1
|
Wu Y, Jing H, Li Y, Li M, Zheng Y, Lin Y, Ma G, Cao H, Yang H. NOR1 promotes the osteoblastic differentiation of human periodontal ligament stem cells via TGF-β signaling pathway. Cell Mol Life Sci 2024; 81:338. [PMID: 39120703 PMCID: PMC11335260 DOI: 10.1007/s00018-024-05356-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
Alveolar bone loss is a main manifestation of periodontitis. Human periodontal ligament stem cells (PDLSCs) are considered as optimal seed cells for alveolar bone regeneration due to its mesenchymal stem cell like properties. Osteogenic potential is the premise for PDLSCs to repair alveolar bone loss. However, the mechanism regulating osteogenic differentiation of PDLSCs remain elusive. In this study, we identified Neuron-derived orphan receptor 1 (NOR1), was particularly expressed in PDL tissue in vivo and gradually increased during osteogenic differentiation of PDLSCs in vitro. Knockdown of NOR1 in hPDLSCs inhibited their osteogenic potential while NOR1 overexpression reversed this effect. In order to elucidate the downstream regulatory network of NOR1, RNA-sequencing was used. We found that downregulated genes were mainly enriched in TGF-β, Hippo, Wnt signaling pathway. Further, by western blot analysis, we verified that the expression level of phosphorylated-SMAD2/3 and phosphorylated-SMAD4 were all decreased after NOR1 knockdown. Additionally, ChIP-qPCR and dual luciferase reporter assay indicated that NOR1 could bind to the promoter of TGFBR1 and regulate its activity. Moreover, overexpression of TGFBR1 in PDLSCs could rescue the damaged osteogenic potential after NOR1 knockdown. Taken together, our results demonstrated that NOR1 could activate TGF-β/SMAD signaling pathway and positively regulates the commitment of osteoblast lineages of PDLSCs by targeting TGFBR1 directly.
Collapse
Affiliation(s)
- Yun Wu
- The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518000, China
- Guangdong Provincial High-level Clinical Key Specialty, Shenzhen, Guangdong, 518000, China
- Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong, 518000, China
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518000, China
| | - Huan Jing
- The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518000, China
- Guangdong Provincial High-level Clinical Key Specialty, Shenzhen, Guangdong, 518000, China
- Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong, 518000, China
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518000, China
| | - Yicun Li
- The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518000, China
- Guangdong Provincial High-level Clinical Key Specialty, Shenzhen, Guangdong, 518000, China
- Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong, 518000, China
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518000, China
| | - Mengqing Li
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong , 518000, China
| | - Yating Zheng
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong , 518000, China
| | - Yuntao Lin
- The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518000, China
- Guangdong Provincial High-level Clinical Key Specialty, Shenzhen, Guangdong, 518000, China
- Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong, 518000, China
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518000, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| | - Hongyu Yang
- The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518000, China.
- Guangdong Provincial High-level Clinical Key Specialty, Shenzhen, Guangdong, 518000, China.
- Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong, 518000, China.
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518000, China.
| |
Collapse
|
2
|
Kumar R, Chhillar N, Gupta DS, Kaur G, Singhal S, Chauhan T. Cholesterol Homeostasis, Mechanisms of Molecular Pathways, and Cardiac Health: A Current Outlook. Curr Probl Cardiol 2024; 49:102081. [PMID: 37716543 DOI: 10.1016/j.cpcardiol.2023.102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
The metabolism of lipoproteins, which regulate the transit of the lipid to and from tissues, is crucial to maintaining cholesterol homeostasis. Cardiac remodeling is referred to as a set of molecular, cellular, and interstitial changes that, following injury, affect the size, shape, function, mass, and geometry of the heart. Acetyl coenzyme A (acetyl CoA), which can be made from glucose, amino acids, or fatty acids, is the precursor for the synthesis of cholesterol. In this article, the authors explain concepts behind cardiac remodeling, its clinical ramifications, and the pathophysiological roles played by numerous various components, such as cell death, neurohormonal activation, oxidative stress, contractile proteins, energy metabolism, collagen, calcium transport, inflammation, and geometry. The levels of cholesterol are traditionally regulated by 2 biological mechanisms at the transcriptional stage. First, the SREBP transcription factor family regulates the transcription of crucial rate-limiting cholesterogenic and lipogenic proteins, which in turn limits cholesterol production. Immune cells become activated, differentiated, and divided, during an immune response with the objective of eradicating the danger signal. In addition to creating ATP, which is used as energy, this process relies on metabolic reprogramming of both catabolic and anabolic pathways to create metabolites that play a crucial role in regulating the response. Because of changes in signal transduction, malfunction of the sarcoplasmic reticulum and sarcolemma, impairment of calcium handling, increases in cardiac fibrosis, and progressive loss of cardiomyocytes, oxidative stress appears to be the primary mechanism that causes the transition from cardiac hypertrophy to heart failure. De novo cholesterol production, intestinal cholesterol absorption, and biliary cholesterol output are consequently crucial processes in cholesterol homeostasis. In the article's final section, the pharmacological management of cardiac remodeling is explored. The route of treatment is explained in different steps: including, promising, and potential strategies. This chapter offers a brief overview of the history of the study of cholesterol absorption as well as the different potential therapeutic targets.
Collapse
Affiliation(s)
| | - Neelam Chhillar
- Deparetment of Biochemistry, School of Medicine, DY Patil University, Navi Mumbai, India
| | - Dhruv Sanjay Gupta
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Ginpreet Kaur
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Shailey Singhal
- Cluster of Applied Sciences, University of Petroleum and Energy Studies, Dehradun, India
| | - Tanya Chauhan
- Division of Forensic Biology, National Forensic Sciences University, Delhi Campus (LNJN NICFS) Delhi, India
| |
Collapse
|
3
|
Zhao L, Lei W, Deng C, Wu Z, Sun M, Jin Z, Song Y, Yang Z, Jiang S, Shen M, Yang Y. The roles of liver X receptor α in inflammation and inflammation-associated diseases. J Cell Physiol 2020; 236:4807-4828. [PMID: 33305467 DOI: 10.1002/jcp.30204] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/19/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
Liver X receptor α (LXRα; also known as NR1H3), an isoform of LXRs, is a member of the nuclear receptor family of transcription factors and plays essential roles in the transcriptional control of cholesterol homeostasis. Previous in-depth phenotypic analyses of mouse models with deficient LXRα have also demonstrated various physiological functions of this receptor within inflammatory responses. LXRα activation exerts a combination of metabolic and anti-inflammatory actions resulting in the modulation and the amelioration of inflammatory disorders. The tight "repercussions" between LXRα and inflammation, as well as cholesterol homeostasis, have suggested that LXRα could be pharmacologically targeted in pathologies such as atherosclerosis, acute lung injury, and Alzheimer's disease. This review gives an overview of the recent advances in understanding the roles of LXRα in inflammation and inflammation-associated diseases, which will help in the design of future experimental researches on the potential of LXRα and advance the investigation of LXRα as pharmacological inflammatory targets.
Collapse
Affiliation(s)
- Lin Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China.,Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wangrui Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhen Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| | - Meng Sun
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yanbin Song
- Department of Cardiology, Affiliated Hospital, Yan'an University, China
| | - Zhi Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| | - Shuai Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| | - Mingzhi Shen
- Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Sanya, Hainan, China.,Hainan Branch of National Clinical Reasearch Center of Geriatrics Disease, Sanya, Hainan, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| |
Collapse
|
4
|
Poirot M, Silvente-Poirot S. The tumor-suppressor cholesterol metabolite, dendrogenin A, is a new class of LXR modulator activating lethal autophagy in cancers. Biochem Pharmacol 2018; 153:75-81. [PMID: 29409832 DOI: 10.1016/j.bcp.2018.01.046] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 01/29/2018] [Indexed: 12/31/2022]
Abstract
Dendrogenin A (DDA) is a mammalian cholesterol metabolite recently identified that displays tumor suppressor properties. The discovery of DDA has revealed the existence in mammals of a new metabolic branch in the cholesterol pathway centered on 5,6α-epoxycholesterol and bridging cholesterol metabolism with histamine metabolism. Metabolic studies showed a drop in DDA levels in cancer cells and tumors compared to normal cells, suggesting a link between DDA metabolism deregulation and oncogenesis. Importantly, complementation of cancer cells with DDA induced 1) cancer cell re-differentiation, 2) blockade of 6-oxo-cholestan-3β,5α-diol (OCDO) production, an endogenous tumor promoter and 3) lethal autophagy in tumors. Importantly, by binding the liver X receptor (LXR), DDA activates the expression of genes controlling autophagy. These genes include NR4A1, NR4A3, LC3 and TFEB. The canonical LXR ligands 22(R)hydroxycholesterol, TO901317 and GW3965 did not induce these effects indicating that DDA delineates a new class of selective LXR modulator (SLiM). The induction of lethal autophagy by DDA was associated with the accumulation in cancer cells of lysosomes and of the pro-lysosomal cholesterol precursor zymostenol due to the inhibition of the 3β-hydroxysteroid-Δ8Δ7-isomerase enzyme (D8D7I). The anti-cancer efficacy of DDA was established on different mouse and human cancers such as breast cancers, melanoma and acute myeloid leukemia, including patient derived xenografts, and did not discriminate bulk cancer cells from cancer cell progenitors. Together these data highlight that the mammalian metabolite DDA is a promising anticancer compound with a broad range of anticancer applications. In addition, DDA and LXR are new actors in the transcriptional control of autophagy and DDA being a "first in line" driver of lethal autophagy in cancers via the LXR.
Collapse
Affiliation(s)
- Marc Poirot
- Team "Cholesterol Metabolism and Therapeutic Innovations", Cancer Research Center of Toulouse, UMR 1037 INSERM-University of Toulouse, Toulouse, France; Cancer Research Center of Toulouse, UMR 1037 INSERM-University of Toulouse, Toulouse, France.
| | - Sandrine Silvente-Poirot
- Team "Cholesterol Metabolism and Therapeutic Innovations", Cancer Research Center of Toulouse, UMR 1037 INSERM-University of Toulouse, Toulouse, France; Cancer Research Center of Toulouse, UMR 1037 INSERM-University of Toulouse, Toulouse, France.
| |
Collapse
|
5
|
Dendrogenin A drives LXR to trigger lethal autophagy in cancers. Nat Commun 2017; 8:1903. [PMID: 29199269 PMCID: PMC5712521 DOI: 10.1038/s41467-017-01948-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 10/27/2017] [Indexed: 01/09/2023] Open
Abstract
Dendrogenin A (DDA) is a newly discovered cholesterol metabolite with tumor suppressor properties. Here, we explored its efficacy and mechanism of cell death in melanoma and acute myeloid leukemia (AML). We found that DDA induced lethal autophagy in vitro and in vivo, including primary AML patient samples, independently of melanoma Braf status or AML molecular and cytogenetic classifications. DDA is a partial agonist on liver-X-receptor (LXR) increasing Nur77, Nor1, and LC3 expression leading to autolysosome formation. Moreover, DDA inhibited the cholesterol biosynthesizing enzyme 3β-hydroxysterol-Δ8,7-isomerase (D8D7I) leading to sterol accumulation and cooperating in autophagy induction. This mechanism of death was not observed with other LXR ligands or D8D7I inhibitors establishing DDA selectivity. The potent anti-tumor activity of DDA, its original mechanism of action and its low toxicity support its clinical evaluation. More generally, this study reveals that DDA can direct control a nuclear receptor to trigger lethal autophagy in cancers.
Collapse
|
6
|
Eicosapentaenoic acid regulates brown adipose tissue metabolism in high-fat-fed mice and in clonal brown adipocytes. J Nutr Biochem 2016; 39:101-109. [PMID: 27833050 DOI: 10.1016/j.jnutbio.2016.08.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 08/03/2016] [Accepted: 08/20/2016] [Indexed: 01/08/2023]
Abstract
Brown adipose tissue (BAT) plays a key role in energy expenditure through its specialized thermogenic function. Therefore, BAT activation may help prevent and/or treat obesity. Interestingly, subcutaneous white adipose tissue (WAT) also has the ability to differentiate into brown-like adipocytes and may potentially contribute to increased thermogenesis. We have previously reported that eicosapentaenoic acid (EPA) reduces high-fat (HF)-diet-induced obesity and insulin resistance in mice. Whether BAT mediates some of these beneficial effects of EPA has not been determined. We hypothesized that EPA activates BAT thermogenic program, contributing to its antiobesity effects. BAT and WAT were harvested from B6 male mice fed HF diets supplemented with or without EPA. HIB 1B clonal brown adipocytes treated with or without EPA were also used. Gene and protein expressions were measured in adipose tissues and H1B 1B cells by quantitative polymerase chain reaction and immunoblotting, respectively. Our results show that BAT from EPA-supplemented mice expressed significantly higher levels of thermogenic genes such as PRDM16 and PGC1α and higher levels of uncoupling protein 1 compared to HF-fed mice. By contrast, both WATs (subcutaneous and visceral) had undetectable levels of these markers with no up regulation by EPA. HIB 1B cells treated with EPA showed significantly higher mRNA expression of PGC1α and SIRT2. EPA treatment significantly increased maximum oxidative and peak glycolytic metabolism in H1B 1B cells. Our results demonstrate a novel and promising role for EPA in preventing obesity via activation of BAT, adding to its known beneficial anti-inflammatory effects.
Collapse
|
7
|
HE KUN, DAI ZHUOYA, LI PEIZHI, ZHU XIWEN, GONG JIANPING. Association between liver X receptor-α and neuron-derived orphan nuclear receptor-1 in Kupffer cells of C57BL/6 mice during inflammation. Mol Med Rep 2015; 12:6098-104. [DOI: 10.3892/mmr.2015.4155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 06/15/2015] [Indexed: 11/05/2022] Open
|
8
|
Mottillo EP, Granneman JG. Intracellular fatty acids suppress β-adrenergic induction of PKA-targeted gene expression in white adipocytes. Am J Physiol Endocrinol Metab 2011; 301:E122-31. [PMID: 21505145 PMCID: PMC3129835 DOI: 10.1152/ajpendo.00039.2011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
β-Adrenergic receptor (β-AR) activation elevates cAMP levels in fat cells and triggers both metabolic and transcriptional responses; however, the potential interactions between these pathways are poorly understood. This study investigated whether lipolysis affects β-AR-mediated gene expression in adipocytes. Acute β(3)-adrenergic receptor (β(3)-AR) stimulation with CL 316,243 (CL) increased expression of PKA-targeted genes PCG-1α, UCP1, and NOR-1 in mouse white fat. Limiting lipolysis via inhibition of hormone-sensitive lipase (HSL), a direct target of PKA, sharply potentiated CL induction of PCG-1α, UCP1, and NOR-1. CL also induced greater expression of PKA-targeted genes in white fat of HSL-null mice compared with wild-type littermates, further indicating that HSL activity limits PKA-mediated gene expression. Inhibiting HSL in 3T3-L1 adipocytes also potentiated the induction of PGC-1α, UCP1, and NOR-1 by β-AR activation, as did siRNA knockdown of adipose triglyceride lipase, the rate-limiting enzyme for lipolysis. Conversely, treatments that promote intracellular fatty acid accumulation suppressed induction of PGC-1α and UCP1 through β-AR stimulation. Analysis of β-adrenergic signaling indicated that excessive intracellular fatty acid production inhibits adenylyl cyclase activity and thereby reduces PKA signaling to the nucleus. Lastly, partially limiting lipolysis by inhibition of HSL increased the induction of oxidative gene expression and mitochondrial electron transport chain activity in white adipose tissue and facilitated fat loss in mice treated for 5 days with CL. Overall, our results demonstrate that fatty acids limit the upregulation of β-AR-responsive genes in white adipocytes and suggest that limiting lipolysis may be a novel means of enhancing β-AR signaling.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Animals
- Coenzyme A Ligases/antagonists & inhibitors
- Cyclic AMP-Dependent Protein Kinases/physiology
- Down-Regulation/drug effects
- Enzyme Inhibitors/pharmacology
- Fatty Acids/metabolism
- Fatty Acids/pharmacology
- Female
- Gene Expression/drug effects
- Intracellular Space/metabolism
- Lipolysis/drug effects
- Lipolysis/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Receptors, Adrenergic, beta/genetics
- Receptors, Adrenergic, beta/metabolism
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/metabolism
- Triazenes/pharmacology
Collapse
Affiliation(s)
- Emilio P Mottillo
- Center for Integrative Metabolic and Endocrine Research, Department of Pathology and Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | |
Collapse
|
9
|
Wang SCM, Myers SA, Eriksson NA, Fitzsimmons RL, Muscat GEO. Nr4a1 siRNA expression attenuates α-MSH regulated gene expression in 3T3-L1 adipocytes. Mol Endocrinol 2011; 25:291-306. [PMID: 21239615 DOI: 10.1210/me.2010-0231] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Several recent investigations have underscored the growing role of melanocortin signaling in the peripheral regulation of lipid, glucose, and energy homeostasis. In addition, the melanocortins play a critical role in the central control of satiety. These observations, and the latest reports highlighting the emerging role of the nuclear hormone receptor (NR) 4A subgroup in metabolism, have prompted us to investigate the cross talk between [Nle(4), d-Phe(7)] (NDP)-α-MSH and Nr4a signaling in adipose. We have shown that NDP-MSH strikingly and preferentially induces the expression of the NR4A subgroup (but not any other members of the NR superfamily) in differentiated 3T3-L1 adipocytes. Utilization of quantitative PCR on custom-designed metabolic TaqMan low-density arrays identified the concomitant and marked induction of the mRNAs encoding Il-6, Cox2, Pdk4, and Pck-1 after NDP-MSH treatment. Similar experiments demonstrated that the mRNA expression profile induced by cAMP and NDP-MSH treatment displayed unique but also overlapping properties and suggested that melanocortin-mediated induction of gene expression involves cAMP-dependent and -independent signaling. Nr4a1/Nur77 small interfering RNA (siRNA) expression suppressed NDP-MSH-mediated induction of Nr4a1/Nur77 and Nr4a3/Nor-1 (but not Nr4a2/Nurr1). Moreover, expression of the siRNA-attenuated NDP-MSH mediated induction of the mRNAs encoding Il-6, Cox2/Ptgs2, and Pck-1 expression. In addition, Nur77 siRNA expression attenuated NDP-MSH-mediated glucose uptake. In vivo, ip administration of NDP-MSH to C57 BL/6J (male) mice significantly induced the expression of the mRNA encoding Nur77 and increased IL-6, Cox2, Pck1, and Pdk4 mRNA expression in (inguinal) adipose tissue. We conclude that Nur77 expression is necessary for MSH-mediated induction of gene expression in differentiated adipocytes. Furthermore, this study demonstrates cross talk between MSH and Nr4a signaling in adipocytes.
Collapse
Affiliation(s)
- S-C Mary Wang
- Obesity Research Centre, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | |
Collapse
|
10
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2010; 17:177-85. [PMID: 20190584 DOI: 10.1097/med.0b013e3283382286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|