1
|
Goldberg D, Buchshtab N, Charni-Natan M, Goldstein I. Transcriptional cascades during fasting amplify gluconeogenesis and instigate a secondary wave of ketogenic gene transcription. Liver Int 2024; 44:2964-2982. [PMID: 39162082 DOI: 10.1111/liv.16077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND AND AIMS During fasting, bodily homeostasis is maintained due to hepatic production of glucose (gluconeogenesis) and ketone bodies (ketogenesis). The main hormones governing hepatic fuel production are glucagon and glucocorticoids that initiate transcriptional programs aimed at supporting gluconeogenesis and ketogenesis. METHODS Using primary mouse hepatocytes as an ex vivo model, we employed transcriptomic analysis (RNA-seq), genome-wide profiling of enhancer dynamics (ChIP-seq), perturbation experiments (inhibitors, shRNA), hepatic glucose production measurements and computational analyses. RESULTS We found that in addition to the known metabolic genes transcriptionally induced by glucagon and glucocorticoids, these hormones induce a set of genes encoding transcription factors (TFs) thereby initiating transcriptional cascades. Upon activation by glucocorticoids, the glucocorticoid receptor (GR) induced the genes encoding two TFs: CCAAT/enhancer-binding protein beta (C/EBPβ) and peroxisome proliferator-activated receptor alpha (PPARα). We found that the GR-C/EBPβ cascade mainly serves as a secondary amplifier of primary hormone-induced gene programs. C/EBPβ augmented gluconeogenic gene expression and hepatic glucose production. Conversely, the GR-PPARα cascade initiated a secondary transcriptional wave of genes supporting ketogenesis. The cascade led to synergistic induction of ketogenic genes which is dependent on protein synthesis. Genome-wide analysis of enhancer dynamics revealed numerous enhancers activated by the GR-PPARα cascade. These enhancers were proximal to ketogenic genes, enriched for the PPARα response element and showed increased PPARα binding. CONCLUSION This study reveals abundant transcriptional cascades occurring during fasting. These cascades serve two separated purposes: the amplification of the gluconeogenic transcriptional program and the induction of a gene program aimed at enhancing ketogenesis.
Collapse
Affiliation(s)
- Dana Goldberg
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Nufar Buchshtab
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Meital Charni-Natan
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ido Goldstein
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
2
|
Lou S, Li T, Kong X, Zhang J, Liu J, Lee D, Gerstein M. TopicNet: a framework for measuring transcriptional regulatory network change. Bioinformatics 2021; 36:i474-i481. [PMID: 32657410 PMCID: PMC7355251 DOI: 10.1093/bioinformatics/btaa403] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Motivation Recently, many chromatin immunoprecipitation sequencing experiments have been carried out for a diverse group of transcription factors (TFs) in many different types of human cells. These experiments manifest large-scale and dynamic changes in regulatory network connectivity (i.e. network ‘rewiring’), highlighting the different regulatory programs operating in disparate cellular states. However, due to the dense and noisy nature of current regulatory networks, directly comparing the gains and losses of targets of key TFs across cell states is often not informative. Thus, here, we seek an abstracted, low-dimensional representation to understand the main features of network change. Results We propose a method called TopicNet that applies latent Dirichlet allocation to extract functional topics for a collection of genes regulated by a given TF. We then define a rewiring score to quantify regulatory-network changes in terms of the topic changes for this TF. Using this framework, we can pinpoint particular TFs that change greatly in network connectivity between different cellular states (such as observed in oncogenesis). Also, incorporating gene expression data, we define a topic activity score that measures the degree to which a given topic is active in a particular cellular state. And we show how activity differences can indicate differential survival in various cancers. Availability and Implementation The TopicNet framework and related analysis were implemented using R and all codes are available at https://github.com/gersteinlab/topicnet. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Shaoke Lou
- Department of Molecular Biophysics and Biochemistry
| | - Tianxiao Li
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | | | - Jing Zhang
- Department of Molecular Biophysics and Biochemistry
| | - Jason Liu
- Department of Molecular Biophysics and Biochemistry
| | - Donghoon Lee
- Department of Molecular Biophysics and Biochemistry
| | - Mark Gerstein
- Department of Molecular Biophysics and Biochemistry
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
3
|
Chen D, Chou FJ, Chen Y, Tian H, Wang Y, You B, Niu Y, Huang CP, Yeh S, Xing N, Chang C. Targeting the radiation-induced TR4 nuclear receptor-mediated QKI/circZEB1/miR-141-3p/ZEB1 signaling increases prostate cancer radiosensitivity. Cancer Lett 2020; 495:100-111. [PMID: 32768524 DOI: 10.1016/j.canlet.2020.07.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 12/24/2022]
Abstract
Early studies indicated that the testicular nuclear receptor 4 (TR4) might play key roles in altering prostate cancer (PCa) progression; however, its ability to alter PCa radiosensitivity remains unclear. Here, we found that suppressing TR4 expression promoted radiosensitivity and better suppressed PCa by modulating the protein quaking (QKI)/circZEB1/miR-141-3p/ZEB1 signaling pathway. Mechanism dissection studies revealed that TR4 could transcriptionally increase the RNA-binding protein QKI to increase circZEB1 levels, which then sponges the miR-141-3p to increase the expression of its host gene ZEB1. Preclinical studies with an in vivo mouse model further proved that combining radiation therapy (RT) with metformin promoted radiosensitivity to suppress PCa progression. Together, these results suggest that TR4 may play key roles in altering PCa radiosensitivity and show that targeting this newly identified TR4-mediated QKI/circZEB1/miR-141-3p/ZEB1 signaling pathway may help in the development of a novel RT to better suppress the progression of PCa.
Collapse
Affiliation(s)
- Dong Chen
- Department of Urology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China; George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester, Rochester, 14642, NY, USA
| | - Fu-Ju Chou
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester, Rochester, 14642, NY, USA
| | - Yuhchyau Chen
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester, Rochester, 14642, NY, USA
| | - Hao Tian
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester, Rochester, 14642, NY, USA; Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin Medical University, 300211, Tianjin, China
| | - Yaqin Wang
- Key Laboratory of Cardiovascular Epidemiology and Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037, Beijing, China
| | - Bosen You
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester, Rochester, 14642, NY, USA
| | - Yuanjie Niu
- Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin Medical University, 300211, Tianjin, China
| | - Chi-Ping Huang
- Sex Hormone Research Center, China Medical University, 404, Taichung, Taiwan
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester, Rochester, 14642, NY, USA
| | - Nianzeng Xing
- Department of Urology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester, Rochester, 14642, NY, USA; Key Laboratory of Cardiovascular Epidemiology and Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037, Beijing, China.
| |
Collapse
|
4
|
Mechanism of anti-remodelling action of treprostinil in human pulmonary arterial smooth muscle cells. PLoS One 2018; 13:e0205195. [PMID: 30383775 PMCID: PMC6211661 DOI: 10.1371/journal.pone.0205195] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 09/20/2018] [Indexed: 12/31/2022] Open
Abstract
Treprostinil is applied for pulmonary arterial hypertension (PAH) therapy. However, the mechanism by which the drug achieves its beneficial effects in PAH vessels is not fully understood. This study investigated the effects of treprostinil on PDGF-BB induced remodelling parameters in isolated human pulmonary arterial smooth muscle cells (PASMC) of four PAH patients. The production of TGF-β1, CTGF, collagen type-I and -IV, and of fibronectin were determined by ELISA and PCR. The role of cAMP was determined by ELISA and di-deoxyadenosine treatment. Proliferation was determined by direct cell count. Treprostinil increased cAMP levels dose and time dependently, which was not affected by PDGF-BB. Treprostinil significantly reduced PDGF-BB induced secretion of TGF-β1 and CTGF, both was counteracted when cAMP generation was blocked. Similarly, the PDGF-BB induced proliferation of PASMC was dose dependently reduced by treprostinil through signalling via cAMP—C/EBP-α p42 –p21(WAf1/Cip1). In regards to extracellular matrix remodelling, treprostinil significantly reduced PDGF-BB—TGF-β1—CTGF induced synthesis and deposition of collagen type I and fibronectin, in a cAMP sensitive manner. In contrast, the deposition of collagen IV was not affected. The data suggest that this action of treprostinil in vessel wall remodelling may benefit patients with PAH and may reduce arterial wall remodelling.
Collapse
|
5
|
Targeting the ERK pathway for the treatment of Cushing's disease. Oncotarget 2018; 7:69149-69158. [PMID: 27708250 PMCID: PMC5342466 DOI: 10.18632/oncotarget.12381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/22/2016] [Indexed: 01/21/2023] Open
Abstract
We recently demonstrated that the orphan nuclear receptor testicular receptor 4 (TR4) is a potent regulator of corticotroph tumor growth and hormone secretion. The Ras/Raf/MEK/ERK pathway is commonly overactivated in human tumors and we have demonstrated that corticotroph tumor TR4 is activated by ERK1/2-mediated phosphorylation. We evaluated effects of MEK-162, a selective, non-ATP-competitive allosteric inhibitor of MEK1/2, on murine and human in vitro and in vivo corticotroph tumor proliferation and adrenocorticotrophic hormone (ACTH) secretion. MEK-162 treatment dose-dependently inhibited corticotroph tumor proliferation, induced apoptosis, reduced pro-opiomelanocortin (POMC) mRNA levels and inhibited ACTH secretion in vitro. Similar findings were obtained in human corticotroph tumor primary cultures (n = 5). These actions of MEK-162 were augmented in the presence of TR4 overexpression, suggesting that TR4 levels may serve as a predictive biomarker of MEK-162 corticotroph tumor responsiveness. Additionally, MEK-162 treatment reduced TR4 protein expression and blocked recruitment of TR4 to bind its consensus site on the POMC promoter (−854bp to −637bp), elucidating multiple mechanisms to control TR4 corticotroph tumor actions. In a murine corticotroph tumor in vivo model of Cushing's disease, MEK-162 treatment inhibited tumor growth and reduced tumor-derived circulating plasma ACTH, and corticosterone levels. These results demonstrate the potent actions of MEK-162 to inhibit corticotroph tumor growth and hormone secretion in vitro and in vivo via TR4-dependent and independent mechanisms, and raise the possibility of MEK-162 as a novel therapy for Cushing's disease.
Collapse
|
6
|
PK/PD studies on non-selective PDE inhibitors in rats using cAMP as a marker of pharmacological response. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:1047-1059. [PMID: 28730281 PMCID: PMC5599463 DOI: 10.1007/s00210-017-1406-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/05/2017] [Indexed: 12/12/2022]
Abstract
In recent years, phosphodiesterase (PDE) inhibitors have been frequently tested for the treatment of experimental inflammatory and immune disorders. It is suggested that anti-inflammatory properties of PDE inhibitors are related to their ability to increase cAMP levels. The aim of this study was to verify the hypothesis that cAMP may be a useful marker of pharmacological response following administration of non-selective PDE inhibitors (pentoxifylline and (±)-lisofylline) to endotoxemic rats. Male Wistar rats were administered LPS (1 mg kg−1, i.v.) simultaneously with either compound given at two doses (40 and 80 mg kg−1, i.v.). Levels of cAMP and both compounds in animal plasma were measured by the validated HPLC methods. Pharmacokinetic-pharmacodynamic analysis was performed using basic and modified indirect response (IDR) models II in Phoenix WinNonlin. The results of this study indicate that, in contrast to pentoxifylline, (±)-lisofylline demonstrates a non-linear pharmacokinetics in rats with endotoxemia. In vitro study using human recombinant PDE4B and PDE7A revealed the occurrence of additive interaction between studied compounds. Moreover, (±)-lisofylline is a more potent inhibitor of PDEs compared to pentoxifylline, as evidenced by lower IC50 values. Following administration of both compounds, levels of cAMP in rat plasma increased in a dose-dependent manner. The modified IDR model II better described cAMP levels over time profiles. The validity of the proposed marker was confirmed by measuring plasma TNF-α levels in the studied animals. In conclusion, cAMP may be used in future preclinical and clinical studies of some PDE inhibitors to evaluate the drug concentration–effect relationship.
Collapse
|
7
|
Abstract
Testicular nuclear receptors 2 and 4 (TR2, TR4), also known as NR2C1 and NR2C2, belong to the nuclear receptor superfamily and were first cloned in 1989 and 1994, respectively. Although classified as orphan receptors, several natural molecules, their metabolites, and synthetic compounds including polyunsaturated fatty acids (PUFAs), PUFA metabolites 13-hydroxyoctadecadienoic acid, 15-hydroxyeicosatetraenoic acid, and the antidiabetic drug thiazolidinediones can transactivate TR4. Importantly, many of these ligands/activators can also transactivate peroxisome proliferator-activated receptor gamma (PPARγ), also known as NR1C3 nuclear receptor. Both TR4 and PPARγ can bind to similar hormone response elements (HREs) located in the promoter of their common downstream target genes. However, these two nuclear receptors, even with shared ligands/activators and shared binding ability for similar HREs, have some distinct functions in many diseases they influence. In cancer, PPARγ inhibits thyroid, lung, colon, and prostate cancers but enhances bladder cancer. In contrast, TR4 inhibits liver and prostate cancer initiation but enhances pituitary corticotroph, liver, and prostate cancer progression. In type 2 diabetes, PPARγ increases insulin sensitivity but TR4 decreases insulin sensitivity. In cardiovascular disease, PPARγ inhibits atherosclerosis but TR4 enhances atherosclerosis through increasing foam cell formation. In bone physiology, PPARγ inhibits bone formation but TR4 increases bone formation. Together, the contrasting impact of TR4 and PPARγ on different diseases may raise a critical issue about drug used to target any one of these nuclear receptors.
Collapse
|
8
|
Li L, Dang Q, Xie H, Yang Z, He D, Liang L, Song W, Yeh S, Chang C. Infiltrating mast cells enhance prostate cancer invasion via altering LncRNA-HOTAIR/PRC2-androgen receptor (AR)-MMP9 signals and increased stem/progenitor cell population. Oncotarget 2016; 6:14179-90. [PMID: 25895025 PMCID: PMC4546459 DOI: 10.18632/oncotarget.3651] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/28/2015] [Indexed: 12/14/2022] Open
Abstract
Early studies indicated that selective inflammatory immune cells in the prostate tumor microenvironment might be able to influence prostate cancer (PCa) progression. Here we found treating PCa cells with androgen deprivation therapy (ADT) results in the recruitment of more mast cells, which might then increase PCa cell invasion via down-regulation of AR signals in 4 different PCa cell lines. Mechanism dissection revealed infiltrating mast cells could decrease AR transcription via modulation of the PRC2 complex with LncRNA-HOTAIR at the AR 5' promoter region in PCa cells. The consequences of suppressing AR may then increase PCa cell invasion via increased MMP9 expression and/or increased stem/progenitor cell population. The in vivo mouse model with orthotopically xenografted PCa CWR22Rv1 cells with/without mast cells also confirmed that infiltrating mast cells could increase PCa cell invasion via suppression of AR signals. Together, our results provide a new mechanism for the ADT-enhanced PCa metastasis via altering the infiltrating mast cells to modulate PCa AR-MMP9 signals and/or AR-stem/progenitor cell population. Targeting these newly identified inflammatory mast cells-AR signals may help us to better suppress PCa metastasis at the castration resistant stage.
Collapse
Affiliation(s)
- Lei Li
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Qiang Dang
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Hongjun Xie
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Zhao Yang
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Dalin He
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Liang
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Wenbing Song
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Shuyuan Yeh
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Chawnshang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA.,Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
9
|
Park SS, Choi H, Kim SJ, Chang C, Kim E. CREB/GSK-3β signaling pathway regulates the expression of TR4 orphan nuclear receptor gene. Mol Cell Endocrinol 2016; 423:22-9. [PMID: 26762765 DOI: 10.1016/j.mce.2015.12.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 12/31/2015] [Indexed: 12/24/2022]
Abstract
In this study, we show that reduction of glucose concentration increases TR4 expression in 3T3-L1 cells via stimulation of the GSK-3β-CREB pathway. While GSK-3β and CREB increased TR4 expression in 3T3-L1 cells, inhibition of CREB expression or activity resulted in loss of GSK-3β-mediated enhancement of TR4 expression. In addition, CREB enhanced murine TR4 promoter activity via direct binding to a cAMP response element located in the promoter, and this CREB effect was further strengthened by GSK-3β. Moreover, silencing of TR4 expression by a gene-specific microRNA inhibited CREB-induced lipid accumulation in 3T3-L1 adipocytes, suggesting that TR4 could be a key mediator of CREB-induced lipogenesis.
Collapse
Affiliation(s)
- Sung-Soo Park
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, South Korea
| | - Hojung Choi
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, South Korea
| | - Seung-Jin Kim
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, South Korea
| | - Chawnshang Chang
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology and Radiation Oncology, and Caner Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Eungseok Kim
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, South Korea.
| |
Collapse
|
10
|
Lee YJ, Liu C, Liao M, Sukhova GK, Shirakawa J, Abdennour M, Iamarene K, Andre S, Inouye K, Clement K, Kulkarni RN, Banks AS, Libby P, Shi GP. Deficiency of FcϵR1 Increases Body Weight Gain but Improves Glucose Tolerance in Diet-Induced Obese Mice. Endocrinology 2015; 156:4047-58. [PMID: 26295369 PMCID: PMC4606759 DOI: 10.1210/en.2015-1184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prior studies demonstrated increased plasma IgE in diabetic patients, but the direct participation of IgE in diabetes or obesity remains unknown. This study found that plasma IgE levels correlated inversely with body weight, body mass index, and body fat mass among a population of randomly selected obese women. IgE receptor FcϵR1-deficient (Fcer1a(-/-)) mice and diet-induced obesity (DIO) mice demonstrated that FcϵR1 deficiency in DIO mice increased food intake, reduced energy expenditure, and increased body weight gain but improved glucose tolerance and glucose-induced insulin secretion. White adipose tissue from Fcer1a(-/-) mice showed an increased expression of phospho-AKT, CCAAT/enhancer binding protein-α, peroxisome proliferator-activated receptor-γ, glucose transporter-4 (Glut4), and B-cell lymphoma 2 (Bcl2) but reduced uncoupling protein 1 (UCP1) and phosphorylated c-Jun N-terminal kinase (JNK) expression, tissue macrophage accumulation, and apoptosis, suggesting that IgE reduces adipogenesis and glucose uptake but induces energy expenditure, adipocyte apoptosis, and white adipose tissue inflammation. In 3T3-L1 cells, IgE inhibited the expression of CCAAT/enhancer binding protein-α and peroxisome proliferator-activated receptor-γ, and preadipocyte adipogenesis and induced adipocyte apoptosis. IgE reduced the 3T3-L1 cell expression of Glut4, phospho-AKT, and glucose uptake, which concurred with improved glucose tolerance in Fcer1a(-/-) mice. This study established two novel pathways of IgE in reducing body weight gain in DIO mice by suppressing adipogenesis and inducing adipocyte apoptosis while worsening glucose tolerance by reducing Glut4 expression, glucose uptake, and insulin secretion.
Collapse
Affiliation(s)
- Yun-Jung Lee
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Conglin Liu
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Mengyang Liao
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Galina K Sukhova
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Jun Shirakawa
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Meriem Abdennour
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karine Iamarene
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Sebastien Andre
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karen Inouye
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karine Clement
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Rohit N Kulkarni
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Alexander S Banks
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Peter Libby
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Guo-Ping Shi
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| |
Collapse
|
11
|
Lin SJ, Yang DR, Wang N, Jiang M, Miyamoto H, Li G, Chang C. TR4 nuclear receptor enhances prostate cancer initiation via altering the stem cell population and EMT signals in the PPARG-deleted prostate cells. Oncoscience 2015; 2:142-50. [PMID: 25859557 PMCID: PMC4381707 DOI: 10.18632/oncoscience.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 02/06/2015] [Indexed: 12/13/2022] Open
Abstract
A recent report indicated that the TR4 nuclear receptor might suppress the prostate cancer (PCa) initiation via modulating the DNA damage/repair system. Knocking-out peroxisome proliferator-activated receptor gamma (PPARG), a nuclear receptor that shares similar ligands/activators with TR4, promoted PCa initiation. Here we found 9% of PCa patients have one allele of PPARG deletion. Results from in vitro cell lines and in vivo mouse model indicated that during PCa initiation TR4 roles might switch from suppressor to enhancer in prostate cells when PPARG was deleted or suppressed (by antagonist GW9662). Mechanism dissection found targeting TR4 in the absence of PPARG might alter the stem cell population and epithelial-mesenchymal transition (EMT) signals. Together, these results suggest that whether TR4 can enhance or suppress PCa initiation may depend on the availability of PPARG and future potential therapy via targeting PPARG to battle PPARG-related diseases may need to consider the potential side effects of TR4 switched roles during the PCa initiation.
Collapse
Affiliation(s)
- Shin-Jen Lin
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Dong-Rong Yang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Nancy Wang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Ming Jiang
- Department of Urologic Surgery, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hiroshi Miyamoto
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Gonghui Li
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA ; Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
12
|
Lin SJ, Yang DR, Li G, Chang C. TR4 Nuclear Receptor Different Roles in Prostate Cancer Progression. Front Endocrinol (Lausanne) 2015; 6:78. [PMID: 26074876 PMCID: PMC4445305 DOI: 10.3389/fendo.2015.00078] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/30/2015] [Indexed: 01/03/2023] Open
Abstract
Nuclear receptors are important to maintain the tissue homeostasis. Each receptor is tightly controlled and under a very complicated balance. In this review, we summarize the current findings regarding the nuclear receptor TR4 and its role in prostate cancer (PCa) progression. In general, TR4 can inhibit the PCa carcinogenesis. However, when PPARγ is knocked out, activation of TR4 can have an opposite effect to promote the PCa carcinogenesis. Clinical data also indicates that higher TR4 expression is found in PCa tissues with high Gleason scores compared to those tissues with low Gleason scores. In vitro and in vivo studies show that TR4 can promote PCa progression. Mechanism dissection indicates that TR4 inhibits PCa carcinogenesis through regulating the tumor suppressor ATM to reduce DNA damages. On the other hand, in the absence of PPARγ, TR4 tends to increase the stem cell population and epithelial-mesenchymal transition (EMT) via regulating CCL2, Oct4, EZH2, and miRNA-373-3p expression that results in increased PCa carcinogenesis. In opposition to PCa initiation, TR4 can increase PCa metastasis via modulating the CCL2 signals. Finally, targeting TR4 enhances the chemotherapy and radiation therapy sensitivity in PCa. Together, these data suggest TR4 is a key player to control PCa progression, and targeting TR4 with small molecules may provide us a new and better therapy to suppress PCa progression.
Collapse
Affiliation(s)
- Shin-Jen Lin
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Dong-Rong Yang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Urology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Gonghui Li
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Urology, Sir-Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Sex Hormone Research Center, China Medical University Hospital, Taichung, Taiwan
- *Correspondence: Chawnshang Chang, George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center. University of Rochester Medical Center, Rochester, NY 14642, USA,
| |
Collapse
|
13
|
Liang L, Li L, Tian J, Lee SO, Dang Q, Huang CK, Yeh S, Erturk E, Bushinsky D, Chang LS, He D, Chang C. Androgen receptor enhances kidney stone-CaOx crystal formation via modulation of oxalate biosynthesis & oxidative stress. Mol Endocrinol 2014; 28:1291-303. [PMID: 24956378 PMCID: PMC4116591 DOI: 10.1210/me.2014-1047] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Males develop kidney stones far more frequently than females with a ratio of 2–3:1, suggesting that androgen receptor (AR) signaling might play a key role in the development of nephrolithiasis. Using the cre-loxP system to selectively knock out AR in glyoxylate-induced calcium oxalate (CaOx) crystal mouse models, we found that the mice lacking hepatic AR had less oxalate biosynthesis, which might lead to lower CaOx crystal formation, and that the mice lacking kidney proximal or distal epithelial AR also had lower CaOx crystal formation. We found that AR could directly up-regulate hepatic glycolate oxidase and kidney epithelial NADPH oxidase subunit p22-PHOX at the transcriptional level. This up-regulation might then increase oxalate biosynthesis and oxidative stress that resulted in induction of kidney tubular injury. Targeting AR with the AR degradation enhancer ASC-J9 led to suppression of CaOx crystal formation via modulation of oxalate biosynthesis and oxidative stress in both in vitro and in vivo studies. Taken together, these results established the roles of AR in CaOx crystal formation.
Collapse
Affiliation(s)
- Liang Liang
- Sex Hormone Research Center (L.Liang, L.Li, Q.D., L.S.C., D.H.), Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China; George H. Whipple Laboratory for Cancer Research (L.Liang, L.Li, J.T., S.O.L., Q.D., C.-K.H., S.Y., E.E., D.B., C.C.), Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642; and Sex Hormone Research Center (C.C.), China Medical University/Hospital, Taichung 404, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Liu S, Lin SJ, Li G, Kim E, Chen YT, Yang DR, Tan MHE, Yong EL, Chang C. Differential roles of PPARγ vs TR4 in prostate cancer and metabolic diseases. Endocr Relat Cancer 2014; 21:R279-300. [PMID: 24623743 DOI: 10.1530/erc-13-0529] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ, NR1C3) and testicular receptor 4 nuclear receptor (TR4, NR2C2) are two members of the nuclear receptor (NR) superfamily that can be activated by several similar ligands/activators including polyunsaturated fatty acid metabolites, such as 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, as well as some anti-diabetic drugs such as thiazolidinediones (TZDs). However, the consequences of the transactivation of these ligands/activators via these two NRs are different, with at least three distinct phenotypes. First, activation of PPARγ increases insulin sensitivity yet activation of TR4 decreases insulin sensitivity. Second, PPARγ attenuates atherosclerosis but TR4 might increase the risk of atherosclerosis. Third, PPARγ suppresses prostate cancer (PCa) development and TR4 suppresses prostate carcinogenesis yet promotes PCa metastasis. Importantly, the deregulation of either PPARγ or TR4 in PCa alone might then alter the other receptor's influences on PCa progression. Knocking out PPARγ altered the ability of TR4 to promote prostate carcinogenesis and knocking down TR4 also resulted in TZD treatment promoting PCa development, indicating that both PPARγ and TR4 might coordinate with each other to regulate PCa initiation, and the loss of either one of them might switch the other one from a tumor suppressor to a tumor promoter. These results indicate that further and detailed studies of both receptors at the same time in the same cells/organs may help us to better dissect their distinct physiological roles and develop better drug(s) with fewer side effects to battle PPARγ- and TR4-related diseases including tumor and cardiovascular diseases as well as metabolic disorders.
Collapse
Affiliation(s)
- Su Liu
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Shin-Jen Lin
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Gonghui Li
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Eungseok Kim
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Yei-Tsung Chen
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Dong-Rong Yang
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - M H Eileen Tan
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Eu Leong Yong
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Chawnshang Chang
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, TaiwanGeorge Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| |
Collapse
|
15
|
Lin SJ, Zhang Y, Liu NC, Yang DR, Li G, Chang C. Minireview: Pathophysiological roles of the TR4 nuclear receptor: lessons learned from mice lacking TR4. Mol Endocrinol 2014; 28:805-21. [PMID: 24702179 DOI: 10.1210/me.2013-1422] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Testicular nuclear receptor 4 (TR4), also known as NR2C2, belongs to the nuclear receptor superfamily and shares high homology with the testicular nuclear receptor 2. The natural ligands of TR4 remained unclear until the recent discoveries of several energy/lipid sensors including the polyunsaturated fatty acid metabolites, 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, and their synthetic ligands, thiazolidinediones, used for treatment of diabetes. TR4 is widely expressed throughout the body and particularly concentrated in the testis, prostate, cerebellum, and hippocampus. It has been shown to play important roles in cerebellar development, forebrain myelination, folliculogenesis, gluconeogenesis, lipogenesis, muscle development, bone development, and prostate cancer progression. Here we provide a comprehensive summary of TR4 signaling including its upstream ligands/activators/suppressors, transcriptional coactivators/repressors, downstream targets, and their in vivo functions with potential impacts on TR4-related diseases. Importantly, TR4 shares similar ligands/activators with another key nuclear receptor, peroxisome proliferator-activated receptor γ, which raised several interesting questions about how these 2 nuclear receptors may collaborate with or counteract each other's function in their related diseases. Clear dissection of such molecular mechanisms and their differential roles in various diseases may help researchers to design new potential drugs with better efficacy and fewer side effects to battle TR4 and peroxisome proliferator-activated receptor γ involved diseases.
Collapse
Affiliation(s)
- Shin-Jen Lin
- George Whipple Laboratory for Cancer Research (S.-J.L., Y.Z., N.-C.L., C.C.), Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14646; Department of Urology (D.-R.Y.), the Second Affiliated Hospital of Suzhou University, Suzhou, 215004 China; Chawnshang Chang Liver Cancer Center and Department of Urology (G.L.), Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016 China; and Sex Hormone Research Center (C.C.), China Medical University/Hospital, Taichung, 404 Taiwan
| | | | | | | | | | | |
Collapse
|
16
|
Overexpression of Jazf1 reduces body weight gain and regulates lipid metabolism in high fat diet. Biochem Biophys Res Commun 2013; 444:296-301. [PMID: 24380856 DOI: 10.1016/j.bbrc.2013.12.094] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 12/18/2013] [Indexed: 12/13/2022]
Abstract
Jazf1 is a 27 kDa nuclear protein containing three putative zinc finger motifs that is associated with diabetes mellitus and prostate cancer; however, little is known about the role that this gene plays in regulation of metabolism. Recent evidence indicates that Jazf1 transcription factors bind to the nuclear orphan receptor TR4. This receptor regulates PEPCK, the key enzyme involved in gluconeogenesis. To elucidate Jazf1's role in metabolism, we fed a 60% fat diet for up to 15 weeks. In Jazf1 overexpression mice, weight gain was found to be significantly decreased. The expression of Jazf1 in the liver also suppressed lipid accumulation and decreased droplet size. These results suggest that Jazf1 plays a critical role in the regulation of lipid homeostasis. Finally, Jazf1 may provide a new therapeutic target in the management of obesity and diabetes.
Collapse
|
17
|
Liu S, Yan SJ, Lee YF, Liu NC, Ting HJ, Li G, Wu Q, Chen LM, Chang C. Testicular nuclear receptor 4 (TR4) regulates UV light-induced responses via Cockayne syndrome B protein-mediated transcription-coupled DNA repair. J Biol Chem 2011; 286:38103-38108. [PMID: 21918225 DOI: 10.1074/jbc.m111.259523] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
UV irradiation is one of the major external insults to cells and can cause skin aging and cancer. In response to UV light-induced DNA damage, the nucleotide excision repair (NER) pathways are activated to remove DNA lesions. We report here that testicular nuclear receptor 4 (TR4), a member of the nuclear receptor family, modulates DNA repair specifically through the transcription-coupled (TC) NER pathway but not the global genomic NER pathway. The level of Cockayne syndrome B protein (CSB), a member of the TC-NER pathway, is 10-fold reduced in TR4-deficient mouse tissues, and TR4 directly regulates CSB at the transcriptional level. Moreover, restored CSB expression rescues UV hypersensitivity of TR4-deficient cells. Together, these results indicate that TR4 modulates UV sensitivity by promoting the TC-NER DNA repair pathway through transcriptional regulation of CSB. These results may lead to the development of new treatments for UV light-sensitive syndromes, skin cancer, and aging.
Collapse
Affiliation(s)
- Su Liu
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, New York 14642
| | - Shian-Jang Yan
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, New York 14642
| | - Yi-Fen Lee
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, New York 14642
| | - Ning-Chun Liu
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, New York 14642
| | - Huei-Ju Ting
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, New York 14642
| | - Gonghui Li
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, New York 14642
| | - Qiao Wu
- Key Lab of the Ministry of Education for Cell Biology and Tumor Cell Engineering, Xiamen University, Xiamen 361005, China
| | - Lu-Min Chen
- Sex Hormone Research Center, China Medical University/Hospital, Taichung 404, Taiwan
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, New York 14642; Sex Hormone Research Center, China Medical University/Hospital, Taichung 404, Taiwan.
| |
Collapse
|
18
|
Xie S, Ni J, Lee YF, Liu S, Li G, Shyr CR, Chang C. Increased acetylation in the DNA-binding domain of TR4 nuclear receptor by the coregulator ARA55 leads to suppression of TR4 transactivation. J Biol Chem 2011; 286:21129-36. [PMID: 21515881 DOI: 10.1074/jbc.m110.208181] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The nuclear receptor TR4 is a key regulator for many physiological processes, including growth, development, and metabolism. However, how the transcriptional activity of TR4 is regulated in the absence of ligand(s) remains largely unknown. Here we found that an androgen receptor (AR) coactivator, ARA55, might function as a corepressor to suppress TR4 transactivation. Molecular mechanistic dissection with mutation analysis found that ARA55 could enhance TR4 acetylation at the conserved acetylation sites of lysine 175 and lysine 176 in the DNA-binding domain via recruiting proteins with histone acetyl transferase activity, which might then reduce significantly the TR4 DNA binding activity that resulted in the suppression of TR4 transactivation. These results are in contrast to the classic ARA55 coactivator function to enhance AR transactivation partially via increased AR acetylation in the hinge/ligand-binding domain. Together, these results not only provide a novel functional mechanism showing that acetylation of different nuclear receptors at different domains by coregulator may lead to differential receptor transactivation activity but also provide a new way for small molecules to control TR4 transactivation via altering TR4 acetylation levels, and such small molecules may have potential therapeutic applications in the future.
Collapse
Affiliation(s)
- Shaozhen Xie
- George Whipple Lab for Cancer Research, Department of Pathology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Kang HS, Okamoto K, Kim YS, Takeda Y, Bortner CD, Dang H, Wada T, Xie W, Yang XP, Liao G, Jetten AM. Nuclear orphan receptor TAK1/TR4-deficient mice are protected against obesity-linked inflammation, hepatic steatosis, and insulin resistance. Diabetes 2011; 60:177-88. [PMID: 20864514 PMCID: PMC3012170 DOI: 10.2337/db10-0628] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 09/14/2010] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The nuclear receptor TAK1/TR4/NR2C2 is expressed in several tissues that are important in the control of energy homeostasis. In this study, we investigate whether TAK1 functions as a regulator of lipid and energy homeostasis and has a role in metabolic syndrome. RESEARCH DESIGN AND METHODS We generated TAK1-deficient (TAK1⁻(/)⁻) mice to study the function of TAK1 in the development of metabolic syndrome in aged mice and mice fed a high-fat diet (HFD). (Immuno)histochemical, biochemical, and gene expression profile analyses were performed to determine the effect of the loss of TAK1 expression on lipid homeostasis in liver and adipose tissues. In addition, insulin sensitivity, energy expenditure, and adipose-associated inflammation were compared in wild-type (WT) and TAK1⁻(/)⁻ mice fed a HFD. RESULTS TAK1-deficient (TAK1⁻(/)⁻) mice are resistant to the development of age- and HFD-induced metabolic syndrome. Histo- and biochemical analyses showed significantly lower hepatic triglyceride levels and reduced lipid accumulation in adipose tissue in TAK1⁻(/)⁻ mice compared with WT mice. Gene expression profiling analysis revealed that the expression of several genes encoding proteins involved in lipid uptake and triglyceride synthesis and storage, including Cidea, Cidec, Mogat1, and CD36, was greatly decreased in the liver and primary hepatocytes of TAK1⁻(/)⁻ mice. Restoration of TAK1 expression in TAK1⁻(/)⁻ hepatocytes induced expression of several lipogenic genes. Moreover, TAK1⁻(/)⁻ mice exhibited reduced infiltration of inflammatory cells and expression of inflammatory genes in white adipose tissue, and were resistant to the development of glucose intolerance and insulin resistance. TAK1⁻(/)⁻ mice consume more oxygen and produce more carbon dioxide than WT mice, suggesting increased energy expenditure. CONCLUSIONS Our data reveal that TAK1 plays a critical role in the regulation of energy and lipid homeostasis, and promotes the development of metabolic syndrome. TAK1 may provide a new therapeutic target in the management of obesity, diabetes, and liver steatosis.
Collapse
Affiliation(s)
- Hong Soon Kang
- Cell Biology Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Kyoko Okamoto
- Cell Biology Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Yong-Sik Kim
- Cell Biology Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Yukimasa Takeda
- Cell Biology Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Carl D. Bortner
- Laboratory of Signal Transduction, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Huaixin Dang
- Cell Biology Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Taira Wada
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiao-Ping Yang
- Cell Biology Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Grace Liao
- Cell Biology Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Anton M. Jetten
- Cell Biology Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|