatin binding and gene regulation in human macrophages. BMC Genomics 2012; 13:50. [PMID: 22292898 PMCID: PMC3295715 DOI: 10.1186/1471-2164-13-50] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 01/31/2012] [Indexed: 12/15/2022] Open
Abstract
Background The liver X receptors (LXRs) are oxysterol sensing nuclear receptors with multiple effects on metabolism and immune cells. However, the complete genome-wide cistrome of LXR in cells of human origin has not yet been provided. Results We performed ChIP-seq in phorbol myristate acetate-differentiated THP-1 cells (macrophage-type) after stimulation with the potent synthetic LXR ligand T0901317 (T09). Microarray gene expression analysis was performed in the same cellular model. We identified 1357 genome-wide LXR locations (FDR < 1%), of which 526 were observed after T09 treatment. De novo analysis of LXR binding sequences identified a DR4-type element as the major motif. On mRNA level T09 up-regulated 1258 genes and repressed 455 genes. Our results show that LXR actions are focused on 112 genomic regions that contain up to 11 T09 target genes per region under the control of highly stringent LXR binding sites with individual constellations for each region. We could confirm that LXR controls lipid metabolism and transport and observed a strong association with apoptosis-related functions. Conclusions This first report on genome-wide binding of LXR in a human cell line provides new insights into the transcriptional network of LXR and its target genes with their link to physiological processes, such as apoptosis. The gene expression microarray and sequence data have been submitted collectively to the NCBI Gene Expression Omnibus http://www.ncbi.nlm.nih.gov/geo under accession number GSE28319.
Collapse
Affiliation(s)
- Petri Pehkonen
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FIN-70210 Kuopio, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Méndez-González J, Julve J, Rotllan N, Llaverias G, Blanco-Vaca F, Escolà-Gil JC. ATP-binding cassette G5/G8 deficiency causes hypertriglyceridemia by affecting multiple metabolic pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:1186-93. [DOI: 10.1016/j.bbalip.2011.07.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 07/23/2011] [Accepted: 07/27/2011] [Indexed: 10/17/2022]
|
38
|
Sukardi H, Zhang X, Lui EY, Ung CY, Mathavan S, Gong Z, Lam SH. Liver X receptor agonist T0901317 induced liver perturbation in zebrafish: histological, gene set enrichment and expression analyses. Biochim Biophys Acta Gen Subj 2011; 1820:33-43. [PMID: 22047996 DOI: 10.1016/j.bbagen.2011.10.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 10/04/2011] [Accepted: 10/16/2011] [Indexed: 01/16/2023]
Abstract
BACKGROUND Liver X receptor (LXR), a ligand-activated transcription factor, regulates important biological processes. It has been associated with pathology and proposed as a therapeutic target. The zebrafish is a new vertebrate model for disease modeling, drug and toxicity screening and will be interesting to test for its potential for LXR-related studies. METHODS Adult male fish were exposed to LXR agonist T0901317 at 20, 200 and 2000nM for 96h and the livers were sampled for histological, microarray and qRT-PCR analyses. RESULTS Histological analysis suggests dose-dependent perturbation of carbohydrate and lipid metabolisms by T0901317 in the liver, which lead to hepatocyte swelling and cell death. Microarray data revealed several conserved effects of T0901317 with mammalian models, including up-regulation of LXR-targeted genes, modulation of biological pathways associated with proteasome, cell death, extracellular matrix and adhesions, maturity onset diabetes of the young and lipid beta oxidation. Interestingly, this study identified the complement and coagulation systems as down-regulated by T0901317 for the first time, potentially via transcriptional repression by LXR activation. qRT-PCR validated the expression of 16 representative genes, confirming activation of LXR signaling and down-regulation of these biological pathways by T0901317 which could be linked to the anti-thrombogenic, anti-atherogenic and anti-inflammatory actions, as well as metabolic disruptions via LXR activation. CONCLUSION AND GENERAL SIGNIFICANCE Our study underscores the potential of using zebrafish model coupled with transcriptomic analysis to capture pharmacological and toxicological or pathological events induced by LXR modulators.
Collapse
Affiliation(s)
- Hendrian Sukardi
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
39
|
Bełtowski J. Inhibition of cell proliferation: a new role of liver X receptors. ACTA ACUST UNITED AC 2011. [DOI: 10.2217/clp.11.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
40
|
Choi SE, Jung IR, Lee YJ, Lee SJ, Lee JH, Kim Y, Jun HS, Lee KW, Park CB, Kang Y. Stimulation of lipogenesis as well as fatty acid oxidation protects against palmitate-induced INS-1 beta-cell death. Endocrinology 2011; 152:816-27. [PMID: 21209018 DOI: 10.1210/en.2010-0924] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Saturated fatty acids are generally cytotoxic to β-cells. Accumulation of lipid intermediates and subsequent activation of lipid-mediated signals has been suggested to play a role in fatty acid-induced toxicity. To determine the effects of lipid metabolism in fatty acid-induced toxicity, lipid metabolism was modulated by up- and down-regulation of a lipogenic or fatty acid oxidation pathway, and the effects of various modulators on palmitate (PA)-induced INS-1 β-cell death were then evaluated. Treatment with the liver X receptor agonist T0901317 reduced PA-induced INS-1 cell death, regardless of its enhanced lipogenic activity. Furthermore, transient expression of a lipogenic transcription factor sterol regulatory element binding protein-1c (SREBP-1c) was also protective against PA-induced cytotoxicity. In contrast, knockdown of SREBP-1c or glycerol-3-phosphate acyltransferase 1 significantly augmented PA-induced cell death and reduced T0901317-induced protective effects. Conversely, T0901317 increased carnitine PA transferease-1 (CPT-1) expression and augmented PA oxidation. CPT-1 inhibitor etomoxir or CPT-1 knockdown augmented PA-induced cell death and reduced T0901317-induced protective effects, whereas the peroxisome proliferator-activated receptor (PPAR)-α agonist bezafibrate reduced PA-induced toxicity. In particular, T0901317 reduced the levels of PA-induced endoplasmic reticulum (ER) stress markers, including phospho-eukaryotic initiation factor-2α, phospho-C-Jun N terminal kinase, and CCAAT/enhancer-binding protein homologous protein. In contrast, knockdown of SREBP-1c or glycerol-3-phosphate acyltransferase 1 augmented PA-induced ER stress responses. Results of these experiments suggested that stimulation of lipid metabolism, including lipogenesis and fatty acid oxidation, protected β-cells from PA-induced lipotoxicity and that protection through enhanced lipogenesis was likely due to reduced ER stress.
Collapse
Affiliation(s)
- Sung-E Choi
- Institute for Medical Science, Ajou University School of Medicine, Wonchon-dong san 5, Yeongtong-gu, Suwon, Gyeonggi-do 442-749, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kim YW, Kim YM, Yang YM, Kay HY, Kim WD, Lee JW, Hwang SJ, Kim SG. Inhibition of LXRα-dependent steatosis and oxidative injury by liquiritigenin, a licorice flavonoid, as mediated with Nrf2 activation. Antioxid Redox Signal 2011; 14:733-45. [PMID: 20677908 DOI: 10.1089/ars.2010.3260] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Liver X receptor-α (LXRα) functions as a major regulator of lipid homeostasis through activation of sterol regulatory element binding protein-1c (SREBP-1c), which promotes hepatic steatosis and steatohepatitis. NF-E2-related factor 2 (Nrf2) is the crucial transcription factor that is necessary for the induction of antioxidant enzymes. This study investigated the potential of liquiritigenin (LQ), a hepatoprotective flavonoid in licorice, to inhibit LXRα-induced hepatic steatosis, and the underlying mechanism of the action. LQ treatment attenuated fat accumulation and lipogenic gene induction in the liver of mice fed a high fat diet. Also, LQ had the ability to inhibit oxidative liver injury, as shown by decreases in thiobarbituric acid reactive substances formation and nitrotyrosinylation. Moreover, LQ treatment antagonized LXRα agonist (T0901317)-mediated SREBP-1c activation, and transactivation of the lipogenic target genes. LQ was found to activate Nrf2, and the ability of LQ to inhibit LXRα-mediated SREBP-1c activation was reversed by Nrf2 deficiency, which supports the inhibitory role of Nrf2 in LXRα-dependent lipogenesis. Consistently, treatment with other Nrf2 activators or forced expression of Nrf2 also inhibited LXRα-mediated SREBP-1c activation. Our results demonstrate that LQ has an efficacy to activate Nrf2, which contributes to inhibiting the activity of LXRα that leads to SREBP-1c induction and hepatic steatosis.
Collapse
Affiliation(s)
- Young Woo Kim
- Innovative Drug Research Center for Metabolic and Inflammatory Diseases, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Lee JW, Choi AH, Ham M, Kim JW, Choe SS, Park J, Lee GY, Yoon KH, Kim JB. G6PD up-regulation promotes pancreatic beta-cell dysfunction. Endocrinology 2011; 152:793-803. [PMID: 21248143 DOI: 10.1210/en.2010-0606] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Increased reactive oxygen species (ROS) induce pancreatic β-cell dysfunction during progressive type 2 diabetes. Glucose-6-phosphate dehydrogenase (G6PD) is a reduced nicotinamide adenine dinucleotide phosphate-producing enzyme that plays a key role in cellular reduction/oxidation regulation. We have investigated whether variations in G6PD contribute to β-cell dysfunction through regulation of ROS accumulation and β-cell gene expression. When the level of G6PD expression in pancreatic islets was examined in several diabetic animal models, such as db/db mice and OLEFT rats, G6PD expression was evidently up-regulated in pancreatic islets in diabetic animals. To investigate the effect of G6PD on β-cell dysfunction, we assessed the levels of cellular ROS, glucose-stimulated insulin secretion and β-cell apoptosis in G6PD-overexpressing pancreatic β-cells. In INS-1 cells, G6PD overexpression augmented ROS accumulation associated with increased expression of prooxidative enzymes, such as inducible nitric oxide synthase and reduced nicotinamide adenine dinucleotide phosphate oxidase. G6PD up-regulation also caused decrease in glucose-stimulated insulin secretion in INS-1 cells and primary pancreatic islets. Moreover, elevated G6PD expression led to β-cell apoptosis, concomitant with the increase in proapoptotic gene expression. On the contrary, suppression of G6PD with small interference RNA attenuated palmitate-induced β-cell apoptosis. Together, these data suggest that up-regulation of G6PD in pancreatic β-cells would induce β-cell dysregulation through ROS accumulation in the development of type 2 diabetes.
Collapse
Affiliation(s)
- Joo-Won Lee
- Department of Biological Sciences, Seoul National University, San 56-1, Sillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Lim S, Rashid MA, Jang M, Kim Y, Won H, Lee J, Woo JT, Kim YS, Murphy MP, Ali L, Ha J, Kim SS. Mitochondria-targeted Antioxidants Protect Pancreatic β-cells against Oxidative Stress and Improve Insulin Secretion in Glucotoxicity and Glucolipotoxicity. Cell Physiol Biochem 2011; 28:873-86. [DOI: 10.1159/000335802] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2011] [Indexed: 01/11/2023] Open
|
44
|
Rashid MA, Lee S, Tak E, Lee J, Choi TG, Lee JW, Kim JB, Youn JH, Kang I, Ha J, Kim SS. Carbonyl reductase 1 protects pancreatic β-cells against oxidative stress-induced apoptosis in glucotoxicity and glucolipotoxicity. Free Radic Biol Med 2010; 49:1522-33. [PMID: 20728534 DOI: 10.1016/j.freeradbiomed.2010.08.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 08/05/2010] [Accepted: 08/12/2010] [Indexed: 01/02/2023]
Abstract
Carbonyl reductase 1 (CBR1) plays an important role in the detoxification of reactive lipid aldehydes. Oxidative stress has been implicated in the pathogenesis of pancreatic β-cell failure. However, the functional role of CBR1 in pancreatic β-cell failure has not been studied yet. Therefore, we investigated the role of CBR1 in pancreatic β-cell failure under glucotoxic and glucolipotoxic conditions. Under both conditions, knockdown of CBR1 by specific siRNA increased β-cell apoptosis, expression of lipogenic enzymes (such as ACC, FAS, and ABCA1), intracellular lipid accumulation, oxidative stress, ER stress, and nuclear SREBP1c, but decreased glucose-stimulated insulin secretion. In contrast, overexpression of CBR1 showed the opposite effects. The antioxidants N-acetyl-l-cysteine and Tiron, as well as the FAS inhibitor cerulenin, reversed the effects of CBR1 knockdown. Interestingly, the expression level and enzyme activity of CBR1 were significantly decreased in pancreatic islets of db/db mice, compared with those of wild-type mice. In conclusion, CBR1 protects pancreatic β-cells against oxidative stress and promotes their survival in glucotoxicity and glucolipotoxicity.
Collapse
Affiliation(s)
- M A Rashid
- Medical Science and Engineering Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute (BK-21), Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ketterer C, Müssig K, Machicao F, Stefan N, Fritsche A, Häring HU, Staiger H. Genetic variation within the NR1H2 gene encoding liver X receptor β associates with insulin secretion in subjects at increased risk for type 2 diabetes. J Mol Med (Berl) 2010; 89:75-81. [PMID: 21042792 DOI: 10.1007/s00109-010-0687-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 08/23/2010] [Accepted: 09/08/2010] [Indexed: 01/17/2023]
Abstract
The liver X receptors (LXRs)-α and -β play a crucial role in control of insulin production and secretion in pancreatic β-cells. We hypothesized that common variants in the NR1H2 and NR1H3 genes, encoding LXR-β and -α, respectively, may alter pancreatic β-cell function. One thousand five hundred seventy-four subjects of European ancestry with elevated risk for type 2 diabetes were genotyped for the two NR1H2 single nucleotide polymorphisms (SNPs) rs2248949 and rs1405655 and for the four NR1H3 SNPs rs11039149, rs3758673, rs12221497 and rs2279238, and association studies with metabolic traits were performed. Metabolic characterization comprised an oral glucose tolerance test (OGTT) in all participants and, in addition, a hyperinsulinemic-euglycemic clamp and an intravenous glucose tolerance test (IVGTT) in subsets. One hundred per cent of common genetic variation (minor allele frequency ≥1%) within the NR1H2 and NR1H3 loci (D' = 1.0; r² ≥ 0.8) were covered by the six chosen tagging SNPs. NR1H2 rs2248949 was nominally associated with OGTT-derived first-phase insulin secretion and proinsulin conversion to insulin and significantly associated with the AUC of insulin levels during the IVGTT (p = 0.007) after adjustment for age, gender, BMI and insulin sensitivity in the dominant model, with the minor allele conferring reduced pancreatic β-cell function to the carriers. In subjects of European ancestry at increased risk for type 2 diabetes, common variation within the NR1H2 gene impaired insulin secretion, which may facilitate the development of type 2 diabetes.
Collapse
Affiliation(s)
- Caroline Ketterer
- Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Department of Internal Medicine, Eberhard Karls University Tübingen, German Center for Diabetes Research (DZD), Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Miyazaki S, Taniguchi H, Moritoh Y, Tashiro F, Yamamoto T, Yamato E, Ikegami H, Ozato K, Miyazaki JI. Nuclear hormone retinoid X receptor (RXR) negatively regulates the glucose-stimulated insulin secretion of pancreatic ß-cells. Diabetes 2010; 59:2854-61. [PMID: 20798333 PMCID: PMC2963544 DOI: 10.2337/db09-1897] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Retinoid X receptors (RXRs) are members of the nuclear hormone receptor superfamily and are thought to be key regulators in differentiation, cellular growth, and gene expression. Although several experiments using pancreatic β-cell lines have shown that the ligands of nuclear hormone receptors modulate insulin secretion, it is not clear whether RXRs have any role in insulin secretion. RESEARCH DESIGN AND METHODS To elucidate the function of RXRs in pancreatic β-cells, we generated a double-transgenic mouse in which a dominant-negative form of RXRβ was inducibly expressed in pancreatic β-cells using the Tet-On system. We also established a pancreatic β-cell line from an insulinoma caused by the β-cell-specific expression of simian virus 40 T antigen in the above transgenic mouse. RESULTS In the transgenic mouse, expression of the dominant-negative RXR enhanced the insulin secretion with high glucose stimulation. In the pancreatic β-cell line, the suppression of RXRs also enhanced glucose-stimulated insulin secretion at a high glucose concentration, while 9-cis-retinoic acid, an RXR agonist, repressed it. High-density oligonucleotide microarray analysis showed that expression of the dominant-negative RXR affected the expression levels of a number of genes, some of which have been implicated in the function and/or differentiation of β-cells. CONCLUSIONS These results suggest that endogenous RXR negatively regulates the glucose-stimulated insulin secretion. Given these findings, we propose that the modulation of endogenous RXR in β-cells may be a new therapeutic approach for improving impaired insulin secretion in type 2 diabetes.
Collapse
Affiliation(s)
- Satsuki Miyazaki
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidenori Taniguchi
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Moritoh
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumi Tashiro
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tsunehiko Yamamoto
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eiji Yamato
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Ikegami
- Department of Endocrinology, Metabolism and Diabetes, Kinki University School of Medicine, Osaka, Japan
| | - Keiko Ozato
- Section on Molecular Genetics of Immunity, Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Jun-ichi Miyazaki
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Osaka, Japan
- Corresponding author: Jun-ichi Miyazaki,
| |
Collapse
|
47
|
Choi SE, Lee YJ, Hwang GS, Chung JH, Lee SJ, Lee JH, Han SJ, Kim HJ, Lee KW, Kim Y, Jun HS, Kang Y. Supplement of TCA cycle intermediates protects against high glucose/palmitate-induced INS-1 beta cell death. Arch Biochem Biophys 2010; 505:231-41. [PMID: 20965146 DOI: 10.1016/j.abb.2010.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/12/2010] [Accepted: 10/14/2010] [Indexed: 01/22/2023]
Abstract
The aim of this study is to investigate the effect of mitochondrial metabolism on high glucose/palmitate (HG/PA)-induced INS-1 beta cell death. Long-term treatment of INS-1 cells with HG/PA impaired energy-producing metabolism accompanying with depletion of TCA cycle intermediates. Whereas an inhibitor of carnitine palmitoyl transferase 1 augmented HG/PA-induced INS-1 cell death, stimulators of fatty acid oxidation protected the cells against the HG/PA-induced death. Furthermore, whereas mitochondrial pyruvate carboxylase inhibitor phenylacetic acid augmented HG/PA-induced INS-1 cell death, supplementation of TCA cycle metabolites including leucine/glutamine, methyl succinate/α-ketoisocaproic acid, dimethyl malate, and valeric acid or treatment with a glutamate dehydrogenase activator, aminobicyclo-heptane-2-carboxylic acid (BCH), significantly protected the cells against the HG/PA-induced death. In particular, the mitochondrial tricarboxylate carrier inhibitor, benzene tricarboxylate (BTA), also showed a strong protective effect on the HG/PA-induced INS-1 cell death. Knockdown of glutamate dehydrogenase or tricarboxylate carrier augmented or reduced the HG/PA-induced INS-1 cell death, respectively. Both BCH and BTA restored HG/PA-induced reduction of energy metabolism as well as depletion of TCA intermediates. These data suggest that depletion of the TCA cycle intermediate pool and impaired energy-producing metabolism may play a role in HG/PA-induced cytotoxicity to beta cells and thus, HG/PA-induced beta cell glucolipotoxicity can be protected by nutritional or pharmacological maneuver enhancing anaplerosis or reducing cataplerosis.
Collapse
Affiliation(s)
- Sung-E Choi
- Institute for Medical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Solaas K, Legry V, Retterstol K, Berg PR, Holven KB, Ferrières J, Amouyel P, Lien S, Romeo J, Valtueña J, Widhalm K, Ruiz JR, Dallongeville J, Tonstad S, Rootwelt H, Halvorsen B, Nenseter MS, Birkeland KI, Thorsby PM, Meirhaeghe A, Nebb HI. Suggestive evidence of associations between liver X receptor β polymorphisms with type 2 diabetes mellitus and obesity in three cohort studies: HUNT2 (Norway), MONICA (France) and HELENA (Europe). BMC MEDICAL GENETICS 2010; 11:144. [PMID: 20939869 PMCID: PMC2958901 DOI: 10.1186/1471-2350-11-144] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 10/12/2010] [Indexed: 01/03/2023]
Abstract
BACKGROUND The liver X receptors (LXR) α and β regulate lipid and carbohydrate homeostasis and inflammation. Lxrβ⁻/⁻ mice are glucose intolerant and at the same time lean. We aimed to assess the associations between single nucleotide polymorphisms (SNPs) in LXRβ and risk of type 2 diabetes mellitus (T2DM), obesity and related traits in 3 separate cohort studies. METHODS Twenty LXRβ SNPs were identified by sequencing and genotyped in the HUNT2 adult nested case-control study for T2DM (n = 835 cases/1986 controls). Five tag-SNPs (rs17373080, rs2695121, rs56151148, rs2303044 and rs3219281), covering 99.3% of the entire common genetic variability of the LXRβ gene were identified and genotyped in the French MONICA adult study (n = 2318) and the European adolescent HELENA cross-sectional study (n = 1144). In silico and in vitro functionality studies were performed. RESULTS We identified suggestive or significant associations between rs17373080 and the risk of (i) T2DM in HUNT2 (OR = 0.82, p = 0.03), (ii) obesity in MONICA (OR = 1.26, p = 0.05) and (iii) overweight/obesity in HELENA (OR = 1.59, p = 0.002). An intron 4 SNP (rs28514894, a perfect proxy for rs17373080) could potentially create binding sites for hepatic nuclear factor 4 alpha (HNF4α) and nuclear factor 1 (NF1). The C allele of rs28514894 was associated with ~1.25-fold higher human LXRβ basal promoter activity in vitro. However, no differences between alleles in terms of DNA binding and reporter gene transactivation by HNF4α or NF1 were observed. CONCLUSIONS Our results suggest that rs17373080 in LXRβ is associated with T2DM and obesity, maybe via altered LXRβ expression.
Collapse
Affiliation(s)
- Karianne Solaas
- Department of Endocrinology, Oslo University Hospital, Aker, Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kim KH, Lee GY, Kim JI, Ham M, Won Lee J, Kim JB. Inhibitory effect of LXR activation on cell proliferation and cell cycle progression through lipogenic activity. J Lipid Res 2010; 51:3425-33. [PMID: 20847297 DOI: 10.1194/jlr.m007989] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Liver X receptor (LXR), a sterol-activated nuclear hormone receptor, has been implicated in cholesterol and fatty acid homeostasis via regulation of reverse cholesterol transport and de novo fatty acid synthesis. LXR is also involved in immune responses, including anti-inflammatory action and T cell proliferation. In this study, we demonstrated that activated LXR suppresses cell cycle progression and proliferation in certain cell types. Stimulation of LXR with synthetic ligand T0901317 or GW3965 inhibited cell growth rate and arrested the cell cycle at the G1/S boundary in several cells, such as RWPE1, THP1, SNU16, LNCaP, and HepG2. However, LXR ligands did not exhibit antiproliferative activity in PC3, HEK293, or HeLa cells. Interestingly, activated LXR-mediated cell cycle arrest is closely correlated with the lipogenic gene expression and triacylglyceride accumulation. In accordance with these findings, suppression of FAS via small-interference RNA (siRNA) partially alleviated the antiproliferative effect of LXR activation in RWPE1 cells. Together, these data suggest that LXR activation with its ligands inhibits cell proliferation and induces G1/S arrest through elevated lipogenic activity, thus proposing a novel effect of activated LXR on cell cycle regulation.
Collapse
Affiliation(s)
- Kang Ho Kim
- Department of Biophysics and Chemical Biology, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
50
|
Weedon-Fekjær M, Johnsen G, Anthonisen E, Sugulle M, Nebb H, Duttaroy A, Staff A. Expression of Liver X Receptors in Pregnancies Complicated by Preeclampsia. Placenta 2010; 31:818-24. [DOI: 10.1016/j.placenta.2010.06.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 05/27/2010] [Accepted: 06/24/2010] [Indexed: 10/19/2022]
|