1
|
Deshwal S, Baidya AT, Kumar R, Sandhir R. Structure-based virtual screening for identification of potential non-steroidal LXR modulators against neurodegenerative conditions. J Steroid Biochem Mol Biol 2022; 223:106150. [PMID: 35787453 DOI: 10.1016/j.jsbmb.2022.106150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
Abstract
Liver X Receptors (LXRs) are members of the nuclear receptor superfamily that regulate cholesterol metabolism. LXRs have been suggested as promising targets against many neurodegenerative diseases (NDDs). The present study was aimed to identify novel non-steroidal molecules that may potentially modulate LXR activity. The structure-based virtual screening (SBVS) was used to search for suitable compounds from the Asinex library. The top hits were selected and filtered based on their binding affinity for LXR α and β isoforms. Based on molecular docking and scoring results, 24 compounds were selected that had binding energy in the range of - 13.9 to - 12 for LXRα and - 12.5 to - 11 for LXRβ, which were higher than the reference ligands (GW3965 and TO901317). Further, the five hits referred to as model 29, 64, 202, 250, 313 were selected by virtue of their binding interactions with amino acid residues at the active site of LXRs. The selected hits were then subjected to absorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis and blood-brain permeability prediction. It was observed that the selected hits had better pharmacokinetic properties with no toxicity and could cross blood-brain barrier. Further, the selected hits were analysed for dynamic evolution of the system with LXRs by molecular dynamics (MD) simulation at 100 ns using GROMACS. The MD simulation results validated that selected hits possess a remarkable amount of flexibility, stability, compactness, binding energy and exhibited limited conformational modification. The root mean square deviation (RMSD) values of the top-scoring hits complexed with LXRα and LXRβ were 0.05-0.6 nm and 0.05-0.45 nm respectively, which is greater than the protein itself. Altogether the study identified potential non-steroidal LXR modulators that appear to be effective against various neurodegenerative conditions involving perturbed cholesterol and lipid homeostasis.
Collapse
Affiliation(s)
- Sonam Deshwal
- Department of Biochemistry, Basic Medical Sciences, Block-II, Panjab University, Chandigarh 160014, India
| | - Anurag Tk Baidya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Rajat Sandhir
- Department of Biochemistry, Basic Medical Sciences, Block-II, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
2
|
Moldavski O, Zushin PJH, Berdan CA, Van Eijkeren RJ, Jiang X, Qian M, Ory DS, Covey DF, Nomura DK, Stahl A, Weiss EJ, Zoncu R. 4β-Hydroxycholesterol is a prolipogenic factor that promotes SREBP1c expression and activity through the liver X receptor. J Lipid Res 2021; 62:100051. [PMID: 33631213 PMCID: PMC8042401 DOI: 10.1016/j.jlr.2021.100051] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/06/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
Oxysterols are oxidized derivatives of cholesterol that play regulatory roles in lipid biosynthesis and homeostasis. How oxysterol signaling coordinates different lipid classes such as sterols and triglycerides remains incompletely understood. Here, we show that 4β-hydroxycholesterol (HC) (4β-HC), a liver and serum abundant oxysterol of poorly defined functions, is a potent and selective inducer of the master lipogenic transcription factor, SREBP1c, but not the related steroidogenic transcription factor SREBP2. By correlating tracing of lipid synthesis with lipogenic gene expression profiling, we found that 4β-HC acts as a putative agonist for the liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of the LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c and triggered de novo lipogenesis both in primary hepatocytes and in the mouse liver. In addition, 4β-HC acted in parallel to insulin-PI3K-dependent signaling to stimulate triglyceride synthesis and lipid-droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
Collapse
Affiliation(s)
- Ofer Moldavski
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA; Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Charles A Berdan
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Robert J Van Eijkeren
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA
| | - Xuntian Jiang
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St Louis, MO, USA
| | - Mingxing Qian
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St Louis, MO, USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel K Nomura
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Ethan J Weiss
- Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
3
|
Buñay J, Fouache A, Trousson A, de Joussineau C, Bouchareb E, Zhu Z, Kocer A, Morel L, Baron S, Lobaccaro JMA. Screening for liver X receptor modulators: Where are we and for what use? Br J Pharmacol 2020; 178:3277-3293. [PMID: 33080050 DOI: 10.1111/bph.15286] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022] Open
Abstract
Liver X receptors (LXRs) are members of the nuclear receptor superfamily that are canonically activated by oxidized derivatives of cholesterol. Since the mid-90s, numerous groups have identified LXRs as endocrine receptors that are involved in the regulation of various physiological functions. As a result, when their expression is genetically modified in mice, phenotypic analyses reveal endocrine disorders ranging from infertility to diabetes and obesity, nervous system pathologies such Alzheimer's or Parkinson's disease, immunological disturbances, inflammatory response, and enhancement of tumour development. Based on such findings, it appears that LXRs could constitute good pharmacological targets to prevent and/or to treat these diseases. This review discusses the various aspects of LXR drug discovery, from the tools available for the screening of potential LXR modulators to the current situational analysis of the drugs in development. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Julio Buñay
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Allan Fouache
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Amalia Trousson
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Cyrille de Joussineau
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Erwan Bouchareb
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Zhekun Zhu
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Ayhan Kocer
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Laurent Morel
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Silvere Baron
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Jean-Marc A Lobaccaro
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
4
|
Balasubramanian B, Kim HJ, Mothana RA, Kim YO, Siddiqui NA. Role of LXR alpha in regulating expression of glucose transporter 4 in adipocytes — Investigation on improvement of health of diabetic patients. J Infect Public Health 2020; 13:244-252. [DOI: 10.1016/j.jiph.2019.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 11/26/2022] Open
|
5
|
El-Gendy BEDM, Goher SS, Hegazy LS, Arief MMH, Burris TP. Recent Advances in the Medicinal Chemistry of Liver X Receptors. J Med Chem 2018; 61:10935-10956. [DOI: 10.1021/acs.jmedchem.8b00045] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Bahaa El-Dien M. El-Gendy
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Shaimaa S. Goher
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Lamees S. Hegazy
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Mohamed M. H. Arief
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Thomas P. Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| |
Collapse
|
6
|
Jennelle LT, Dandekar AP, Magoro T, Hahn YS. Immunometabolic Signaling Pathways Contribute to Macrophage and Dendritic Cell Function. Crit Rev Immunol 2018; 36:379-394. [PMID: 28605345 DOI: 10.1615/critrevimmunol.2017018803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Understanding of antigen-presenting cell (APC) participation in tissue inflammation and metabolism has advanced through numerous studies using systems biology approaches. Previously unrecognized connections between these research areas have been elucidated in the context of inflammatory disease involving innate and adaptive immune responses. A new conceptual framework bridges APC biology, metabolism, and cytokines in the generation of effective T-cell responses. Exploring these connections is paramount to addressing the rising tide of multi-organ system diseases, particularly chronic diseases associated with metabolic syndrome, infection, and cancer. Focused research in these areas will aid the development of strategies to harness and manipulate innate immunology to improve vaccine development, anti-viral, anti-inflammatory, and anti-tumor therapies. This review highlights recent advances in APC "immunometabolism" specifically related to chronic viral and metabolic disease in humans. The goal of this review is to develop an abridged and consolidated outlook on recent thematic updates to APC immunometabolism in the areas of regulation and crosstalk between metabolic and inflammatory signaling and the integrated stress response and how these signals dictate APC function in providing T-cell activation Signal 3.
Collapse
Affiliation(s)
- Lucas T Jennelle
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | - Aditya P Dandekar
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | - Tshifhiwa Magoro
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | - Young S Hahn
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
7
|
Takeuchi Y, Yahagi N, Aita Y, Murayama Y, Sawada Y, Piao X, Toya N, Oya Y, Shikama A, Takarada A, Masuda Y, Nishi M, Kubota M, Izumida Y, Yamamoto T, Sekiya M, Matsuzaka T, Nakagawa Y, Urayama O, Kawakami Y, Iizuka Y, Gotoda T, Itaka K, Kataoka K, Nagai R, Kadowaki T, Yamada N, Lu Y, Jain MK, Shimano H. KLF15 Enables Rapid Switching between Lipogenesis and Gluconeogenesis during Fasting. Cell Rep 2016; 16:2373-86. [PMID: 27545894 PMCID: PMC5031553 DOI: 10.1016/j.celrep.2016.07.069] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/08/2016] [Accepted: 07/25/2016] [Indexed: 11/17/2022] Open
Abstract
Hepatic lipogenesis is nutritionally regulated (i.e., downregulated during fasting and upregulated during the postprandial state) as an adaptation to the nutritional environment. While alterations in the expression level of the transcription factor SREBP-1c are known to be critical for nutritionally regulated lipogenesis, upstream mechanisms governing Srebf1 expression remain unclear. Here, we show that the fasting-induced transcription factor KLF15, a key regulator of gluconeogenesis, forms a complex with LXR/RXR, specifically on the Srebf1 promoter. This complex recruits the corepressor RIP140 instead of the coactivator SRC1, resulting in reduced Srebf1 and thus downstream lipogenic enzyme expression during the early and euglycemic period of fasting prior to hypoglycemia and PKA activation. Through this mechanism, KLF15 overexpression specifically ameliorates hypertriglyceridemia without affecting LXR-mediated cholesterol metabolism. These findings reveal a key molecular link between glucose and lipid metabolism and have therapeutic implications for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Yoshinori Takeuchi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Naoya Yahagi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan.
| | - Yuichi Aita
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Murayama
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshikazu Sawada
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Xiaoying Piao
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Naoki Toya
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yukari Oya
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Akito Shikama
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Ayako Takarada
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yukari Masuda
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Makiko Nishi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Midori Kubota
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshihiko Izumida
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Yamamoto
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshimi Nakagawa
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Osamu Urayama
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yasushi Kawakami
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yoko Iizuka
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Takanari Gotoda
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Keiji Itaka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Ryozo Nagai
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Takashi Kadowaki
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Nobuhiro Yamada
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yuan Lu
- Case Cardiovascular Research Institute, Cleveland, OH 44106, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Cleveland, OH 44106, USA
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
8
|
Olivares AM, Moreno-Ramos OA, Haider NB. Role of Nuclear Receptors in Central Nervous System Development and Associated Diseases. J Exp Neurosci 2016; 9:93-121. [PMID: 27168725 PMCID: PMC4859451 DOI: 10.4137/jen.s25480] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 11/13/2022] Open
Abstract
The nuclear hormone receptor (NHR) superfamily is composed of a wide range of receptors involved in a myriad of important biological processes, including development, growth, metabolism, and maintenance. Regulation of such wide variety of functions requires a complex system of gene regulation that includes interaction with transcription factors, chromatin-modifying complex, and the proper recognition of ligands. NHRs are able to coordinate the expression of genes in numerous pathways simultaneously. This review focuses on the role of nuclear receptors in the central nervous system and, in particular, their role in regulating the proper development and function of the brain and the eye. In addition, the review highlights the impact of mutations in NHRs on a spectrum of human diseases from autism to retinal degeneration.
Collapse
Affiliation(s)
- Ana Maria Olivares
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Oscar Andrés Moreno-Ramos
- Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Neena B Haider
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
LXRα represses LPS-induced inflammatory responses by competing with IRF3 for GRIP1 in Kupffer cells. Int Immunopharmacol 2016; 35:272-279. [PMID: 27085678 DOI: 10.1016/j.intimp.2016.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 04/03/2016] [Accepted: 04/06/2016] [Indexed: 12/16/2022]
Abstract
Liver X receptors (LXRs) in the nucleus play important roles in lipid metabolism and inflammation. The mechanism of LXR regulation of the LPS-induced Toll-like receptor 4 (TLR4) inflammatory signaling pathway remains to be elucidated. C57/BL6 mice were randomly divided into four groups: control, T0901317 (a LXRs agonist), LPS and T0901317+LPS. Additionally, Kupffer cells isolated from male C57/BL6 mice were divided into the same four groups. A decreased amount of inflammatory cells infiltrated the portal areas and the hepatic sinusoids in the livers of mice in the T0901317+LPS group than in those of mice in the LPS group. In the T0901317+LPS group, the serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and tumor necrosis factor alpha (TNF-α) were lower, while the serum level of interleukin-10 (IL-10) was higher. In vitro, Kupffer cells pretreated with T0901317 for 24h presented reduced TNF-α, interferon-beta (IFN-β) and interleukin-1 beta (IL-1β) levels, while the IL-10 level increased; however, the mRNA and protein expression levels of interferon regulatory factor 3 (IRF3) and glucocorticoid receptor-interacting protein 1 (GRIP1) were not significantly reduced. The co-IP data illustrated that LXRα bound to GRIP1 specifically in the T0901317+LPS group, while less IRF3 was bound to GRIP1 in the T0901317+LPS group than in the LPS group. Furthermore, the DNA-binding activity of NF-κB was decreased by pretreating Kupffer cells with T0901317 for 24h. These results suggest that activated LXRα competes with IRF3 for GRIP1 binding, thus repressing IRF3 and NF-κB transcriptional activity and inhibiting the inflammatory response initiated by LPS in Kupffer cells.
Collapse
|
10
|
Shrestha E, Hussein MA, Savas JN, Ouimet M, Barrett TJ, Leone S, Yates JR, Moore KJ, Fisher EA, Garabedian MJ. Poly(ADP-ribose) Polymerase 1 Represses Liver X Receptor-mediated ABCA1 Expression and Cholesterol Efflux in Macrophages. J Biol Chem 2016; 291:11172-84. [PMID: 27026705 DOI: 10.1074/jbc.m116.726729] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Indexed: 11/06/2022] Open
Abstract
Liver X receptors (LXR) are oxysterol-activated nuclear receptors that play a central role in reverse cholesterol transport through up-regulation of ATP-binding cassette transporters (ABCA1 and ABCG1) that mediate cellular cholesterol efflux. Mouse models of atherosclerosis exhibit reduced atherosclerosis and enhanced regression of established plaques upon LXR activation. However, the coregulatory factors that affect LXR-dependent gene activation in macrophages remain to be elucidated. To identify novel regulators of LXR that modulate its activity, we used affinity purification and mass spectrometry to analyze nuclear LXRα complexes and identified poly(ADP-ribose) polymerase-1 (PARP-1) as an LXR-associated factor. In fact, PARP-1 interacted with both LXRα and LXRβ. Both depletion of PARP-1 and inhibition of PARP-1 activity augmented LXR ligand-induced ABCA1 expression in the RAW 264.7 macrophage line and primary bone marrow-derived macrophages but did not affect LXR-dependent expression of other target genes, ABCG1 and SREBP-1c. Chromatin immunoprecipitation experiments confirmed PARP-1 recruitment at the LXR response element in the promoter of the ABCA1 gene. Further, we demonstrated that LXR is poly(ADP-ribosyl)ated by PARP-1, a potential mechanism by which PARP-1 influences LXR function. Importantly, the PARP inhibitor 3-aminobenzamide enhanced macrophage ABCA1-mediated cholesterol efflux to the lipid-poor apolipoprotein AI. These findings shed light on the important role of PARP-1 on LXR-regulated lipid homeostasis. Understanding the interplay between PARP-1 and LXR may provide insights into developing novel therapeutics for treating atherosclerosis.
Collapse
Affiliation(s)
- Elina Shrestha
- From the Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - Maryem A Hussein
- From the Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - Jeffery N Savas
- the Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611
| | - Mireille Ouimet
- the Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York 10016, and
| | - Tessa J Barrett
- the Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York 10016, and
| | - Sarah Leone
- From the Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - John R Yates
- the Department of Chemical Physiology, Scripps Research Institute, La Jolla, California 92037
| | - Kathryn J Moore
- the Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York 10016, and
| | - Edward A Fisher
- the Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York 10016, and
| | - Michael J Garabedian
- From the Department of Microbiology, New York University School of Medicine, New York, New York 10016,
| |
Collapse
|
11
|
Nelson JK, Cook ECL, Loregger A, Hoeksema MA, Scheij S, Kovacevic I, Hordijk PL, Ovaa H, Zelcer N. Deubiquitylase Inhibition Reveals Liver X Receptor-independent Transcriptional Regulation of the E3 Ubiquitin Ligase IDOL and Lipoprotein Uptake. J Biol Chem 2015; 291:4813-25. [PMID: 26719329 DOI: 10.1074/jbc.m115.698688] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Indexed: 01/05/2023] Open
Abstract
Cholesterol metabolism is subject to complex transcriptional and nontranscriptional regulation. Herein, the role of ubiquitylation is emerging as an important post-translational modification that regulates cholesterol synthesis and uptake. Similar to other post-translational modifications, ubiquitylation is reversible in a process dependent on activity of deubiquitylating enzymes (DUBs). Yet whether these play a role in cholesterol metabolism is largely unknown. As a first step to test this possibility, we used pharmacological inhibition of cellular DUB activity. Short term (2 h) inhibition of DUBs resulted in accumulation of high molecular weight ubiquitylated proteins. This was accompanied by a dramatic decrease in abundance of the LDLR and attenuated LDL uptake into hepatic cells. Importantly, this occurred in the absence of changes in the mRNA levels of the LDLR or other SREBP2-regulated genes, in line with this phenotype being a post-transcriptional event. Mechanistically, we identify transcriptional induction of the E3 ubiquitin ligase IDOL in human and rodent cells as the underlying cause for ubiquitylation-dependent lysosomal degradation of the LDLR following DUB inhibition. In contrast to the established transcriptional regulation of IDOL by the sterol-responsive liver X receptor (LXR) transcription factors, induction of IDOL by DUB inhibition is LXR-independent and occurs in Lxrαβ(-/-) MEFs. Consistent with the role of DUBs in transcriptional regulation, we identified a 70-bp region in the proximal promoter of IDOL, distinct from that containing the LXR-responsive element, which mediates the response to DUB inhibition. In conclusion, we identify a sterol-independent mechanism to regulate IDOL expression and IDOL-mediated lipoprotein receptor degradation.
Collapse
Affiliation(s)
- Jessica Kristine Nelson
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Emma Clare Laura Cook
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Anke Loregger
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Marten Anne Hoeksema
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Saskia Scheij
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Igor Kovacevic
- the Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1066 CX Amsterdam, The Netherlands, and
| | - Peter Lodewijk Hordijk
- the Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1066 CX Amsterdam, The Netherlands, and
| | - Huib Ovaa
- the Department of Cell Biology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Noam Zelcer
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands,
| |
Collapse
|
12
|
Fan H, Dong W, Li Q, Zou X, Zhang Y, Wang J, Li S, Liu W, Dong Y, Sun H, Hou Z. Ajuba Preferentially Binds LXRα/RXRγ Heterodimer to Enhance LXR Target Gene Expression in Liver Cells. Mol Endocrinol 2015; 29:1608-18. [PMID: 26389695 DOI: 10.1210/me.2015-1046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The liver X receptors (LXRs) are important regulators of lipid, cholesterol, and glucose homeostasis by transcriptional regulation of many key genes in these processes, and the transcriptional activities of LXRs are finely controlled by cooperating with retinoid X receptors and many other coregulators. Here, we report that the LIM protein Ajuba binds to the hinge and the ligand binding domains of LXRα via its C-terminal tandem LIM motifs and enhances LXR target gene expression in liver cells. Depletion of Ajuba in HepG2 cells and in mouse primary hepatocytes decreases LXR target gene expression, whereas stable expression of Ajuba in HepG2 cells results in increased expression of these genes. Mechanistic investigations found that Ajuba selectively interacts with LXRα/retinoid X receptor-γ heterodimer to form a ternary complex, which displays a higher transactivation activity to LXR target genes. Moreover, Ajuba and LXR mutually affect their DNA binding activity at endogenous target chromatins and the cooperation between Ajuba and LXRα is dependent on the functional LXR response elements located in the target promoters. Together, our studies demonstrate that Ajuba is a novel coactivator for LXRs and may play important role in lipid and glucose metabolism.
Collapse
Affiliation(s)
- Hongyan Fan
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Weibing Dong
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Qi Li
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Xiuqun Zou
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Yihong Zhang
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Jiamin Wang
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Shengxian Li
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Wei Liu
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Ying Dong
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Haipeng Sun
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Zhaoyuan Hou
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
13
|
Gillespie MA, Gold ES, Ramsey SA, Podolsky I, Aderem A, Ranish JA. An LXR-NCOA5 gene regulatory complex directs inflammatory crosstalk-dependent repression of macrophage cholesterol efflux. EMBO J 2015; 34:1244-58. [PMID: 25755249 DOI: 10.15252/embj.201489819] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 02/13/2015] [Indexed: 02/04/2023] Open
Abstract
LXR-cofactor complexes activate the gene expression program responsible for cholesterol efflux in macrophages. Inflammation antagonizes this program, resulting in foam cell formation and atherosclerosis; however, the molecular mechanisms underlying this antagonism remain to be fully elucidated. We use promoter enrichment-quantitative mass spectrometry (PE-QMS) to characterize the composition of gene regulatory complexes assembled at the promoter of the lipid transporter Abca1 following downregulation of its expression. We identify a subset of proteins that show LXR ligand- and binding-dependent association with the Abca1 promoter and demonstrate they differentially control Abca1 expression. We determine that NCOA5 is linked to inflammatory Toll-like receptor (TLR) signaling and establish that NCOA5 functions as an LXR corepressor to attenuate Abca1 expression. Importantly, TLR3-LXR signal crosstalk promotes recruitment of NCOA5 to the Abca1 promoter together with loss of RNA polymerase II and reduced cholesterol efflux. Together, these data significantly expand our knowledge of regulatory inputs impinging on the Abca1 promoter and indicate a central role for NCOA5 in mediating crosstalk between pro-inflammatory and anti-inflammatory pathways that results in repression of macrophage cholesterol efflux.
Collapse
Affiliation(s)
| | | | - Stephen A Ramsey
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| | | | - Alan Aderem
- Seattle Biomedical Research Institute, Seattle, WA, USA
| | | |
Collapse
|
14
|
Kurakula K, Sommer D, Sokolovic M, Moerland PD, Scheij S, van Loenen PB, Koenis DS, Zelcer N, van Tiel CM, de Vries CJM. LIM-only protein FHL2 is a positive regulator of liver X receptors in smooth muscle cells involved in lipid homeostasis. Mol Cell Biol 2015; 35:52-62. [PMID: 25332231 PMCID: PMC4295390 DOI: 10.1128/mcb.00525-14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/06/2014] [Accepted: 10/07/2014] [Indexed: 11/20/2022] Open
Abstract
The LIM-only protein FHL2 is expressed in smooth muscle cells (SMCs) and inhibits SMC-rich-lesion formation. To further elucidate the role of FHL2 in SMCs, we compared the transcriptomes of SMCs derived from wild-type (WT) and FHL2 knockout (KO) mice. This revealed that in addition to the previously recognized involvement of FHL2 in SMC proliferation, the cholesterol synthesis and liver X receptor (LXR) pathways are altered in the absence of FHL2. Using coimmunoprecipitation experiments, we found that FHL2 interacts with the two LXR isoforms, LXRα and LXRβ. Furthermore, FHL2 strongly enhances transcriptional activity of LXR element (LXRE)-containing reporter constructs. Chromatin immunoprecipitation (ChIP) experiments on the ABCG1 promoter revealed that FHL2 enhances the association of LXRβ with DNA. In line with these observations, we observed reduced basal transcriptional LXR activity in FHL2-KO SMCs compared to WT SMCs. This was also reflected in reduced expression of LXR target genes in intact aorta and aortic SMCs of FHL2-KO mice. Functionally, the absence of FHL2 resulted in attenuated cholesterol efflux to both ApoA-1 and high-density lipoprotein (HDL), in agreement with reduced LXR signaling. Collectively, our findings demonstrate that FHL2 is a transcriptional coactivator of LXRs and points toward FHL2 being an important determinant of cholesterol metabolism in SMCs.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Daniela Sommer
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Milka Sokolovic
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands European Food Information Council, Brussels, Belgium
| | - Perry D Moerland
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, The Netherlands
| | - Saskia Scheij
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Pieter B van Loenen
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Duco S Koenis
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Noam Zelcer
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Claudia M van Tiel
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Carlie J M de Vries
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Hong C, Tontonoz P. Liver X receptors in lipid metabolism: opportunities for drug discovery. Nat Rev Drug Discov 2014; 13:433-44. [DOI: 10.1038/nrd4280] [Citation(s) in RCA: 401] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
16
|
Han SI, Komatsu Y, Murayama A, Steffensen KR, Nakagawa Y, Nakajima Y, Suzuki M, Oie S, Parini P, Vedin LL, Kishimoto H, Shimano H, Gustafsson JÅ, Yanagisawa J. Estrogen receptor ligands ameliorate fatty liver through a nonclassical estrogen receptor/Liver X receptor pathway in mice. Hepatology 2014; 59:1791-802. [PMID: 24277692 DOI: 10.1002/hep.26951] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 11/19/2013] [Indexed: 12/17/2022]
Abstract
UNLABELLED Liver X receptor (LXR) activation stimulates triglyceride (TG) accumulation in the liver. Several lines of evidence indicate that estradiol-17β (E2) reduces TG levels in the liver; however, the molecular mechanism underlying the E2 effect remains unclear. Here, we show that administration of E2 attenuated sterol regulatory element-binding protein (SREBP)-1 expression and TG accumulation induced by LXR activation in mouse liver. In estrogen receptor alpha (ERα) knockout (KO) and liver-specific ERα KO mice, E2 did not affect SREBP-1 expression or TG levels. Molecular analysis revealed that ERα is recruited to the SREBP-1c promoter through direct binding to LXR and inhibits coactivator recruitment to LXR in an E2-dependent manner. Our findings demonstrate the existence of a novel liver-dependent mechanism controlling TG accumulation through the nonclassical ER/LXR pathway. To confirm that a nonclassical ER/LXR pathway regulates ERα-dependent inhibition of LXR activation, we screened ERα ligands that were able to repress LXR activation without enhancing ERα transcriptional activity, and, as a result, we identified the phytoestrogen, phloretin. In mice, phloretin showed no estrogenic activity; however, it did reduce SREBP-1 expression and TG levels in liver of mice fed a high-fat diet to an extent similar to that of E2. CONCLUSION We propose that ER ligands reduce TG levels in the liver by inhibiting LXR activation through a nonclassical pathway. Our results also indicate that the effects of ER on TG accumulation can be distinguished from its estrogenic effects by a specific ER ligand.
Collapse
Affiliation(s)
- Song-iee Han
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Meffre D, Grenier J, Bernard S, Courtin F, Dudev T, Shackleford G, Jafarian-Tehrani M, Massaad C. Wnt and lithium: a common destiny in the therapy of nervous system pathologies? Cell Mol Life Sci 2014; 71:1123-48. [PMID: 23749084 PMCID: PMC11113114 DOI: 10.1007/s00018-013-1378-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/26/2013] [Accepted: 05/16/2013] [Indexed: 02/07/2023]
Abstract
Wnt signaling is required for neurogenesis, the fate of neural progenitors, the formation of neuronal circuits during development, neuron positioning and polarization, axon and dendrite development and finally for synaptogenesis. This signaling pathway is also implicated in the generation and differentiation of glial cells. In this review, we describe the mechanisms of action of Wnt signaling pathways and their implication in the development and correct functioning of the nervous system. We also illustrate how a dysregulated Wnt pathway could lead to psychiatric, neurodegenerative and demyelinating pathologies. Lithium, used for the treatment of bipolar disease, inhibits GSK3β, a central enzyme of the Wnt/β-catenin pathway. Thus, lithium could, to some extent, mimic Wnt pathway. We highlight the possible dialogue between lithium therapy and modulation of Wnt pathway in the treatment of the diseases of the nervous system.
Collapse
Affiliation(s)
- Delphine Meffre
- UMR 8194 CNRS, University Paris Descartes, 45 rue des Saints-Pères, 75270 Paris Cedex 6, France
| | - Julien Grenier
- UMR 8194 CNRS, University Paris Descartes, 45 rue des Saints-Pères, 75270 Paris Cedex 6, France
| | - Sophie Bernard
- UMR 8194 CNRS, University Paris Descartes, 45 rue des Saints-Pères, 75270 Paris Cedex 6, France
| | - Françoise Courtin
- UMR 8194 CNRS, University Paris Descartes, 45 rue des Saints-Pères, 75270 Paris Cedex 6, France
| | - Todor Dudev
- Institute of Biomedical Sciences, Academia Sinica, 11529 Taipei, Taiwan, R.O.C
- Faculty of Chemistry and Pharmacy, University of Sofia, 1 James Bourchier Avenue, 1164 Sofia, Bulgaria
| | | | | | - Charbel Massaad
- UMR 8194 CNRS, University Paris Descartes, 45 rue des Saints-Pères, 75270 Paris Cedex 6, France
| |
Collapse
|
18
|
Ramayo-Caldas Y, Ballester M, Fortes MRS, Esteve-Codina A, Castelló A, Noguera JL, Fernández AI, Pérez-Enciso M, Reverter A, Folch JM. From SNP co-association to RNA co-expression: novel insights into gene networks for intramuscular fatty acid composition in porcine. BMC Genomics 2014; 15:232. [PMID: 24666776 PMCID: PMC3987146 DOI: 10.1186/1471-2164-15-232] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 03/21/2014] [Indexed: 12/19/2022] Open
Abstract
Background Fatty acids (FA) play a critical role in energy homeostasis and metabolic diseases; in the context of livestock species, their profile also impacts on meat quality for healthy human consumption. Molecular pathways controlling lipid metabolism are highly interconnected and are not fully understood. Elucidating these molecular processes will aid technological development towards improvement of pork meat quality and increased knowledge of FA metabolism, underpinning metabolic diseases in humans. Results The results from genome-wide association studies (GWAS) across 15 phenotypes were subjected to an Association Weight Matrix (AWM) approach to predict a network of 1,096 genes related to intramuscular FA composition in pigs. To identify the key regulators of FA metabolism, we focused on the minimal set of transcription factors (TF) that the explored the majority of the network topology. Pathway and network analyses pointed towards a trio of TF as key regulators of FA metabolism: NCOA2, FHL2 and EP300. Promoter sequence analyses confirmed that these TF have binding sites for some well-know regulators of lipid and carbohydrate metabolism. For the first time in a non-model species, some of the co-associations observed at the genetic level were validated through co-expression at the transcriptomic level based on real-time PCR of 40 genes in adipose tissue, and a further 55 genes in liver. In particular, liver expression of NCOA2 and EP300 differed between pig breeds (Iberian and Landrace) extreme in terms of fat deposition. Highly clustered co-expression networks in both liver and adipose tissues were observed. EP300 and NCOA2 showed centrality parameters above average in the both networks. Over all genes, co-expression analyses confirmed 28.9% of the AWM predicted gene-gene interactions in liver and 33.0% in adipose tissue. The magnitude of this validation varied across genes, with up to 60.8% of the connections of NCOA2 in adipose tissue being validated via co-expression. Conclusions Our results recapitulate the known transcriptional regulation of FA metabolism, predict gene interactions that can be experimentally validated, and suggest that genetic variants mapped to EP300, FHL2, and NCOA2 modulate lipid metabolism and control energy homeostasis in pigs.
Collapse
Affiliation(s)
- Yuliaxis Ramayo-Caldas
- Centre de Recerca en Agrigenòmica (CRAG), Consorci CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra 08193, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Gabbi C, Warner M, Gustafsson JÅ. Action mechanisms of Liver X Receptors. Biochem Biophys Res Commun 2013; 446:647-50. [PMID: 24300092 DOI: 10.1016/j.bbrc.2013.11.077] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 11/19/2013] [Indexed: 12/18/2022]
Abstract
The two Liver X Receptors, LXRα and LXRβ, are nuclear receptors belonging to the superfamily of ligand-activated transcription factors. They share more than 78% homology in amino acid sequence, a common profile of oxysterol ligands and the same heterodimerization partner, Retinoid X Receptor. LXRs play crucial roles in several metabolic pathways: lipid metabolism, in particular in preventing cellular cholesterol accumulation; glucose homeostasis; inflammation; central nervous system functions and water transport. As with all nuclear receptors, the transcriptional activity of LXR is the result of an orchestration of numerous cellular factors including ligand bioavailability, presence of corepressors and coactivators and cellular context i.e., what other pathways are activated in the cell at the time the receptor recognizes its ligand. In this mini-review we summarize the factors regulating the transcriptional activity and the mechanisms of action of these two receptors.
Collapse
Affiliation(s)
- Chiara Gabbi
- Center for Nuclear Receptors and Cell Signaling, University of Houston, 3056 Cullen Blv, 77204 Houston, Texas, USA
| | - Margaret Warner
- Center for Nuclear Receptors and Cell Signaling, University of Houston, 3056 Cullen Blv, 77204 Houston, Texas, USA
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, University of Houston, 3056 Cullen Blv, 77204 Houston, Texas, USA; Department of Biosciences and Nutrition, Karolinska Institutet, Novum S-141 86, Sweden.
| |
Collapse
|
20
|
Lee JM, Gang GT, Kim DK, Kim YD, Koo SH, Lee CH, Choi HS. Ursodeoxycholic acid inhibits liver X receptor α-mediated hepatic lipogenesis via induction of the nuclear corepressor SMILE. J Biol Chem 2013; 289:1079-91. [PMID: 24265317 DOI: 10.1074/jbc.m113.491522] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Small heterodimer partner interacting leucine zipper protein (SMILE) has been identified as a nuclear corepressor of the nuclear receptor (NRs) family. Here, we examined the role of SMILE in the regulation of nuclear receptor liver X receptor (LXR)-mediated sterol regulatory element binding protein-1c (SREBP-1c) gene expression. We found that SMILE inhibited T0901317 (T7)-induced transcriptional activity of LXR, which functions as a major regulator of lipid metabolism by inducing SREBP-1c, fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC) gene expression. Moreover, we demonstrated that SMILE physically interacts with LXR and represses T7-induced LXR transcriptional activity by competing with coactivator SRC-1. Adenoviral overexpression of SMILE (Ad-SMILE) attenuated fat accumulation and lipogenic gene induction in the liver of T7 administered or of high fat diet (HFD)-fed mice. Furthermore, we investigated the mechanism by which ursodeoxycholic acid (UDCA) inhibits LXR-induced lipogenic gene expression. Interestingly, UDCA treatment significantly increased SMILE promoter activity and gene expression in an adenosine monophosphate-activated kinase-dependent manner. Furthermore, UDCA treatment repressed T7-induced SREBP-1c, FAS, and ACC protein levels, whereas knockdown of endogenous SMILE gene expression by adenovirus SMILE shRNA (Ad-shSMILE) significantly reversed UDCA-mediated repression of SREBP-1c, FAS, and ACC protein levels. Collectively, these results demonstrate that UDCA activates SMILE gene expression through adenosine monophosphate-activated kinase phosphorylation, which leads to repression of LXR-mediated hepatic lipogenic enzyme gene expression.
Collapse
Affiliation(s)
- Ji-Min Lee
- From the National Creative Research Initiatives Center for Nuclear Receptor Signals and
| | | | | | | | | | | | | |
Collapse
|
21
|
Ren J, Li D, Li Y, Lan X, Zheng J, Wang X, Ma J, Lu S. HDAC3 interacts with sumoylated C/EBPα to negatively regulate the LXRα expression in rat hepatocytes. Mol Cell Endocrinol 2013; 374:35-45. [PMID: 23639777 DOI: 10.1016/j.mce.2013.04.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 04/16/2013] [Accepted: 04/19/2013] [Indexed: 01/22/2023]
Abstract
The expression changes of liver X receptor alpha (LXRα), histone deacetylase 3 (HDAC3) and CCAAT/enhancer binding protein alpha (C/EBPα) were detected in liver tissues of our high-fat-diet E3 rat model. The aim of this study is to pinpoint the molecular mechanism of HDAC3 and C/EBPα to orchestrate LXRα expression in hepatocytes. We confirmed that LXRα and its target genes were negatively regulated by HDAC3 in stable expressed clones with pEGFP-Hdac3 or shRNA-Hdac3 vector. However, transient pEGFP-C/EBPα plasmid transfection showed an upregulation of LXRα expression and C/EBPα enhanced LXRα promoter activity in a dose-dependent manner in CBRH-7919 cells. By using 5'-serial deletion reporter analysis, we identified that fragment from -2881 to -1181bp of LXRα promoter was responsible for C/EBPα binding to the promoter, especially CBS1 and CBS4 were identified essentially by using ChIP and luciferase reporter assay. Co-IP, qRT-PCR and ChIP revealed that HDAC3 interacted with C/EBPα co-regulated LXRα expression. Sumoylation of C/EBPα at lysine 159 was detected in CBRH-7919 cells with transient overexpressed C/EBPα, and Co-IP assay detected that sumoylated C/EBPα interacted with more HDAC3 than C/EBPα K159L mutant. Luciferase reporter assay demonstrated that C/EBPα participated in HDAC3-repressed LXRα transcription, and HDAC3 was involved in sumoylated C/EBPα-inactivated LXRα activity. Luciferase reporter assay demonstrated that sumoylation of C/EBPα by SUMO-1 directly reversed the activation of C/EBPα on LXRα promoter. The results suggested that HDAC3 interacts with sumoylated C/EBPα to negatively regulate the LXRα expression.
Collapse
Affiliation(s)
- Juan Ren
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, PR China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Hoang JJ, Baron S, Volle DH, Lobaccaro JMA, Trousson A. Lipids, LXRs and prostate cancer: Are HDACs a new link? Biochem Pharmacol 2013; 86:168-74. [DOI: 10.1016/j.bcp.2013.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/05/2013] [Accepted: 04/05/2013] [Indexed: 12/29/2022]
|
23
|
A regulatory role for microRNA 33* in controlling lipid metabolism gene expression. Mol Cell Biol 2013; 33:2339-52. [PMID: 23547260 DOI: 10.1128/mcb.01714-12] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
hsa-miR-33a and hsa-miR-33b, intronic microRNAs (miRNAs) located within the sterol regulatory element-binding protein 2 and 1 genes (Srebp-2 and -1), respectively, have recently been shown to regulate lipid homeostasis in concert with their host genes. Although the functional role of miR-33a and -b has been highly investigated, the role of their passenger strands, miR-33a* and -b*, remains unclear. Here, we demonstrate that miR-33a* and -b* accumulate to steady-state levels in human, mouse, and nonhuman primate tissues and share a similar lipid metabolism target gene network as their sister strands. Analogous to miR-33, miR-33* represses key enzymes involved in cholesterol efflux (ABCA1 and NPC1), fatty acid metabolism (CROT and CPT1a), and insulin signaling (IRS2). Moreover, miR-33* also targets key transcriptional regulators of lipid metabolism, including SRC1, SRC3, NFYC, and RIP140. Importantly, inhibition of either miR-33 or miR-33* rescues target gene expression in cells overexpressing pre-miR-33. Consistent with this, overexpression of miR-33* reduces fatty acid oxidation in human hepatic cells. Altogether, these data support a regulatory role for the miRNA* species and suggest that miR-33 regulates lipid metabolism through both arms of the miR-33/miR-33* duplex.
Collapse
|
24
|
Interplay between LXR and Wnt/β-catenin signaling in the negative regulation of peripheral myelin genes by oxysterols. J Neurosci 2011; 31:9620-9. [PMID: 21715627 DOI: 10.1523/jneurosci.0761-11.2011] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Oxysterols are reactive molecules generated from the oxidation of cholesterol. Their implication in cholesterol homeostasis and in the progression of neurodegenerative disorders is well known, but few data are available for their functions in the peripheral nervous system. Our aim was to study the influence of oxysterols on myelin gene expression and myelin sheath formation in peripheral nerves. We show by gas chromatography/mass spectrometry that Schwann cells and sciatic nerves contain 24(S)-hydroxycholesterol, 25-hydroxycholesterol, and 27-hydroxycholesterol and that they express their biosynthetic enzymes and receptors (liver X receptors LXRα and LXRβ). We demonstrate that oxysterols inhibit peripheral myelin gene expression [myelin protein zero (MPZ) and peripheral myelin protein-22 (PMP22)] in a Schwann cell line. This downregulation is mediated by either LXRα or LXRβ, depending on the promoter context, as suggested by siRNA strategy and chromatin immunoprecipitation assays in Schwann cells and in the sciatic nerve of LXR knock-out mice. Importantly, the knock-out of LXR in mice results in thinner myelin sheaths surrounding the axons. Oxysterols repress myelin genes via two mechanisms: by binding of LXRs to myelin gene promoters and by inhibiting the Wnt/β-catenin pathway that is crucial for the expression of myelin genes. The Wnt signaling components (Disheveled, TCF/LEF, β-catenin) are strongly repressed by oxysterols. Furthermore, the recruitment of β-catenin at the levels of the MPZ and PMP22 promoters is decreased. Our data reveal new endogenous mechanisms for the negative regulation of myelin gene expression, highlight the importance of oxysterols and LXR in peripheral nerve myelination, and open new perspectives of treating demyelinating diseases with LXR agonists.
Collapse
|
25
|
Huwait EA, Greenow KR, Singh NN, Ramji DP. A novel role for c-Jun N-terminal kinase and phosphoinositide 3-kinase in the liver X receptor-mediated induction of macrophage gene expression. Cell Signal 2011; 23:542-9. [PMID: 21070853 PMCID: PMC3126994 DOI: 10.1016/j.cellsig.2010.11.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/29/2010] [Accepted: 11/03/2010] [Indexed: 11/01/2022]
Abstract
Liver X receptors (LXRs) are ligand-dependent transcription factors that are activated by metabolites of cholesterol, oxysterols, and a number of synthetic agonists. LXRs play potent anti-atherogenic roles in part by stimulating the efflux of cholesterol from macrophage foam cells. The LXR-induced expression of ATP-binding cassette transporter (ABC)-A1 and Apolipoprotein E (ApoE) in macrophages is essential for the stimulation of cholesterol efflux and the prevention of atherosclerotic development. Unfortunately, the signaling pathways underlying such regulation are poorly understood and were therefore investigated in human macrophages. The expression of ApoE and ABCA1 induced by synthetic or natural LXR ligands [TO901317, GW3965, and 22-(R)-hydroxycholesterol (22-(R)-HC), respectively] was attenuated by inhibitors of c-Jun N-terminal kinase (JNK) (curcumin and SP600125) and phosphoinositide 3-kinase (PI3K) (LY294002). Similar results were obtained with ABCG1 and LXR-α, two other LXR target genes. LXR agonists activated several components of the JNK pathway (SEK1, JNK and c-Jun) along with AKT, a downstream target for PI3K. In addition, dominant negative mutants of JNK and PI3K pathways inhibited the LXR-agonists-induced activity of the ABCA1 and LXR-α gene promoters in transfected cells. LXR agonists also induced the binding of activator protein-1 (AP-1), a key transcription factor family regulated by JNK, to recognition sequences present in the regulatory regions of the ApoE and ABCA1 genes. These studies reveal a novel role for JNK and PI3K/AKT signaling in the LXR-regulated expression in macrophages of several key genes implicated in atherosclerosis.
Collapse
Affiliation(s)
| | | | - Nishi N. Singh
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Dipak P. Ramji
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| |
Collapse
|
26
|
Nedumaran B, Kim GS, Hong S, Yoon YS, Kim YH, Lee CH, Lee YC, Koo SH, Choi HS. Orphan nuclear receptor DAX-1 acts as a novel corepressor of liver X receptor alpha and inhibits hepatic lipogenesis. J Biol Chem 2010; 285:9221-32. [PMID: 20080977 DOI: 10.1074/jbc.m109.073650] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
DAX-1 (dosage-sensitive sex reversal adrenal hypoplasia congenital critical region on X chromosome, gene 1) is a member of the nuclear receptor superfamily that can repress diverse nuclear receptors and has a key role in adreno-gonadal development. Our previous report has demonstrated that DAX-1 can inhibit hepatocyte nuclear factor 4alpha transactivity and negatively regulate gluconeogenic gene expression (Nedumaran, B., Hong, S., Xie, Y. B., Kim, Y. H., Seo, W. Y., Lee, M. W., Lee, C. H., Koo, S. H., and Choi, H. S. (2009) J. Biol. Chem. 284, 27511-27523). Here, we further expand the role of DAX-1 in hepatic energy metabolism. Transfection assays have demonstrated that DAX-1 can inhibit the transcriptional activity of nuclear receptor liver X receptor alpha (LXRalpha). Physical interaction between DAX-1 and LXRalpha was confirmed Immunofluorescent staining in mouse liver shows that LXRalpha and DAX-1 are colocalized in the nucleus. Domain mapping analysis shows that the entire region of DAX-1 is involved in the interaction with the ligand binding domain region of LXRalpha. Competition analyses demonstrate that DAX-1 competes with the coactivator SRC-1 for repressing LXRalpha transactivity. Chromatin immunoprecipitation assay showed that endogenous DAX-1 recruitment on the SREBP-1c gene promoter was decreased in the presence of LXRalpha agonist. Overexpression of DAX-1 inhibits T7-induced LXRalpha target gene expression, whereas knockdown of endogenous DAX-1 significantly increases T7-induced LXRalpha target gene expression in HepG2 cells. Finally, overexpression of DAX-1 in mouse liver decreases T7-induced LXRalpha target gene expression, liver triglyceride level, and lipid accumulation. Overall, this study suggests that DAX-1, a novel corepressor of LXRalpha, functions as a negative regulator of lipogenic enzyme gene expression in liver.
Collapse
Affiliation(s)
- Balachandar Nedumaran
- Hormone Research Center, School of Biological Science and Technology, Chonnam National University, Gwangju 500-757, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Expression of sterol 27-hydroxylase in glial cells and its regulation by liver X receptor signaling. Neuroscience 2009; 164:530-40. [DOI: 10.1016/j.neuroscience.2009.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 07/31/2009] [Accepted: 08/01/2009] [Indexed: 11/21/2022]
|
28
|
Trousson A, Makoukji J, Petit PX, Bernard S, Slomianny C, Schumacher M, Massaad C. Cross-talk between oxysterols and glucocorticoids: differential regulation of secreted phopholipase A2 and impact on oligodendrocyte death. PLoS One 2009; 4:e8080. [PMID: 19956653 PMCID: PMC2779104 DOI: 10.1371/journal.pone.0008080] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 11/05/2009] [Indexed: 11/19/2022] Open
Abstract
Background Oxysterols are oxidized forms of cholesterol. They have been shown to be implicated in cholesterol turnover, inflammation and in neurodegenerative diseases such as Alzheimer's disease and multiple sclerosis. Glial cells are targets of oxysterols: they inhibit astrocyte proliferation after brain injury, and we have previously shown that 25-hydroxycholesterol (25OH) provokes oligodendrocyte apoptosis and stimulates the expression of sPLA2 type IIA (sPLA2-IIA), which has a protective effect. Methodology/Principal Findings As glucocorticoids are well-known for their anti-inflammatory effects, our aim was to understand their direct effects on oxysterol-induced responses in oligodendrocytes (sPLA2-IIA stimulation and apoptosis). We demonstrate that the synthetic glucocorticoid dexamethasone (Dex) abolishes the stimulation of sPLA2-IIA by 25-hydroxycholesterol (25-OH). This inhibition is mediated by the glucocorticoid receptor (GR), which decreases the expression of the oxysterol receptor Pregnane X Receptor (PXR) and interferes with oxysterol signaling by recruiting a common limiting coactivator PGC1α. Consistent with the finding that sPLA2-IIA can partially protect oligodendrocytes against oxysterol-triggered apoptosis, we demonstrate here that the inhibition of sPLA2-IIA by Dex accelerates the apoptotic phenomenon, leading to a shift towards necrosis. We have shown by atomic force microscopy and electron microscopy that 25-OH and Dex alters oligodendrocyte shape and disorganizes the cytoplasm. Conclusions/Significance Our results provide a new understanding of the cross-talk between oxysterol and glucocorticoid signaling pathways and their respective roles in apoptosis and oligodendrocyte functions.
Collapse
Affiliation(s)
- Amalia Trousson
- UMR788, Inserm and University Paris-Sud 11, IFR 93, Le Kremlin-Bicêtre, France
- UPR 2228, CNRS and University Paris Descartes, IFR95, Paris, France
| | - Joelle Makoukji
- UPR 2228, CNRS and University Paris Descartes, IFR95, Paris, France
| | - Patrice X. Petit
- Cancer, Apoptosis, and Mitochondria Team, UMR8104 CNRS, Institut Cochin, Paris, France
| | - Sophie Bernard
- UPR 2228, CNRS and University Paris Descartes, IFR95, Paris, France
| | | | - Michael Schumacher
- UMR788, Inserm and University Paris-Sud 11, IFR 93, Le Kremlin-Bicêtre, France
| | - Charbel Massaad
- UMR788, Inserm and University Paris-Sud 11, IFR 93, Le Kremlin-Bicêtre, France
- UPR 2228, CNRS and University Paris Descartes, IFR95, Paris, France
- * E-mail:
| |
Collapse
|
29
|
Son YL, Lee YC. Molecular determinants of the interactions between LXR/RXR heterodimers and TRAP220. Biochem Biophys Res Commun 2009; 384:389-93. [PMID: 19410560 DOI: 10.1016/j.bbrc.2009.04.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Accepted: 04/29/2009] [Indexed: 10/20/2022]
|
30
|
Hepatitis B virus X protein induces lipogenic transcription factor SREBP1 and fatty acid synthase through the activation of nuclear receptor LXRalpha. Biochem J 2008; 416:219-30. [PMID: 18782084 DOI: 10.1042/bj20081336] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
HBV (hepatitis B virus) is a primary cause of chronic liver disease, which frequently results in hepatitis, cirrhosis and ultimately HCC (hepatocellular carcinoma). Recently, we showed that HBx (HBV protein X) expression induces lipid accumulation in hepatic cells mediated by the induction of SREBP1 (sterol-regulatory-element-binding protein 1), a key regulator of lipogenic genes in the liver. However, the molecular mechanisms by which HBx increases SREBP1 expression and transactivation remain to be clearly elucidated. In the present study, we demonstrated that HBx interacts with LXRalpha (liver X receptor alpha) and enhances the binding of LXRalpha to LXRE (LXR-response element), thereby resulting in the up-regulation of SREBP1 and FAS (fatty acid synthase) in the presence or absence of the LXR agonist T0901317 in the hepatic cells and HBx-transgenic mice. Furthermore, HBx also augments the ability to recruit ASC2 (activating signal co-integrator 2), a transcriptional co-activator that controls liver lipid metabolic pathways, to the LXRE with LXRalpha. These studies place LXRalpha in a key position within the HBx-induced lipogenic pathways, and suggest a molecular mechanism through which HBV infection can stimulate the SREBP1-mediated control of hepatic lipid accumulation.
Collapse
|
31
|
Uno S, Endo K, Jeong Y, Kawana K, Miyachi H, Hashimoto Y, Makishima M. Suppression of beta-catenin signaling by liver X receptor ligands. Biochem Pharmacol 2008; 77:186-95. [PMID: 18983830 DOI: 10.1016/j.bcp.2008.10.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 10/03/2008] [Accepted: 10/03/2008] [Indexed: 12/21/2022]
Abstract
The nuclear receptors liver X receptor (LXR) alpha and LXRbeta serve as oxysterol receptors and play an important role in the regulation of lipid metabolism. We investigated the potential effects of LXRs on pathways of colon carcinogenesis and found that LXR activation suppresses the transactivation activity of beta-catenin, a key molecule in Wnt signaling. LXRalpha and LXRbeta inhibited beta-catenin transactivation of T cell factor-mediated transcription in a ligand-dependent manner. LXR activation suppressed an oncogenic beta-catenin, which has phosphorylation site mutations, and did not change beta-catenin protein expression in cells. In contrast, beta-catenin enhanced LXR transactivation activity. Nuclear LXRs and beta-catenin were coimmunoprecipitated in colon cancer HCT116 cells, and in vitro experiments showed that LXRs bind directly to the Armadillo repeat region of beta-catenin in a ligand-independent manner. LXR ligand decreased mRNA expression of beta-catenin targets, MYC, MMP7 and BMP4, and recruited LXRs to MYC and MMP7 promoters. Transfection of a dominant negative LXR to HCT116 cells and experiments using LXR-null cells showed the involvement of cellular LXRs in beta-catenin suppression and proliferation inhibition. The results show lipid-sensing receptor LXRs regulate the beta-catenin activity and cellular proliferation.
Collapse
Affiliation(s)
- Shigeyuki Uno
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Prüfer K, Hernandez C, Gilbreath M. Mutations in the AF-2 region abolish ligand-induced intranuclear immobilization of the liver X receptor alpha. Exp Cell Res 2008; 314:2652-60. [PMID: 18599038 DOI: 10.1016/j.yexcr.2008.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 05/14/2008] [Accepted: 05/15/2008] [Indexed: 11/19/2022]
Abstract
The liver X receptors (LXR) alpha and beta are ligand-induced transcription factors that regulate the expression of genes important for cholesterol metabolism, lipogenesis, and other metabolic pathways. Despite their high degree of similarity, LXRs have redundant as well as nonredundant functions. The regulation of LXRs' intranuclear mobility most likely plays a major role in the regulation of their transcriptional activities. In order to elucidate how ligand binding, receptor-protein and receptor-DNA interactions affect intranuclear receptor mobility, we expressed transcriptionally active yellow fluorescent protein (YFP)-LXR alpha and YFP-LXR beta in Cos-7 cells. We used the fluorescence recovery after photobleaching (FRAP) technique and confocal laser scanning microscopy as well as Triton X-100 permeabilization experiments and fluorescence microscopy to measure differences in the intranuclear mobility between LXR alpha and LXR beta. The image analyses revealed that after agonist binding, LXR alpha exhibits slower intranuclear trafficking and greater intranuclear immobilization compared with LXR beta. In addition, mutational analysis showed that the integrity of the Activation Function (AF)-2 region of LXR alpha is essential for its immobilization whereas the integrity of the DNA binding domain is not. These findings imply that specific protein interactions with the AF-2 region of LXR alpha play a role in its intranuclear immobilization.
Collapse
Affiliation(s)
- Kirsten Prüfer
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
| | | | | |
Collapse
|
33
|
Raghow R, Yellaturu C, Deng X, Park EA, Elam MB. SREBPs: the crossroads of physiological and pathological lipid homeostasis. Trends Endocrinol Metab 2008; 19:65-73. [PMID: 18291668 DOI: 10.1016/j.tem.2007.10.009] [Citation(s) in RCA: 243] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 10/22/2007] [Accepted: 10/29/2007] [Indexed: 12/14/2022]
Abstract
The uptake, biosynthesis and metabolism of cholesterol and other lipids are exquisitely regulated by feedback and feed-forward pathways in organisms ranging from Caenorhabditis elegans to humans. As endoplasmic reticulum (ER) membrane-embedded transcription factors that are activated in the Golgi apparatus, sterol regulatory element-binding proteins (SREBPs) are central to the intracellular surveillance of lipid catabolism and de novo biogenesis. The biosynthesis of SREBP proteins, their migration from the ER to the Golgi compartment, intra-membrane proteolysis, nuclear translocation and trans-activation potential are tightly controlled in vivo. Here we summarize recent studies elucidating the transcriptional and post-transcriptional regulation of SREBP-1c through nutrition and the action of hormones, particularly insulin, and the resulting implications for dyslipidemia of obesity, metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- Rajendra Raghow
- Department of Pharmacology, University of Tennessee Health Science Center, 874 Union Avenue, Memphis, TN 38163, USA.
| | | | | | | | | |
Collapse
|
34
|
RXR heterodimerization allosterically activates LXR binding to the second NR box of activating signal co-integrator-2. Biochem J 2008; 410:319-30. [PMID: 18031289 DOI: 10.1042/bj20070837] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
ASC-2 (activating signal co-integrator-2) is a transcriptional co-activator that mediates the transactivation of NRs (nuclear receptors) via direct interactions with these receptors. ASC-2 contains two separate NR-interaction domains harbouring a core signature motif, LXXLL (where X is any amino acid), named the NR box. Although the first NR box (NR box-1) of ASC-2 interacts with many different NRs, the second NR box (NR box-2) specifically interacts with only LXR (liver X receptor), whose transactivation in vivo requires heterodimerization with RXR (retinoid X receptor). Interestingly, RXR has been shown to enhance the LXR transactivation, even in the absence of LXR ligand via a unique mechanism of allosteric regulation. In the present study we demonstrate that LXR binding to an ASC-2 fragment containing NR box-2 (Co4aN) is enhanced by RXR and even further by liganded RXR. We also identified specific residues in Co4aN involved in its interaction with LXR that were also required for the ASC-2-mediated transactivation of LXR in mammalian cells. Using these mutants, we found that the Co4aN–LXR interaction surface is not altered by the presence of RXR and RXR ligand and that the Ser1490 residue is the critical determinant for the LXR-specific interaction of Co4aN. Notably the NR box-2, but not the NR box-1, is essential for ASC-2-mediated transactivation of LXR in vivo and for the interaction between LXR–RXR and ASC-2 in vitro. These results indicate that RXR does not interact directly with NR box-1 of ASC-2, but functions as an allosteric activator of LXR binding to NR box-2 of ASC-2.
Collapse
|
35
|
Deng X, Yellaturu C, Cagen L, Wilcox HG, Park EA, Raghow R, Elam MB. Expression of the rat sterol regulatory element-binding protein-1c gene in response to insulin is mediated by increased transactivating capacity of specificity protein 1 (Sp1). J Biol Chem 2007; 282:17517-29. [PMID: 17449871 DOI: 10.1074/jbc.m702228200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The induction of genes involved in lipid biosynthesis by insulin is mediated in part by the sterol regulatory element-binding protein-1c (SREBP-1c). SREBP-1c is directly regulated by insulin by transcriptional and post-transcriptional mechanisms. Previously, we have demonstrated that the insulin-responsive cis-acting unit of the rat SREBP-1c promoter is composed of several elements that include a sterol regulatory element, two liver X receptor elements, and a number of conserved GC boxes. Here we systematically dissected the role of these GC boxes and report that five bona fide Sp1-binding elements of the SREBP-1c promoter determine its basal and insulin-induced activation. Luciferase expression driven by the rat SREBP-1c promoter was accelerated by ectopic expression of Sp1, and insulin further enhanced the transactivation potential of Sp1. Introduction of a small interfering RNA against Sp1 reduced both basal and insulin-induced activation of the SREBP-1c promoter. We also found that Sp1 interacted with both SREBP-1c and LXRalpha proteins and that insulin promoted these interactions. Chromatin immunoprecipitation studies revealed that insulin facilitated the recruitment of the steroid receptor coactivator-1 to the SREBP-1c promoter. These studies identify a novel mechanism by which maximal activation of the rat SREBP-1c gene expression by insulin is mediated by Sp1 and its enhanced ability to interact with other transcriptional regulatory proteins.
Collapse
Affiliation(s)
- Xiong Deng
- Medical and Research Service, Department of Veterans Affairs Medical Center, Memphis, Tennessee 38104, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Moore DD, Kato S, Xie W, Mangelsdorf DJ, Schmidt DR, Xiao R, Kliewer SA. International Union of Pharmacology. LXII. The NR1H and NR1I receptors: constitutive androstane receptor, pregnene X receptor, farnesoid X receptor alpha, farnesoid X receptor beta, liver X receptor alpha, liver X receptor beta, and vitamin D receptor. Pharmacol Rev 2007; 58:742-59. [PMID: 17132852 DOI: 10.1124/pr.58.4.6] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The nuclear receptors of the NR1H and NR1I subgroups include the constitutive androstane receptor, pregnane X receptor, farnesoid X receptors, liver X receptors, and vitamin D receptor. The newly emerging functions of these related receptors are under the control of metabolic pathways, including metabolism of xenobiotics, bile acids, cholesterol, and calcium. This review summarizes results of structural, pharmacologic, and genetic studies of these receptors.
Collapse
Affiliation(s)
- David D Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Prüfer K, Boudreaux J. Nuclear localization of liver X receptor α and β is differentially regulated. J Cell Biochem 2007; 100:69-85. [PMID: 16888799 DOI: 10.1002/jcb.21006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Activity of nuclear receptors is regulated by their nuclear localization. Liver X receptors (LXR) alpha and beta are nuclear receptors that regulate transcription of genes for cholesterol metabolism, cholesterol transport, and lipogenesis. While LXR alpha and beta are very similar in structure and exhibit similar ligand binding properties, their physiological roles are quite different. Since the LXRs fall into a class of receptors that move between the nucleus and cytoplasm, experiments were conducted to determine whether LXR alpha and LXR beta show differences in their nuclear localization pattern. To determine the location of each receptor, cell lines stably expressing yellow fluorescent protein (YFP) chimeras with either LXR alpha or LXR beta were examined. Retention in the nucleus of the chimeric proteins in the presence or absence of ligands was assessed using fluorescence microscopy coupled with digitonin permeabilization assays. Surprisingly, differences were found between LXR alpha and LXR beta. Whereas unliganded LXR alpha was retained in the nucleus, unliganded LXR beta was partially exported. Mutations were then introduced into putative nuclear localization sequences (NLS) to determine which sequences are important for nuclear localization and function. Mutation in one such sequence abolished nuclear localization of LXR alpha, whereas the analogous change in LXR beta had a much less dramatic effect. Mutations in analogous putative NLS also differentially affected transcriptional activation by LXR alpha and LXR beta. These data demonstrate for the first time that nuclear retention and localization as well as function of LXR alpha and LXR beta are differentially regulated.
Collapse
Affiliation(s)
- Kirsten Prüfer
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA.
| | | |
Collapse
|
38
|
Chen M, Li W, Wang N, Zhu Y, Wang X. ROS and NF-kappaB but not LXR mediate IL-1beta signaling for the downregulation of ATP-binding cassette transporter A1. Am J Physiol Cell Physiol 2006; 292:C1493-501. [PMID: 17135302 DOI: 10.1152/ajpcell.00016.2006] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
ATP-binding cassette transporter A1 (ABCA1), a pivotal regulator of cholesterol efflux from cells to apolipoproteins, plays an important role in cholesterol homeostasis. As an inflammatory factor, IL-1beta has been shown to downregulate ABCA1 in macrophages and facilitates foam cell formation. However, the molecular mechanism underlining the downregulated ABCA1 by IL-1beta is still elusive. In the present study, we demonstrated that IL-1beta downregulated ABCA1 but not ABCG1 at mRNA and protein levels in a time- and dose-dependent manner in THP-1 and A549 cells. IL-1beta attenuated ABCA1 promoter activity through an LXR (liver X receptor)-independent pathway, since IL-1beta did not alter the expression and activities of LXRalpha/beta, and deletion of the LXR responsive element from the ABCA1 promoter failed to reverse the IL-1beta effect. In contrast, NF-kappaB inhibition by pyrrolidine dithiocarbamate and MG132 prevented the suppression of ABCA1 by IL-1beta. Cotransfection with ABCA1 luciferase reporter and the expression plasmids of Rel A decreased ABCA1 promoter activities. An adenovirus expressing NF-kappaB inhibitor subunit-alpha inhibited NF-kappaB activities and also reversed the IL-1beta effect at the promoter activity and protein levels of ABCA1. In addition, IL-1beta could induce the production of reactive oxygen species (ROS), and N-acetyl-L-cysteine, a scavenger of ROS, reversed the decreased level of ABCA1 induced by IL-1beta. H(2)O(2) decreased ABCA1 at the mRNA and protein levels and the promoter activity. Thus our data provide strong evidence that ROS and NF-kappaB, but not LXR, mediate the IL-1beta-induced downregulation of ABCA1 via a novel transcriptional mechanism, which might play an important role of proinflammation in the alteration of lipid metabolism.
Collapse
Affiliation(s)
- Min Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing 100083, People's Republic of China
| | | | | | | | | |
Collapse
|
39
|
Huuskonen J, Vishnu M, Chau P, Fielding PE, Fielding CJ. Liver X Receptor Inhibits the Synthesis and Secretion of Apolipoprotein A1 by Human Liver-Derived Cells. Biochemistry 2006; 45:15068-74. [PMID: 17154544 DOI: 10.1021/bi061378y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The liver X receptor (LXR) agonist TO901317 inhibited the synthesis of apolipoprotein A1 (apo A1) by human liver-derived cells, including the formation of lipid-poor, prebeta-migrating high-density lipoprotein (HDL). Despite activation of the lipid transporter ABCA1 under these conditions, cellular efflux of PL and cholesterol from liver cells was also reduced. By assaying transcription from full-length and truncated promoters and by site-directed mutagenesis, the effect of LXR and its ligand was localized to a binding site for hepatic nuclear factor-4 (HNF4) in the proximal apo A1 promoter (-132/-119 bp). Chromatin immunoprecipitation analysis of apo A1 transcription complexes from control and ligand-activated cells showed an increase in the binding of reported apo A1 transcriptional inhibitor COUP-TF, which competes with HNF4 for DNA binding. It also identified LXR in the apo A1 transcription complex of TO901317-treated cells. Displacement of HNF4 from the -132/-119 bp promoter DNA sequence in the presence of TO901317 was confirmed by gel shift analysis. These data indicate that LXR can be a significant negative regulator of apo A1 transcription and HDL synthesis.
Collapse
Affiliation(s)
- J Huuskonen
- Cardiovascular Research Institute, University of California, San Francisco, California 94143, USA
| | | | | | | | | |
Collapse
|
40
|
Talukdar S, Hillgartner FB. The mechanism mediating the activation of acetyl-coenzyme A carboxylase-alpha gene transcription by the liver X receptor agonist T0-901317. J Lipid Res 2006; 47:2451-61. [PMID: 16931873 DOI: 10.1194/jlr.m600276-jlr200] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In birds and mammals, agonists of the liver X receptor (LXR) increase the expression of enzymes that make up the fatty acid synthesis pathway. Here, we investigate the mechanism by which the synthetic LXR agonist, T0-901317, increases the transcription of the acetyl-coenzyme A carboxylase-alpha (ACC alpha) gene in chick embryo hepatocyte cultures. Transfection analyses demonstrate that activation of ACC alpha transcription by T0-901317 is mediated by a cis-acting regulatory unit (-101 to -71 bp) that is composed of a liver X receptor response element (LXRE) and a sterol-regulatory element (SRE). The SRE enhances the ability of the LXRE to activate ACC alpha transcription in the presence of T0-901317. Treating hepatocytes with T0-901317 increases the concentration of mature sterol-regulatory element binding protein-1 (SREBP-1) in the nucleus and the acetylation of histone H3 and histone H4 at the ACC alpha LXR response unit. These results indicate that T0-901317 increases hepatic ACC alpha transcription by directly activating LXR*retinoid X receptor (RXR) heterodimers and by increasing the activity of an accessory transcription factor (SREBP-1) that enhances ligand induced-LXR*RXR activity.
Collapse
Affiliation(s)
- Saswata Talukdar
- Department of Biochemistry and Molecular Pharmacology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | | |
Collapse
|
41
|
Sevov M, Elfineh L, Cavelier LB. Resveratrol regulates the expression of LXR-alpha in human macrophages. Biochem Biophys Res Commun 2006; 348:1047-54. [PMID: 16901463 DOI: 10.1016/j.bbrc.2006.07.155] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Accepted: 07/24/2006] [Indexed: 02/04/2023]
Abstract
The naturally occurring polyphenol resveratrol has been associated with the beneficial effects of red wine consumption on cardiovascular disease and shown to inhibit atherosclerosis in animal models. To determine if resveratrol affects the expression of genes that control lipid homeostasis in human macrophages, we measured expression changes in the LXR-alpha pathway, crucial to cholesterol efflux, and in genes that mediate lipoprotein uptake. Resveratrol treatment of THP-1 macrophages induced LXR-alpha at mRNA and protein levels. Increased recruitment of RNA polymerase II to the LXR-alpha promoter suggested that up-regulation was at least partly mediated by transcriptional mechanisms. Resveratrol also induced LXR-alpha in human monocyte-derived macrophages together with elevated ABCA1 and ABCG1 mRNA levels. Moreover, resveratrol repressed the expression of the lipid uptake genes LPL and SR-AII. The ability of resveratrol to modulate expression of the genes involved in lipid uptake and efflux suggests that polyphenols can potentially limit cholesterol accumulation in human macrophages.
Collapse
Affiliation(s)
- Marie Sevov
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | | | | |
Collapse
|
42
|
Verreault M, Senekeo-Effenberger K, Trottier J, Bonzo JA, Bélanger J, Kaeding J, Staels B, Caron P, Tukey RH, Barbier O. The liver X-receptor alpha controls hepatic expression of the human bile acid-glucuronidating UGT1A3 enzyme in human cells and transgenic mice. Hepatology 2006; 44:368-78. [PMID: 16871576 DOI: 10.1002/hep.21259] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Glucuronidation, an important bile acid detoxification pathway, is catalyzed by enzymes belonging to the UDP-glucuronosyltransferase (UGT) family. Among UGT enzymes, UGT1A3 is considered the major human enzyme for the hepatic C24-glucuronidation of the primary chenodeoxycholic (CDCA) and secondary lithocholic (LCA) bile acids. We identify UGT1A3 as a positively regulated target gene of the oxysterol-activated nuclear receptor liver X-receptor alpha (LXRalpha). In human hepatic cells and human UGT1A transgenic mice, LXRalpha activators induce UGT1A3 mRNA levels and the formation of CDCA-24glucuronide (24G) and LCA-24G. Furthermore, a functional LXR response element (LXRE) was identified in the UGT1A3 promoter by site-directed mutagenesis, electrophoretic mobility shift assays and chromatin immunoprecipitation experiment. In addition, LXRalpha is found to interact with the SRC-1alpha and NCoR cofactors to regulate the UGT1A3 gene, but not with PGC-1beta. In conclusion, these observations establish LXRalpha as a crucial regulator of bile acid glucuronidation in humans and suggest that accumulation of oxysterols in hepatocytes during cholestasis favors bile acid detoxification as glucuronide conjugates. LXR agonists may be useful for stimulating both bile acid detoxification and cholesterol removal in cholestatic or hypercholesterolemic patients, respectively.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cells, Cultured
- Chromatin Immunoprecipitation
- DNA-Binding Proteins/drug effects
- DNA-Binding Proteins/metabolism
- Gene Expression
- Glucuronosyltransferase/drug effects
- Glucuronosyltransferase/genetics
- Glucuronosyltransferase/metabolism
- Hepatocytes/cytology
- Hepatocytes/metabolism
- Humans
- Hydrocarbons, Fluorinated
- In Vitro Techniques
- Liver X Receptors
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Orphan Nuclear Receptors
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- Receptors, Cytoplasmic and Nuclear/drug effects
- Receptors, Cytoplasmic and Nuclear/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sulfonamides/pharmacology
Collapse
Affiliation(s)
- Mélanie Verreault
- Molecular Endocrinology and Oncology Research Center, CHUL Research Center and the Faculty of Pharmacy, Laval University, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Steffensen KR, Gustafsson JÅ. Liver X receptors: new drug targets to treat Type 2 diabetes? ACTA ACUST UNITED AC 2006. [DOI: 10.2217/17460875.1.2.181] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
44
|
Abstract
Liver X receptors (LXRs) and farnesoid X receptor (FXR) are nuclear receptors that function as intracellular sensors for sterols and bile acids, respectively. In response to their ligands, these receptors induce transcriptional responses that maintain a balanced, finely tuned regulation of cholesterol and bile acid metabolism. LXRs also permit the efficient storage of carbohydrate- and fat-derived energy, whereas FXR activation results in an overall decrease in triglyceride levels and modulation of glucose metabolism. The elegant, dual interplay between these two receptor systems suggests that they coevolved to constitute a highly sensitive and efficient system for the maintenance of total body fat and cholesterol homeostasis. Emerging evidence suggests that the tissue-specific action of these receptors is also crucial for the proper function of the cardiovascular, immune, reproductive, endocrine pancreas, renal, and central nervous systems. Together, LXRs and FXR represent potential therapeutic targets for the treatment and prevention of numerous metabolic and lipid-related diseases.
Collapse
Affiliation(s)
- Nada Y Kalaany
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.
| | | |
Collapse
|
45
|
Yin L, Wang Y, Dridi S, Vinson C, Hillgartner FB. Role of CCAAT/enhancer-binding protein, histone acetylation, and coactivator recruitment in the regulation of malic enzyme transcription by thyroid hormone. Mol Cell Endocrinol 2005; 245:43-52. [PMID: 16293364 DOI: 10.1016/j.mce.2005.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2005] [Revised: 10/07/2005] [Accepted: 10/11/2005] [Indexed: 11/23/2022]
Abstract
In chick embryo hepatocytes, activation of malic enzyme gene transcription by triiodothyronine (T3) is mediated by a T3 response unit (T3RU) that contains five T3 response elements (T3REs) plus five accessory elements that enhance T3 responsiveness conferred by the T3REs. Results from in vitro binding assays indicate that one of the accessory elements (region F) binds CCAAT/enhancer-binding protein-alpha (C/EBPalpha). Here, we investigated the role of C/EBPalpha in the regulation of malic enzyme transcription by T3. Transfection analyses demonstrated that the stimulation of T3RE function by region F did not require the presence of additional malic enzyme gene promoter sequences. Expression of a dominant negative C/EBP inhibited the ability of region F to stimulate T3 responsiveness. In chromatin immunoprecipitation assays, C/EBPalpha and TR associated with the malic enzyme T3RU in the absence and presence of T3 with the extent of the association being greater in the presence of T3. These observations indicate that C/EBPalpha interacts with TR on the malic enzyme T3RU to enhance T3 regulation of malic enzyme gene transcription. T3 treatment increased the acetylation of histones, decreased the recruitment of nuclear receptor corepressor and increased the recruitment of steroid receptor coactivator-1, CREB binding protein, and the thyroid hormone associated protein/mediator complex at the malic enzyme T3RU. In contrast, T3 treatment had no effect on the acetylation of histones and the recruitment of corepressors and coactivators at the T3RU that mediates the T3 activation of acetyl-CoA carboxylase-alpha gene transcription. We propose that differences between the malic enzyme T3RU and the ACCalpha T3RU in the ability of T3 to modulate histone acetylation and coregulatory protein recruitment are due to differences in the composition of the nuclear receptor complexes that bind these regulatory regions.
Collapse
Affiliation(s)
- Liya Yin
- Department of Biochemistry and Molecular Pharmacology, School of Medicine, P.O. Box 9142, West Virginia University, Morgantown, 26506-9142, USA
| | | | | | | | | |
Collapse
|
46
|
Sabol SL, Brewer HB, Santamarina-Fojo S. The human ABCG1 gene: identification of LXR response elements that modulate expression in macrophages and liver. J Lipid Res 2005; 46:2151-67. [PMID: 16024918 DOI: 10.1194/jlr.m500080-jlr200] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ABC transporter ABCG1 (ATP binding cassette transporter G1), expressed in macrophages, liver, and other tissues, has been implicated in the efflux of cholesterol to high density lipoprotein. The ABCG1 gene is transcriptionally activated by cholesterol loading and activators of liver X receptors (LXRs) and retinoid X receptors (RXRs) through genomic sequences that have not been fully characterized. Here we show that ABCG1 mRNA is induced by LXR agonists in RAW264.7 macrophage cells, HepG2 hepatoma cells, and primary mouse hepatocytes. We identify two evolutionarily highly conserved LXR response elements (LXREs), LXRE-A and LXRE-B, located in the first and second introns of the human ABCG1 gene. Each element conferred robust LXR-agonist responsiveness to ABCG1 promoter-directed luciferase gene constructs in RAW264.7 and HepG2 cells. Overexpression of LXR/RXR activated the ABCG1 promoter in the presence of LXRE-A or LXRE-B sequences. In gel-shift assays, LXR/RXR heterodimers bound to wild-type but not to mutated LXRE-A and LXRE-B sequences. In chromatin immunoprecipitation assays, LXR and RXR were detected at LXRE-A and -B regions of DNA of human THP-1 macrophages. These studies clarify the mechanism of transcriptional upregulation of the ABCG1 gene by oxysterols in macrophages and liver, two key tissues where ABCG1 expression may affect cholesterol balance and atherogenesis.
Collapse
Affiliation(s)
- Steven L Sabol
- Molecular Disease Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
47
|
Schmitz G, Langmann T. Transcriptional regulatory networks in lipid metabolism control ABCA1 expression. Biochim Biophys Acta Mol Cell Biol Lipids 2005; 1735:1-19. [PMID: 15922656 DOI: 10.1016/j.bbalip.2005.04.004] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Revised: 04/22/2005] [Accepted: 04/22/2005] [Indexed: 10/25/2022]
Abstract
The ATP-binding cassette transporters, ABCA1 and ABCG1, are major players in mediating cellular efflux of phospholipids and cholesterol to apoA-I containing lipoproteins including prebeta-HDL and alphaHDL and thereby exert important antiatherogenic properties. Although the exact mechanisms how ABC transporters mediate lipid transport are not completely resolved, recent evidence from several laboratories including ours suggests that vesicular transport processes involving different interactive proteins like beta2-syntrophin, alpha1-syntrophin, Lin7, and cdc42 are critically involved in cellular lipid homeostasis controlled by ABCA1 and ABCG1. Besides sterols and fatty acids as known physiological modulators of the LXR/RXR and SREBP pathways, a growing list of natural and synthetic substances and metabolic regulators such as retinoids, PPAR-ligands, hormones, cytokines, and drugs are particularly effective in modulating ABCA1 and ABCG1 gene expression. Although ABCA1 protein amounts are regulated at the level of stability, the majority of potent activating and repressing mechanisms on ABCA1 function directly act on the ABCA1 gene promoter. Among the inducing factors, liver-X-receptors (LXR), retinoic acid receptors (RAR) and peroxisome proliferator-activated receptors (PPARs) along with their coactivators provide an amplification loop for ABCA1 and ABCG1 expression. The ABCA1 promoter is further stimulated by the ubiquitous factor Sp1 and the hypoxia-induced factor 1 (HIF1), which bind to GC-boxes and the E-box, respectively. Shutdown of ABCA1 expression in the absence of sterols or in certain tissues is mediated by corepressor complexes involving unliganded LXR, sterol-regulatory element binding protein 2 (SREBP2), Sp3, and the SCAN-domain protein ZNF202, which also impacts nuclear receptor signaling. Thus, a highly sophisticated transcriptional network controls the balanced expression of ABCA1.
Collapse
Affiliation(s)
- Gerd Schmitz
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93042 Regensburg, Germany.
| | | |
Collapse
|
48
|
Gerin I, Dolinsky VW, Shackman JG, Kennedy RT, Chiang SH, Burant CF, Steffensen KR, Gustafsson JA, MacDougald OA. LXRβ Is Required for Adipocyte Growth, Glucose Homeostasis, and β Cell Function. J Biol Chem 2005; 280:23024-31. [PMID: 15831500 DOI: 10.1074/jbc.m412564200] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Liver X receptors (LXR) alpha and beta are nuclear oxysterol receptors with established roles in cholesterol, lipid, and carbohydrate metabolism. Although LXRs have been extensively studied in liver and macrophages, the importance for development and metabolism of other tissues and cell types is not as well characterized. We demonstrate here that although LXRalpha and LXRbeta are not required for adipocyte development per se, LXRbeta is required for the increase in adipocyte size that normally occurs with aging and diet-induced obesity. Similar food intake and oxygen consumption in LXRbeta-/- mice suggests that reduced storage of lipid in adipose tissue is not due to altered energy balance. Despite reduced amounts of adipose tissue, LXRbeta-/- mice on a chow diet have insulin sensitivity and levels of adipocyte hormones similar to wild type mice. However, these mice are glucose-intolerant due to impaired glucose-induced insulin secretion. Lipid droplets in pancreatic islets may result from accumulation of cholesterol esters as analysis of islet gene expression reveals that LXRbeta is required for expression of the cholesterol transporters, ABCA1 and ABCG1. Our data establish novel roles for LXRbeta in adipocyte growth, glucose homeostasis, and beta cell function.
Collapse
Affiliation(s)
- Isabelle Gerin
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, 48109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Huuskonen J, Vishnu M, Fielding PE, Fielding CJ. Activation of ATP-Binding Cassette Transporter A1 Transcription by Chromatin Remodeling Complex. Arterioscler Thromb Vasc Biol 2005; 25:1180-5. [PMID: 15774904 DOI: 10.1161/01.atv.0000163186.58462.c5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Liver X receptor (LXR) regulates the transcription of ATP-binding cassette transporter A1 (ABCA1) by binding to the DR-4 promoter element as a heterodimer with retinoid X receptor (RXR). The role of chromatin remodeling complex in LXR or ABCA1 activation has not been established previously. In this study, we investigated the activation of ABCA1 by brahma-related gene 1 (BRG-1) and brahma, members of the SWI/SNF (mating type switching/sucrose nonfermenting) chromatin remodeling complex.
Methods and Results—
Overexpression of wild-type BRG-1 in SW-13 cells, but not a catalytically inactive mutant, increased ABCA1 mRNA levels determined by RT-PCR. These effects were enhanced by LXR and RXR agonists. In 293T (epithelial kidney cell line) and Hep3B (hepatocyte cell line) cells, small interfering RNA against BRG-1/brm also affected ABCA1 mRNA levels. Synergistic activation of ABCA1 was obtained after coexpressing BRG-1 and SRC-1, a coactivator of LXR. Luciferase assays showed that this activation of ABCA1 was dependent on the promoter DR-4 element. Coimmunoprecipitation and chromatin immunoprecipitation studies indicated that the mechanism of BRG-1–mediated activation of ABCA1 involved interaction of LXR/RXR with BRG-1 and binding of this complex to ABCA1 promoter.
Conclusions—
Catalytic subunits of SWI/SNF chromatin remodeling complex, BRG-1 and brahma, play significant roles in enhancing LXR/RXR–mediated transcription of ABCA1 via the promoter DR-4 element.
Collapse
Affiliation(s)
- Jarkko Huuskonen
- Cardiovascular Research Institute, University of California San Francisco, CA 94143, USA.
| | | | | | | |
Collapse
|
50
|
Terasaka N, Hiroshima A, Ariga A, Honzumi S, Koieyama T, Inaba T, Fujiwara T. Liver X receptor agonists inhibit tissue factor expression in macrophages. FEBS J 2005; 272:1546-56. [PMID: 15752369 DOI: 10.1111/j.1742-4658.2005.04599.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Exposure of blood to tissue factor (TF) rapidly initiates the coagulation serine protease cascades. TF is expressed by macrophages and other types of cell within atherosclerotic lesions and plays an important role in thrombus formation after plaque rupture. Macrophage TF expression is induced by pro-inflammatory stimuli including lipopolysaccharide (LPS), interleukin-1beta and tumor necrosis factor-alpha. Here we demonstrate that activation of liver X receptors (LXRs) LXRalpha and LXRbeta suppresses TF expression. Treatment of mouse peritoneal macrophages with synthetic LXR agonist T0901317 or GW3965 reduced TF expression induced by pro-inflammatory stimuli. LXR agonists also suppressed TF expression and its activity in human monocytes. Human and mouse TF promoters contain binding sites for the transcription factors AP-1, NFkappaB, Egr-1 and Sp1, but no LXR-binding sites could be found. Cotransfection assays with LXR and TF promoter constructs in RAW 264.7 cells revealed that LXR agonists suppressed LPS-induced TF promoter activity. Analysis of TF promoter also showed that inhibition of TF promoter activity by LXR was at least in part through inhibition of the NFkappaB signaling pathway. In addition, in vivo, LXR agonists reduced TF expression within aortic lesions in an atherosclerosis mouse model as well as in kidney and lung in mice stimulated with LPS. These findings indicate that activation of LXR results in reduction of TF expression, which may influence atherothrombosis in patients with vascular disease.
Collapse
Affiliation(s)
- Naoki Terasaka
- Pharmacology and Molecular Biology Research Laboratories, Sankyo Co. Ltd, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|