1
|
Lincoln K, Zhou J, Oster H, de Assis LVM. Circadian Gating of Thyroid Hormone Action in Hepatocytes. Cells 2024; 13:1038. [PMID: 38920666 PMCID: PMC11202020 DOI: 10.3390/cells13121038] [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: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Thyroid hormones, thyroxin (T4) and the biologically active triiodothyronine (T3), play important roles in liver metabolic regulation, including fatty acid biosynthesis, beta-oxidation, and cholesterol homeostasis. These functions position TH signaling as a potential target for the treatment of metabolic dysfunction-associated steatotic liver disease (MASLD). Elevated T3 levels in the circulation are associated with increased hepatic lipid turnover, which is also under the control of the circadian clock system. In this study, we developed a cell system to study the impact of hepatocyte circadian rhythms on the metabolic response to T3 treatment under control and steatotic conditions. Synchronized AML-12 circadian reporter hepatocytes were treated with T3 at different circadian phases and metabolic conditions. T3 treatment increased metabolic activity in a dose-independent fashion and had no significant effect on circadian rhythms in AML-12 cells. T3 had marked time-of-treatment-dependent effects on metabolic transcript expression. Steatosis induction altered metabolic transcript expression in AML-12 cells. In this condition, the circadian rhythm period was lengthened, and this effect was independent of T3. Under steatotic conditions, T3 had marked time-of-treatment dependent effects on metabolic transcript expression, which differed from those observed under control conditions. These findings reveal a time-of-day-dependent response of hepatocytes to T3, which is further modulated by the metabolic state. Our data suggest that time has a strong influence on liver TH action, which might be considered when treating MASLD.
Collapse
Affiliation(s)
- Karla Lincoln
- Institute of Neurobiology, Center of Brain Behavior & Metabolism, University of Lübeck, 23562 Lübeck, Germany; (K.L.); (J.Z.)
| | - Jingxuan Zhou
- Institute of Neurobiology, Center of Brain Behavior & Metabolism, University of Lübeck, 23562 Lübeck, Germany; (K.L.); (J.Z.)
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain Behavior & Metabolism, University of Lübeck, 23562 Lübeck, Germany; (K.L.); (J.Z.)
- University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Leonardo Vinicius Monteiro de Assis
- Institute of Neurobiology, Center of Brain Behavior & Metabolism, University of Lübeck, 23562 Lübeck, Germany; (K.L.); (J.Z.)
- University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| |
Collapse
|
2
|
Podtelezhnikov AA, Monroe JJ, Aslamkhan AG, Pearson K, Qin C, Tamburino AM, Loboda AP, Glaab WE, Sistare FD, Tanis KQ. Quantitative Transcriptional Biomarkers of Xenobiotic Receptor Activation in Rat Liver for the Early Assessment of Drug Safety Liabilities. Toxicol Sci 2021; 175:98-112. [PMID: 32119089 DOI: 10.1093/toxsci/kfaa026] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The robust transcriptional plasticity of liver mediated through xenobiotic receptors underlies its ability to respond rapidly and effectively to diverse chemical stressors. Thus, drug-induced gene expression changes in liver serve not only as biomarkers of liver injury, but also as mechanistic sentinels of adaptation in metabolism, detoxification, and tissue protection from chemicals. Modern RNA sequencing methods offer an unmatched opportunity to quantitatively monitor these processes in parallel and to contextualize the spectrum of dose-dependent stress, adaptation, protection, and injury responses induced in liver by drug treatments. Using this approach, we profiled the transcriptional changes in rat liver following daily oral administration of 120 different compounds, many of which are known to be associated with clinical risk for drug-induced liver injury by diverse mechanisms. Clustering, correlation, and linear modeling analyses were used to identify and optimize coexpressed gene signatures modulated by drug treatment. Here, we specifically focused on prioritizing 9 key signatures for their pragmatic utility for routine monitoring in initial rat tolerability studies just prior to entering drug development. These signatures are associated with 5 canonical xenobiotic nuclear receptors (AHR, CAR, PXR, PPARα, ER), 3 mediators of reactive metabolite-mediated stress responses (NRF2, NRF1, P53), and 1 liver response following activation of the innate immune response. Comparing paradigm chemical inducers of each receptor to the other compounds surveyed enabled us to identify sets of optimized gene expression panels and associated scoring algorithms proposed as quantitative mechanistic biomarkers with high sensitivity, specificity, and quantitative accuracy. These findings were further qualified using public datasets, Open TG-GATEs and DrugMatrix, and internal development compounds. With broader collaboration and additional qualification, the quantitative toxicogenomic framework described here could inform candidate selection prior to committing to drug development, as well as complement and provide a deeper understanding of the conventional toxicology study endpoints used later in drug development.
Collapse
Affiliation(s)
| | - James J Monroe
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Amy G Aslamkhan
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Kara Pearson
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Chunhua Qin
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | | | | | - Warren E Glaab
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Frank D Sistare
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | | |
Collapse
|
3
|
Aguado-Fraile E, Tassinari A, Ishii Y, Sigel C, Lowery MA, Goyal L, Gliser C, Jiang L, Pandya SS, Wu B, Bardeesy N, Choe S, Deshpande V. Molecular and morphological changes induced by ivosidenib correlate with efficacy in mutant- IDH1 cholangiocarcinoma. Future Oncol 2021; 17:2057-2074. [PMID: 33709779 DOI: 10.2217/fon-2020-1274] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: IDH1 mutations occur in approximately 13% of intrahepatic cholangiocarcinomas (IHCCs). The oral, targeted, mutant IDH1 (mIDH1) inhibitor ivosidenib (AG-120) suppresses production of the oncometabolite D-2-hydroxyglutarate, promoting disease stabilization and improved progression-free survival (PFS) in mIDH1 IHCC. Materials & methods: Harnessing matched baseline and on-treatment biopsies, we investigate the potential mechanisms underlying ivosidenib's efficacy. Results: mIDH1 inhibition leads to decreased cytoplasm and expression of hepatocyte lineage markers in patients with prolonged PFS. These findings are accompanied by downregulation of biliary fate, cell cycle progression and AKT pathway activity. Conclusion: Ivosidenib stimulates a hepatocyte differentiation program in mIDH1 IHCC, a phenotype associated with clinical benefit. mIDH1 inhibition could be a paradigm for differentiation-based therapy in solid tumors. Clinical trial registration: NCT02073994 (ClinicalTrials.gov).
Collapse
Affiliation(s)
| | | | - Yuko Ishii
- Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Carlie Sigel
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maeve A Lowery
- Trinity St James Cancer Institute, Trinity College Dublin, Dublin D02, Ireland
| | - Lipika Goyal
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | | | - Liewen Jiang
- Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | | | - Bin Wu
- Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Nabeel Bardeesy
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Sung Choe
- Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Vikram Deshpande
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA.,Department of Pathology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
4
|
Bonato M, Corrà F, Bellio M, Guidolin L, Tallandini L, Irato P, Santovito G. PFAS Environmental Pollution and Antioxidant Responses: An Overview of the Impact on Human Field. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:8020. [PMID: 33143342 PMCID: PMC7663035 DOI: 10.3390/ijerph17218020] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 01/09/2023]
Abstract
Due to their unique properties, perfluorinated substances (PFAS) are widely used in multiple industrial and commercial applications, but they are toxic for animals, humans included. This review presents some available data on the PFAS environmental distribution in the world, and in particular in Europe and in the Veneto region of Italy, where it has become a serious problem for human health. The consumption of contaminated food and drinking water is considered one of the major source of exposure for humans. Worldwide epidemiological studies report the negative effects that PFAS have on human health, due to environmental pollution, including infertility, steroid hormone perturbation, thyroid, liver and kidney disorders, and metabolic disfunctions. In vitro and in vivo researches correlated PFAS exposure to oxidative stress effects (in mammals as well as in other vertebrates of human interest), produced by a PFAS-induced increase of reactive oxygen species formation. The cellular antioxidant defense system is activated by PFAS, but it is only partially able to avoid the oxidative damage to biomolecules.
Collapse
Affiliation(s)
| | | | | | | | | | - Paola Irato
- Department of Biology, University of Padova, 35131 Padova, Italy; (M.B.); (F.C.); (M.B.); (L.G.); (L.T.)
| | - Gianfranco Santovito
- Department of Biology, University of Padova, 35131 Padova, Italy; (M.B.); (F.C.); (M.B.); (L.G.); (L.T.)
| |
Collapse
|
5
|
Nault R, Fader KA, Bhattacharya S, Zacharewski TR. Single-Nuclei RNA Sequencing Assessment of the Hepatic Effects of 2,3,7,8-Tetrachlorodibenzo-p-dioxin. Cell Mol Gastroenterol Hepatol 2020; 11:147-159. [PMID: 32791302 PMCID: PMC7674514 DOI: 10.1016/j.jcmgh.2020.07.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Characterization of cell specific transcriptional responses to hepatotoxicants is lost in the averages of bulk RNA-sequencing (RNA-seq). Single-cell/nuclei RNA-seq technologies enable the transcriptomes of individual cell (sub)types to be assessed within the context of in vivo models. METHODS Single-nuclei RNA-sequencing (snSeq) of frozen liver samples from male C57BL/6 mice gavaged with sesame oil vehicle or 30 μg/kg 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) every 4 days for 28 days was used to demonstrate the application of snSeq for the evaluation of xenobiotics. RESULTS A total of 19,907 genes were detected across 16,015 nuclei from control and TCDD-treated livers. Eleven cell (sub)types reflected the expected cell diversity of the liver including distinct pericentral, midzonal, and periportal hepatocyte subpopulations. TCDD altered relative proportions of cell types and elicited cell-specific gene expression profiles. For example, macrophages increased from 0.5% to 24.7%, while neutrophils were only present in treated samples, consistent with histological evaluation. The number of differentially expressed genes (DEGs) in each cell type ranged from 122 (cholangiocytes) to 7625 (midcentral hepatocytes), and loosely correlated with the basal expression level of Ahr, the canonical mediator of TCDD and related compounds. In addition to the expected functions within each cell (sub)types, RAS signaling and related pathways were specifically enriched in nonparenchymal cells while metabolic process enrichment occurred primarily in hepatocytes. snSeq also identified the expansion of a Kupffer cell subtype highly expressing Gpnmb, as reported in a dietary NASH model. CONCLUSIONS We show that snSeq of frozen liver samples can be used to assess cell-specific transcriptional changes and population shifts in models of hepatotoxicity when examining freshly isolated cells is not feasible.
Collapse
Affiliation(s)
- Rance Nault
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Kelly A Fader
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Sudin Bhattacharya
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Tim R Zacharewski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
6
|
Hatchwell L, Harney DJ, Cielesh M, Young K, Koay YC, O’Sullivan JF, Larance M. Multi-omics Analysis of the Intermittent Fasting Response in Mice Identifies an Unexpected Role for HNF4α. Cell Rep 2020; 30:3566-3582.e4. [DOI: 10.1016/j.celrep.2020.02.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/24/2020] [Accepted: 02/11/2020] [Indexed: 12/21/2022] Open
|
7
|
Goemann IM, Marczyk VR, Romitti M, Wajner SM, Maia AL. Current concepts and challenges to unravel the role of iodothyronine deiodinases in human neoplasias. Endocr Relat Cancer 2018; 25:R625-R645. [PMID: 30400023 DOI: 10.1530/erc-18-0097] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Thyroid hormones (THs) are essential for the regulation of several metabolic processes and the energy consumption of the organism. Their action is exerted primarily through interaction with nuclear receptors controlling the transcription of thyroid hormone-responsive genes. Proper regulation of TH levels in different tissues is extremely important for the equilibrium between normal cellular proliferation and differentiation. The iodothyronine deiodinases types 1, 2 and 3 are key enzymes that perform activation and inactivation of THs, thus controlling TH homeostasis in a cell-specific manner. As THs seem to exert their effects in all hallmarks of the neoplastic process, dysregulation of deiodinases in the tumoral context can be critical to the neoplastic development. Here, we aim at reviewing the deiodinases expression in different neoplasias and exploit the mechanisms by which they play an essential role in human carcinogenesis. TH modulation by deiodinases and other classical pathways may represent important targets with the potential to oppose the neoplastic process.
Collapse
Affiliation(s)
- Iuri Martin Goemann
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vicente Rodrigues Marczyk
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Mirian Romitti
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels, Belgium
| | - Simone Magagnin Wajner
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Luiza Maia
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
8
|
Singh BK, Sinha RA, Yen PM. Novel Transcriptional Mechanisms for Regulating Metabolism by Thyroid Hormone. Int J Mol Sci 2018; 19:3284. [PMID: 30360449 PMCID: PMC6214012 DOI: 10.3390/ijms19103284] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/11/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
The thyroid hormone plays a key role in energy and nutrient metabolisms in many tissues and regulates the transcription of key genes in metabolic pathways. It has long been believed that thyroid hormones (THs) exerted their effects primarily by binding to nuclear TH receptors (THRs) that are associated with conserved thyroid hormone response elements (TREs) located on the promoters of target genes. However, recent transcriptome and ChIP-Seq studies have challenged this conventional view as discordance was observed between TH-responsive genes and THR binding to DNA. While THR association with other transcription factors bound to DNA, TH activation of THRs to mediate effects that do not involve DNA-binding, or TH binding to proteins other than THRs have been invoked as potential mechanisms to explain this discrepancy, it appears that additional novel mechanisms may enable TH to regulate the mRNA expression. These include activation of transcription factors by SIRT1 via metabolic actions by TH, the post-translational modification of THR, the THR co-regulation of transcription with other nuclear receptors and transcription factors, and the microRNA (miR) control of RNA transcript expression to encode proteins involved in the cellular metabolism. Together, these novel mechanisms enlarge and diversify the panoply of metabolic genes that can be regulated by TH.
Collapse
Affiliation(s)
- Brijesh Kumar Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore.
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow 226014, Uttar Pradesh, India.
| | - Paul Michael Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore.
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
9
|
Pei Y, Yao Q, Yuan S, Xie B, Liu Y, Ye C, Zhuo H. GATA4 promotes hepatoblastoma cell proliferation by altering expression of miR125b and DKK3. Oncotarget 2018; 7:77890-77901. [PMID: 27788486 PMCID: PMC5363629 DOI: 10.18632/oncotarget.12839] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 10/14/2016] [Indexed: 12/02/2022] Open
Abstract
GATA4 is a zinc finger DNA-binding protein that plays an important role in mammalian liver development. However, the effects of GATA4 in hepatoblastoma (HB), a common liver cancer in pediatric patients, remain largely unknown. In this study, we demonstrate that GATA4 promotes growth and survival in the Huh6 human hepatoblastoma cell line. GATA4 expression was high in Huh6 cells, and its knockdown decreased expression of Dickkopf-related protein 3 (DKK3), a gene that may contribute to premature or undifferentiated phenotypes in HB. GATA4 also directly bound to the promoter regions of the miRNA miR125b and inhibited its expression in Huh6 cells. DKK3 was a direct target of miR125b in Huh6 cells. Inhibition of miR125b or overexpression of DKK3 promoted proliferation, survival, migration, and invasion in Huh6 cells. This is the first report to demonstrate that GATA4 promotes oncogenesis by inhibiting miR125b-dependent suppression of DKK3 expression. This GATA4/miR125b/DKK3 axis may be a major regulator of growth, migration, invasion, and survival in hepatoma cells, and is therefore a potential therapeutic target or biomarker for progression in HB patients.
Collapse
Affiliation(s)
- Yihua Pei
- Central Laboratory, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - Qin Yao
- Central Laboratory, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - Sibo Yuan
- Department of Gastrointestinal Surgery, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - Bozhen Xie
- Department of Spine Surgery, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - Yan Liu
- Department of Pathology, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - Chunsheng Ye
- Department Otolaryngology, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - Huiqin Zhuo
- Department of Gastrointestinal Surgery, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| |
Collapse
|
10
|
Sun Q, Zong G, Valvi D, Nielsen F, Coull B, Grandjean P. Plasma Concentrations of Perfluoroalkyl Substances and Risk of Type 2 Diabetes: A Prospective Investigation among U.S. Women. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:037001. [PMID: 29498927 PMCID: PMC6071816 DOI: 10.1289/ehp2619] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 05/18/2023]
Abstract
BACKGROUND Emerging evidence suggests that perfluoroalkyl substances (PFASs) are endocrine disruptors and may contribute to the etiology of type 2 diabetes (T2D), but this hypothesis needs to be clarified in prospective human studies. OBJECTIVES Our objective was to examine the associations between PFAS exposures and subsequent incidence of T2D in the Nurses' Health Study II (NHSII). In addition, we aimed to evaluate potential demographic and lifestyle determinants of plasma PFAS concentrations. METHODS A prospective nested case-control study of T2D was conducted among participants who were free of diabetes, cardiovascular disease, and cancer in 1995-2000 [(mean±SD): 45.3±4.4 y) of age]. We identified and ascertained 793 incident T2D cases through 2011 (mean±SD) years of follow-up: 6.7±3.7 y). Each case was individually matched to a control (on age, month and fasting status at sample collection, and menopausal status and hormone replacement therapy). Plasma concentrations of five major PFASs, including perfluorooctanesulfonic acid (PFOS), perfluorooctanoic acid (PFOA), perfluorohexanesulfonate, perfluorononanoic acid, and perfluorodecanoic acid were measured. Odds ratios (ORs) of T2D by PFAS tertiles were estimated by conditional logistic regression. RESULTS Shorter breastfeeding duration and higher intake of certain foods, such as seafood and popcorn, were significantly associated with higher plasma concentrations of PFASs among controls. After multivariate adjustment for T2D risk factors, including body mass index, family history, physical activity, and other covariates, higher plasma concentrations of PFOS and PFOA were associated with an elevated risk of T2D. Comparing extreme tertiles of PFOS or PFOA, ORs were 1.62 (95% CI: 1.09, 2.41; ptrend=0.02) and 1.54 (95% CI: 1.04, 2.28; ptrend=0.03), respectively. Other PFASs were not clearly associated with T2D risk. CONCLUSIONS Background exposures to PFASs in the late 1990s were associated with higher T2D risk during the following years in a prospective case-control study of women from the NHSII. These findings support a potential diabetogenic effect of PFAS exposures. https://doi.org/10.1289/EHP2619.
Collapse
Affiliation(s)
- Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health , Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts, USA
| | - Geng Zong
- Department of Nutrition, Harvard T.H. Chan School of Public Health , Boston, Massachusetts, USA
| | - Damaskini Valvi
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Flemming Nielsen
- Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Brent Coull
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health , Boston, Massachusetts, USA
| | - Philippe Grandjean
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Institute of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
11
|
Singh BK, Sinha RA, Ohba K, Yen PM. Role of thyroid hormone in hepatic gene regulation, chromatin remodeling, and autophagy. Mol Cell Endocrinol 2017; 458:160-168. [PMID: 28216439 DOI: 10.1016/j.mce.2017.02.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 01/21/2023]
Abstract
Thyroid hormone (TH) actions on development and metabolism have been studied ever since the discovery of thyroxine almost a century ago. Initial studies focused on the physiological and biochemical actions of TH. Later, the cloning of the thyroid hormone receptor (THR) isoforms and the development of techniques enabled the study of TH regulation of complex cellular processes (such as gene transcription). Recently we found that TH activates secondary transcription factors such as FOXO1, to amplify gene transcription; and also is a potent inducer of autophagy that was critical for fatty acid β-oxidation in the liver. This review summarizes the recent advancements in our understanding of TH regulation of gene expression of metabolic genes (via co-regulators/transcription factors and epigenetic control) and autophagy in the liver. Our deeper understanding of TH action recently has led to the development of tissue- and THR isoform-specific TH mimetics that may be useful for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Brijesh Kumar Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 169857, Singapore
| | - Rohit Anthony Sinha
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 169857, Singapore
| | - Kenji Ohba
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 169857, Singapore; Department of Internal Medicine, Enshu Hospital, Hamamatsu, Shizuoka 430-0929, Japan
| | - Paul Michael Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 169857, Singapore.
| |
Collapse
|
12
|
Piccolo P, Annunziata P, Soria LR, Attanasio S, Barbato A, Castello R, Carissimo A, Quagliata L, Terracciano LM, Brunetti-Pierri N. Down-regulation of hepatocyte nuclear factor-4α and defective zonation in livers expressing mutant Z α1-antitrypsin. Hepatology 2017; 66:124-135. [PMID: 28295475 DOI: 10.1002/hep.29160] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/09/2017] [Accepted: 03/10/2017] [Indexed: 12/19/2022]
Abstract
UNLABELLED α1 -Antitrypsin (AAT) deficiency is one of the most common genetic disorders and the liver disease due to the Z mutant of AAT (ATZ) is a prototype of conformational disorder due to protein misfolding with consequent aberrant intermolecular protein aggregation. In the present study, we found that livers of PiZ transgenic mice expressing human ATZ have altered expression of a network of hepatocyte transcriptional factors, including hepatocyte nuclear factor-4α, that is early down-regulated and induces a transcriptional repression of ATZ expression. Reduced hepatocyte nuclear factor-4α was associated with activation of β-catenin, which regulates liver zonation. Livers of PiZ mice and human patients with AAT deficiency were both found to have a severe perturbation of liver zonation. Functionally, PiZ mice showed a severe defect of ureagenesis, as shown by increased baseline ammonia, and reduced urea production and survival after an ammonia challenge. Down-regulation of hepatocyte nuclear factor-4α expression and defective zonation in livers have not been recognized so far as features of the liver disease caused by ATZ and are likely involved in metabolic disturbances and in the increased risk of hepatocellular carcinoma in patients with AAT deficiency. CONCLUSION The findings of this study are consistent with the concept that abnormal AAT protein conformation and intrahepatic accumulation have broad effects on metabolic liver functions. (Hepatology 2017;66:124-135).
Collapse
Affiliation(s)
- Pasquale Piccolo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | | | - Leandro R Soria
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Sergio Attanasio
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Anna Barbato
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Raffaele Castello
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | | | - Luca Quagliata
- Molecular Pathology Division, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Luigi M Terracciano
- Molecular Pathology Division, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy.,Department of Translational Medicine, Federico II University, Naples, Italy
| |
Collapse
|
13
|
Wijetunga NA, Pascual M, Tozour J, Delahaye F, Alani M, Adeyeye M, Wolkoff AW, Verma A, Greally JM. A pre-neoplastic epigenetic field defect in HCV-infected liver at transcription factor binding sites and polycomb targets. Oncogene 2017; 36:2030-2044. [PMID: 27721404 PMCID: PMC5383522 DOI: 10.1038/onc.2016.340] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 07/26/2016] [Accepted: 08/05/2016] [Indexed: 12/11/2022]
Abstract
The predisposition of patients with Hepatitis C virus (HCV) infection to hepatocellular carcinoma (HCC) involves components of viral infection, inflammation and time. The development of multifocal, genetically distinct tumours is suggestive of a field defect affecting the entire liver. The molecular susceptibility mediating such a field defect is not understood. One potential mediator of long-term cellular reprogramming is heritable (epigenetic) regulation of transcription, exemplified by DNA methylation. We studied epigenetic and transcriptional changes in HCV-infected livers in comparison with control, uninfected livers and HCC, allowing us to identify pre-neoplastic epigenetic and transcriptional events. We find the HCV-infected liver to have a pattern of acquisition of DNA methylation targeted to candidate enhancers active in liver cells, enriched for the binding sites of the FOXA1, FOXA2 and HNF4A transcription factors. These enhancers can be subdivided into those proximal to genes implicated in liver cancer or to genes involved in stem cell development, the latter distinguished by increased CG dinucleotide density and polycomb-mediated repression, manifested by the additional acquisition of histone H3 lysine 27 trimethylation (H3K27me3). Transcriptional studies on our samples showed that the increased DNA methylation at enhancers was associated with decreased local gene expression, results validated in independent samples from The Cancer Genome Atlas. Pharmacological depletion of H3K27me3 using the EZH2 inhibitor GSK343 in HepG2 cells suppressed cell growth and also revealed that local acquired DNA methylation was not dependent upon the presence of polycomb-mediated repression. The results support a model of HCV infection influencing the binding of transcription factors to cognate sites in the genome, with consequent local acquisition of DNA methylation, and the added repressive influence of polycomb at a subset of CG-dense cis-regulatory sequences. These epigenetic events occur before neoplastic transformation, resulting in what may be a pharmacologically reversible epigenetic field defect in HCV-infected liver.
Collapse
Affiliation(s)
- N A Wijetunga
- Department of Genetics and Center for Epigenomics, Bronx, NY, USA
| | - M Pascual
- Department of Genetics and Center for Epigenomics, Bronx, NY, USA
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, Oncohematology Department, Pamplona, Spain
| | - J Tozour
- Department of Genetics and Center for Epigenomics, Bronx, NY, USA
| | - F Delahaye
- Department of Obstetrics, Gynecology and Women's Health, Bronx, NY, USA
| | - M Alani
- Department of Medicine (Division of Gastroenterology and Liver Diseases), Bronx, NY, USA
- Marion Bessin Liver Research Center, Bronx, NY, USA
| | - M Adeyeye
- Department of Genetics and Center for Epigenomics, Bronx, NY, USA
| | - A W Wolkoff
- Department of Medicine (Division of Gastroenterology and Liver Diseases), Bronx, NY, USA
- Marion Bessin Liver Research Center, Bronx, NY, USA
| | - A Verma
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - J M Greally
- Department of Genetics and Center for Epigenomics, Bronx, NY, USA
- Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx NY 10461, USA. E-mail:
| |
Collapse
|
14
|
Crosstalk of HNF4 α with extracellular and intracellular signaling pathways in the regulation of hepatic metabolism of drugs and lipids. Acta Pharm Sin B 2016; 6:393-408. [PMID: 27709008 PMCID: PMC5045537 DOI: 10.1016/j.apsb.2016.07.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/05/2016] [Accepted: 05/11/2016] [Indexed: 12/15/2022] Open
Abstract
The liver is essential for survival due to its critical role in the regulation of metabolic homeostasis. Metabolism of xenobiotics, such as environmental chemicals and drugs by the liver protects us from toxic effects of these xenobiotics, whereas metabolism of cholesterol, bile acids (BAs), lipids, and glucose provide key building blocks and nutrients to promote the growth or maintain the survival of the organism. As a well-established master regulator of liver development and function, hepatocyte nuclear factor 4 alpha (HNF4α) plays a critical role in regulating a large number of key genes essential for the metabolism of xenobiotics, metabolic wastes, and nutrients. The expression and activity of HNF4α is regulated by diverse hormonal and signaling pathways such as growth hormone, glucocorticoids, thyroid hormone, insulin, transforming growth factor-β, estrogen, and cytokines. HNF4α appears to play a central role in orchestrating the transduction of extracellular hormonal signaling and intracellular stress/nutritional signaling onto transcriptional changes in the liver. There have been a few reviews on the regulation of drug metabolism, lipid metabolism, cell proliferation, and inflammation by HNF4α. However, the knowledge on how the expression and transcriptional activity of HNF4α is modulated remains scattered. Herein I provide comprehensive review on the regulation of expression and transcriptional activity of HNF4α, and how HNF4α crosstalks with diverse extracellular and intracellular signaling pathways to regulate genes essential in liver pathophysiology.
Collapse
|
15
|
Amano I, Takatsuru Y, Toya S, Haijima A, Iwasaki T, Grasberger H, Refetoff S, Koibuchi N. Aberrant Cerebellar Development in Mice Lacking Dual Oxidase Maturation Factors. Thyroid 2016; 26:741-52. [PMID: 26914863 PMCID: PMC4860669 DOI: 10.1089/thy.2015.0034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Thyroid hormone (TH) plays a key role in the developing brain, including the cerebellum. TH deficiency induces organizational changes of the cerebellum, causing cerebellar ataxia. However, the mechanisms causing these abnormalities are poorly understood. Various animal models have been used to study the mechanism. Lacking dual oxidase (DUOX) and its maturation factor (DUOXA) are major inducers of congenital hypothyroidism. Thus, this study examined the organizational changes of the cerebellum using knockout mice of the Duoxa gene (Duoxa-/-). METHODS The morphological, behavioral, and electrophysiological changes were analyzed in wild type (Wt) and Duoxa-deficient (Duoxa-/-) mice from postnatal day (P) 10 to P30. To detect the changes in the expression levels of presynaptic proteins, Western blot analysis was performed. RESULTS The proliferation and migration of granule cells was delayed after P15 in Duoxa-/- mice. However, these changes disappeared by P25. Although the cerebellar structure of Duoxa-/- mice was not significantly different from that of Wt mice at P25, motor coordination was impaired. It was also found that the amplitude of paired-pulse facilitation at parallel fiber-Purkinje cell synapses decreased in Duoxa-/- mice, particularly at P15. There were no differences between expression levels of presynaptic proteins regulating neurotransmitter release at P25. CONCLUSIONS These results indicate that the anatomical catch-up growth of the cerebellum did not normalize its function because of the disturbance of neuronal circuits by the combined effect of hypothyroidism and functional disruption of the DUOX/DUOXA complex.
Collapse
Affiliation(s)
- Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yusuke Takatsuru
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Syutaro Toya
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Asahi Haijima
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Toshiharu Iwasaki
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Helmut Grasberger
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
- Department of Genetics, The University of Chicago, Chicago, Illinois
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
16
|
Ohba K, Leow MKS, Singh BK, Sinha RA, Lesmana R, Liao XH, Ghosh S, Refetoff S, Sng JCG, Yen PM. Desensitization and Incomplete Recovery of Hepatic Target Genes After Chronic Thyroid Hormone Treatment and Withdrawal in Male Adult Mice. Endocrinology 2016; 157:1660-1672. [PMID: 26866609 PMCID: PMC4816733 DOI: 10.1210/en.2015-1848] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/03/2016] [Indexed: 02/08/2023]
Abstract
Clinical symptoms may vary and not necessarily reflect serum thyroid hormone (TH) levels during acute and chronic hyperthyroidism as well as recovery from hyperthyroidism. We thus examined changes in hepatic gene expression and serum TH/TSH levels in adult male mice treated either with a single T3 (20 μg per 100 g body weight) injection (acute T3) or daily injections for 14 days (chronic T3) followed by 10 days of withdrawal. Gene expression arrays from livers harvested at these time points showed that among positively-regulated target genes, 320 were stimulated acutely and 429 chronically by T3. Surprisingly, only 69 of 680 genes (10.1%) were induced during both periods, suggesting desensitization of the majority of acutely stimulated target genes. About 90% of positively regulated target genes returned to baseline expression levels after 10 days of withdrawal; however, 67 of 680 (9.9%) did not return to baseline despite normalization of serum TH/TSH levels. Similar findings also were observed for negatively regulated target genes. Chromatin immunoprecipitation analysis of representative positively regulated target genes suggested that acetylation of H3K9/K14 was associated with acute stimulation, whereas trimethylation of H3K4 was associated with chronic stimulation. In an in vivo model of chronic intrahepatic hyperthyroidism since birth, adult male monocarboxylate transporter-8 knockout mice also demonstrated desensitization of most acutely stimulated target genes that were examined. In summary, we have identified transcriptional desensitization and incomplete recovery of gene expression during chronic hyperthyroidism and recovery. Our findings may be a potential reason for discordance between clinical symptoms and serum TH levels observed in these conditions.
Collapse
Affiliation(s)
- Kenji Ohba
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Melvin Khee-Shing Leow
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Rohit Anthony Sinha
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Ronny Lesmana
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Xiao-Hui Liao
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Sujoy Ghosh
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Samuel Refetoff
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Judy Chia Ghee Sng
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| | - Paul Michael Yen
- Cardiovascular and Metabolic Disorders Program (K.O., B.K.S., R.A.S., R.L., S.G., P.M.Y.), Duke-NUS Medical School, Singapore, Singapore 169857; Department of Endocrinology (M.K.-S.L.), Tan Tock Seng Hospital, Singapore, Singapore 229899; Singapore Institute for Clinical Sciences (M.K.-S.L.), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore 117609; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung, West Java 45363, Indonesia; Departments of Medicine (X.-H.L., S.R.) and Pediatrics and Committee on Genetics (S.R.), The University of Chicago, Chicago, Illinois 60637; and Department of Pharmacology (J.C.G.S.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| |
Collapse
|
17
|
Ohguchi H, Hideshima T, Bhasin MK, Gorgun GT, Santo L, Cea M, Samur MK, Mimura N, Suzuki R, Tai YT, Carrasco RD, Raje N, Richardson PG, Munshi NC, Harigae H, Sanda T, Sakai J, Anderson KC. The KDM3A-KLF2-IRF4 axis maintains myeloma cell survival. Nat Commun 2016; 7:10258. [PMID: 26728187 PMCID: PMC4728406 DOI: 10.1038/ncomms10258] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 11/23/2015] [Indexed: 12/14/2022] Open
Abstract
KDM3A is implicated in tumorigenesis; however, its biological role in multiple myeloma (MM) has not been elucidated. Here we identify KDM3A–KLF2–IRF4 axis dependence in MM. Knockdown of KDM3A is toxic to MM cells in vitro and in vivo. KDM3A maintains expression of KLF2 and IRF4 through H3K9 demethylation, and knockdown of KLF2 triggers apoptosis. Moreover, KLF2 directly activates IRF4 and IRF4 reciprocally upregulates KLF2, forming a positive autoregulatory circuit. The interaction of MM cells with bone marrow milieu mediates survival of MM cells. Importantly, silencing of KDM3A, KLF2 or IRF4 both decreases MM cell adhesion to bone marrow stromal cells and reduces MM cell homing to the bone marrow, in association with decreased ITGB7 expression in MAF-translocated MM cell lines. Our results indicate that the KDM3A–KLF2–IRF4 pathway plays an essential role in MM cell survival and homing to the bone marrow, and therefore represents a therapeutic target. Several histone modifiers have been implicated in the survival of multiple myeloma cells. Here, the authors reveal a role for the histone demethylase KDM3A in the survival of this haematologic cancer, and show that mechanistically KDM3A removes H3K9 methylation from the promoters of KLF2 and IRF4, genes essential for myeloma cell survival.
Collapse
Affiliation(s)
- Hiroto Ohguchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Teru Hideshima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Manoj K Bhasin
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115, USA
| | - Gullu T Gorgun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Loredana Santo
- MGH Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Michele Cea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Mehmet K Samur
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Naoya Mimura
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Rikio Suzuki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Yu-Tzu Tai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Ruben D Carrasco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Noopur Raje
- MGH Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Nikhil C Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,West Roxbury Division, VA Boston Healthcare System, West Roxbury, MA 02132, USA
| | - Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8574, Japan
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, Department of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Juro Sakai
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan
| | - Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| |
Collapse
|
18
|
Cvoro A, Devito L, Milton FA, Noli L, Zhang A, Filippi C, Sakai K, Suh JH, Sieglaff DH, Dhawan A, Sakai T, Ilic D, Webb P. A thyroid hormone receptor/KLF9 axis in human hepatocytes and pluripotent stem cells. Stem Cells 2015; 33:416-428. [PMID: 25330987 PMCID: PMC6317531 DOI: 10.1002/stem.1875] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 08/21/2014] [Accepted: 09/13/2014] [Indexed: 12/15/2022]
Abstract
Biological processes require close cooperation of multiple transcription factors that integrate different signals. Thyroid hormone receptors (TRs) induce Krüppel-like factor 9 (KLF9) to regulate neurogenesis. Here, we show that triiodothyronine (T3) also works through TR to induce KLF9 in HepG2 liver cells, mouse liver, and mouse and human primary hepatocytes and sought to understand TR/KLF9 network function in the hepatocyte lineage and stem cells. Knockdown experiments reveal that KLF9 regulates hundreds of HepG2 target genes and modulates T3 response. Together, T3 and KLF9 target genes influence pathways implicated in stem cell self-renewal and differentiation, including Notch signaling, and we verify that T3 and KLF9 cooperate to regulate key Notch pathway genes and work independently to regulate others. T3 also induces KLF9 in human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSC) and this effect persists during differentiation to definitive endoderm and hiPSC-derived hepatocytes. Microarray analysis reveals that T3 regulates hundreds of hESC and hiPSC target genes that cluster into many of the same pathways implicated in TR and KLF9 regulation in HepG2 cells. KLF9 knockdown confirms that TR and KLF9 cooperate to regulate Notch pathway genes in hESC and hiPSC, albeit in a partly cell-specific manner. Broader analysis of T3 responsive hESC/hiPSC genes suggests that TRs regulate multiple early steps in ESC differentiation. We propose that TRs cooperate with KLF9 to regulate hepatocyte proliferation and differentiation and early stages of organogenesis and that TRs exert widespread and important influences on ESC biology.
Collapse
Affiliation(s)
- Aleksandra Cvoro
- Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Liani Devito
- Stem Cell Laboratory, Assisted Conception Unit, Division of Women’s Health, King’s College London, London, United Kingdom
| | - Flora A. Milton
- Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Laila Noli
- Stem Cell Laboratory, Assisted Conception Unit, Division of Women’s Health, King’s College London, London, United Kingdom
| | - Aijun Zhang
- Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Celine Filippi
- Guy’s and St. Thomas NHS Foundation Trust and King’s College Biomedical Research Centre, London, United Kingdom
- Institute of Liver Studies, King’s College London, London, United Kingdom
| | - Keiko Sakai
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, The University of Liverpool, Liverpool, United Kingdom
| | - Ji Ho Suh
- Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Anil Dhawan
- Institute of Liver Studies, King’s College London, London, United Kingdom
| | - Takao Sakai
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, The University of Liverpool, Liverpool, United Kingdom
| | - Dusko Ilic
- Stem Cell Laboratory, Assisted Conception Unit, Division of Women’s Health, King’s College London, London, United Kingdom
| | - Paul Webb
- Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
19
|
Inagaki T, Iwasaki S, Matsumura Y, Kawamura T, Tanaka T, Abe Y, Yamasaki A, Tsurutani Y, Yoshida A, Chikaoka Y, Nakamura K, Magoori K, Nakaki R, Osborne TF, Fukami K, Aburatani H, Kodama T, Sakai J. The FBXL10/KDM2B scaffolding protein associates with novel polycomb repressive complex-1 to regulate adipogenesis. J Biol Chem 2014; 290:4163-77. [PMID: 25533466 DOI: 10.1074/jbc.m114.626929] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Polycomb repressive complex 1 (PRC1) plays an essential role in the epigenetic repression of gene expression during development and cellular differentiation via multiple effector mechanisms, including ubiquitination of H2A and chromatin compaction. However, whether it regulates the stepwise progression of adipogenesis is unknown. Here, we show that FBXL10/KDM2B is an anti-adipogenic factor that is up-regulated during the early phase of 3T3-L1 preadipocyte differentiation and in adipose tissue in a diet-induced model of obesity. Interestingly, inhibition of adipogenesis does not require the JmjC demethylase domain of FBXL10, but it does require the F-box and leucine-rich repeat domains, which we show recruit a noncanonical polycomb repressive complex 1 (PRC1) containing RING1B, SKP1, PCGF1, and BCOR. Knockdown of either RING1B or SKP1 prevented FBXL10-mediated repression of 3T3-L1 preadipocyte differentiation indicating that PRC1 formation mediates the inhibitory effect of FBXL10 on adipogenesis. Using ChIP-seq, we show that FBXL10 recruits RING1B to key specific genomic loci surrounding the key cell cycle and the adipogenic genes Cdk1, Uhrf1, Pparg1, and Pparg2 to repress adipogenesis. These results suggest that FBXL10 represses adipogenesis by targeting a noncanonical PRC1 complex to repress key genes (e.g. Pparg) that control conversion of pluripotent cells into the adipogenic lineage.
Collapse
Affiliation(s)
- Takeshi Inagaki
- From the Division of Metabolic Medicine, the Translational Systems Biology and Medicine Initiative Center for Disease Biology and Integrative Medicine, University of Tokyo, Tokyo 153-8904, Japan,
| | | | | | - Takeshi Kawamura
- the Translational Systems Biology and Medicine Initiative Center for Disease Biology and Integrative Medicine, University of Tokyo, Tokyo 153-8904, Japan, Laboratory for Systems Biology and Medicine
| | - Toshiya Tanaka
- From the Division of Metabolic Medicine, the Translational Systems Biology and Medicine Initiative Center for Disease Biology and Integrative Medicine, University of Tokyo, Tokyo 153-8904, Japan, Laboratory for Systems Biology and Medicine
| | - Yohei Abe
- From the Division of Metabolic Medicine
| | | | | | - Ayano Yoshida
- From the Division of Metabolic Medicine, the Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Science, Tokyo 192-0392, Japan
| | - Yoko Chikaoka
- the Translational Systems Biology and Medicine Initiative Center for Disease Biology and Integrative Medicine, University of Tokyo, Tokyo 153-8904, Japan, Laboratory for Systems Biology and Medicine
| | - Kanako Nakamura
- From the Division of Metabolic Medicine, the Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Science, Tokyo 192-0392, Japan
| | - Kenta Magoori
- From the Division of Metabolic Medicine, the Translational Systems Biology and Medicine Initiative Center for Disease Biology and Integrative Medicine, University of Tokyo, Tokyo 153-8904, Japan
| | - Ryo Nakaki
- Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan
| | - Timothy F Osborne
- the Metabolic Disease Program, Sanford-Burnham Medical Research Institute, Orlando, Florida 32827, and
| | - Kiyoko Fukami
- the Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Science, Tokyo 192-0392, Japan
| | - Hiroyuki Aburatani
- the Translational Systems Biology and Medicine Initiative Center for Disease Biology and Integrative Medicine, University of Tokyo, Tokyo 153-8904, Japan, Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan
| | | | - Juro Sakai
- From the Division of Metabolic Medicine, the Translational Systems Biology and Medicine Initiative Center for Disease Biology and Integrative Medicine, University of Tokyo, Tokyo 153-8904, Japan,
| |
Collapse
|
20
|
Koh KH, Pan X, Zhang W, McLachlan A, Urrutia R, Jeong H. Krüppel-like factor 9 promotes hepatic cytochrome P450 2D6 expression during pregnancy in CYP2D6-humanized mice. Mol Pharmacol 2014; 86:727-35. [PMID: 25217496 PMCID: PMC4244597 DOI: 10.1124/mol.114.093666] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 09/12/2014] [Indexed: 11/22/2022] Open
Abstract
Cytochrome P450 2D6 (CYP2D6), a major drug-metabolizing enzyme, is responsible for metabolism of approximately 25% of marketed drugs. Clinical evidence indicates that metabolism of CYP2D6 substrates is increased during pregnancy, but the underlying mechanisms remain unclear. To identify transcription factors potentially responsible for CYP2D6 induction during pregnancy, a panel of genes differentially expressed in the livers of pregnant versus nonpregnant CYP2D6-humanized (tg-CYP2D6) mice was compiled via microarray experiments followed by real-time quantitative reverse-transcription polymerase chain reaction(qRT-PCR) verification. As a result, seven transcription factors-activating transcription factor 5 (ATF5), early growth response 1 (EGR1), forkhead box protein A3 (FOXA3), JUNB, Krüppel-like factor 9 (KLF9), KLF10, and REV-ERBα-were found to be up-regulated in liver during pregnancy. Results from transient transfection and promoter reporter gene assays indicate that KLF9 itself is a weak transactivator of CYP2D6 promoter but significantly enhances CYP2D6 promoter transactivation by hepatocyte nuclear factor 4 (HNF4α), a known transcriptional activator of CYP2D6 expression. The results from deletion and mutation analysis of CYP2D6 promoter activity identified a KLF9 putative binding motif at -22/-14 region to be critical in the potentiation of HNF4α-induced transactivation of CYP2D6. Electrophoretic mobility shift assays revealed a direct binding of KLF9 to the putative KLF binding motif. Results from chromatin immunoprecipitation assay showed increased recruitment of KLF9 to CYP2D6 promoter in the livers of tg-CYP2D6 mice during pregnancy. Taken together, our data suggest that increased KLF9 expression is in part responsible for CYP2D6 induction during pregnancy via the potentiation of HNF4α transactivation of CYP2D6.
Collapse
Affiliation(s)
- Kwi Hye Koh
- Department of Pharmacy Practice (K.H.K., H.J.), Department of Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, and Department of Pediatrics (W.Z.) and Department of Microbiology and Immunology (A.M.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Laboratory of Epigenetics and Chromatin Dynamics, and Departments of Biology and Molecular Biology, Epigenomics Translational Program, Mayo Clinic Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota (R.U.)
| | - Xian Pan
- Department of Pharmacy Practice (K.H.K., H.J.), Department of Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, and Department of Pediatrics (W.Z.) and Department of Microbiology and Immunology (A.M.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Laboratory of Epigenetics and Chromatin Dynamics, and Departments of Biology and Molecular Biology, Epigenomics Translational Program, Mayo Clinic Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota (R.U.)
| | - Wei Zhang
- Department of Pharmacy Practice (K.H.K., H.J.), Department of Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, and Department of Pediatrics (W.Z.) and Department of Microbiology and Immunology (A.M.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Laboratory of Epigenetics and Chromatin Dynamics, and Departments of Biology and Molecular Biology, Epigenomics Translational Program, Mayo Clinic Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota (R.U.)
| | - Alan McLachlan
- Department of Pharmacy Practice (K.H.K., H.J.), Department of Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, and Department of Pediatrics (W.Z.) and Department of Microbiology and Immunology (A.M.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Laboratory of Epigenetics and Chromatin Dynamics, and Departments of Biology and Molecular Biology, Epigenomics Translational Program, Mayo Clinic Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota (R.U.)
| | - Raul Urrutia
- Department of Pharmacy Practice (K.H.K., H.J.), Department of Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, and Department of Pediatrics (W.Z.) and Department of Microbiology and Immunology (A.M.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Laboratory of Epigenetics and Chromatin Dynamics, and Departments of Biology and Molecular Biology, Epigenomics Translational Program, Mayo Clinic Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota (R.U.)
| | - Hyunyoung Jeong
- Department of Pharmacy Practice (K.H.K., H.J.), Department of Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, and Department of Pediatrics (W.Z.) and Department of Microbiology and Immunology (A.M.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Laboratory of Epigenetics and Chromatin Dynamics, and Departments of Biology and Molecular Biology, Epigenomics Translational Program, Mayo Clinic Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota (R.U.)
| |
Collapse
|
21
|
Nagao H, Sasaki M, Imazu T, Takahashi K, Aoki H, Minato K. Effects of triiodothyronine on turnover rate and metabolizing enzymes for thyroxine in thyroidectomized rats. Life Sci 2014; 116:74-82. [DOI: 10.1016/j.lfs.2014.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/09/2014] [Accepted: 09/13/2014] [Indexed: 01/14/2023]
|
22
|
The associations between immunity-related genes and breast cancer prognosis in Korean women. PLoS One 2014; 9:e103593. [PMID: 25075970 PMCID: PMC4116221 DOI: 10.1371/journal.pone.0103593] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/02/2014] [Indexed: 12/15/2022] Open
Abstract
We investigated the role of common genetic variation in immune-related genes on breast cancer disease-free survival (DFS) in Korean women. 107 breast cancer patients of the Seoul Breast Cancer Study (SEBCS) were selected for this study. A total of 2,432 tag single nucleotide polymorphisms (SNPs) in 283 immune-related genes were genotyped with the GoldenGate Oligonucleotide pool assay (OPA). A multivariate Cox-proportional hazard model and polygenic risk score model were used to estimate the effects of SNPs on breast cancer prognosis. Harrell’s C index was calculated to estimate the predictive accuracy of polygenic risk score model. Subsequently, an extended gene set enrichment analysis (GSEA-SNP) was conducted to approximate the biological pathway. In addition, to confirm our results with current evidence, previous studies were systematically reviewed. Sixty-two SNPs were statistically significant at p-value less than 0.05. The most significant SNPs were rs1952438 in SOCS4 gene (hazard ratio (HR) = 11.99, 95% CI = 3.62–39.72, P = 4.84E-05), rs2289278 in TSLP gene (HR = 4.25, 95% CI = 2.10–8.62, P = 5.99E-05) and rs2074724 in HGF gene (HR = 4.63, 95% CI = 2.18–9.87, P = 7.04E-05). In the polygenic risk score model, the HR of women in the 3rd tertile was 6.78 (95% CI = 1.48–31.06) compared to patients in the 1st tertile of polygenic risk score. Harrell’s C index was 0.813 with total patients and 0.924 in 4-fold cross validation. In the pathway analysis, 18 pathways were significantly associated with breast cancer prognosis (P<0.1). The IL-6R, IL-8, IL-10RB, IL-12A, and IL-12B was associated with the prognosis of cancer in data of both our study and a previous study. Therefore, our results suggest that genetic polymorphisms in immune-related genes have relevance to breast cancer prognosis among Korean women.
Collapse
|
23
|
Knoedler JR, Denver RJ. Krüppel-like factors are effectors of nuclear receptor signaling. Gen Comp Endocrinol 2014; 203:49-59. [PMID: 24642391 PMCID: PMC4339045 DOI: 10.1016/j.ygcen.2014.03.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 02/28/2014] [Accepted: 03/02/2014] [Indexed: 01/09/2023]
Abstract
Binding of steroid and thyroid hormones to their cognate nuclear receptors (NRs) impacts virtually every aspect of postembryonic development, physiology and behavior, and inappropriate signaling by NRs may contribute to disease. While NRs regulate genes by direct binding to hormone response elements in the genome, their actions may depend on the activity of other transcription factors (TFs) that may or may not bind DNA. The Krüppel-like family of transcription factors (KLF) is an evolutionarily conserved class of DNA-binding proteins that influence many aspects of development and physiology. Several members of this family have been shown to play diverse roles in NR signaling. For example, KLFs (1) act as accessory transcription factors for NR actions, (2) regulate expression of NR genes, and (3) as gene products of primary NR response genes function as key players in NR-dependent transcriptional networks. In mouse models, deletion of different KLFs leads to aberrant transcriptional and physiological responses to hormones, underscoring the importance of these proteins in the regulation of hormonal signaling. Understanding the functional relationships between NRs and KLFs will yield important insights into mechanisms of NR signaling. In this review we present a conceptual framework for understanding how KLFs participate in NR signaling, and we provide examples of how these proteins function to effect hormone action.
Collapse
Affiliation(s)
- Joseph R Knoedler
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Robert J Denver
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109-1048, USA; Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA.
| |
Collapse
|
24
|
Cold-induced changes in gene expression in brown adipose tissue, white adipose tissue and liver. PLoS One 2013; 8:e68933. [PMID: 23894377 PMCID: PMC3718809 DOI: 10.1371/journal.pone.0068933] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 06/05/2013] [Indexed: 01/30/2023] Open
Abstract
Cold exposure imposes a metabolic challenge to mammals that is met by a coordinated response in different tissues to prevent hypothermia. This study reports a transcriptomic analysis in brown adipose tissue (BAT), white adipose (WAT) and liver of mice in response to 24 h cold exposure at 8°C. Expression of 1895 genes were significantly (P<0.05) up- or down-regulated more than two fold by cold exposure in all tissues but only 5 of these genes were shared by all three tissues, and only 19, 14 and 134 genes were common between WAT and BAT, WAT and liver, and BAT and liver, respectively. We confirmed using qRT-PCR, the increased expression of a number of characteristic BAT genes during cold exposure. In both BAT and the liver, the most common direction of change in gene expression was suppression (496 genes in BAT and 590 genes in liver). Gene ontology analysis revealed for the first time significant (P<0.05) down regulation in response to cold, of genes involved in oxidoreductase activity, lipid metabolic processes and protease inhibitor activity, in both BAT and liver, but not WAT. The results reveal an unexpected importance of down regulation of cytochrome P450 gene expression and apolipoprotein, in both BAT and liver, but not WAT, in response to cold exposure. Pathway analysis suggests a model in which down regulation of the nuclear transcription factors HNF4α and PPARα in both BAT and liver may orchestrate the down regulation of genes involved in lipoprotein and steroid metabolism as well as Phase I enzymes belonging to the cytochrome P450 group in response to cold stress in mice. We propose that the response to cold stress involves decreased gene expression in a range of cellular processes in order to maximise pathways involved in heat production.
Collapse
|
25
|
Kogiso T, Nagahara H, Otsuka M, Shiratori K, Dowdy SF. Transdifferentiation of human fibroblasts into hepatocyte-like cells by defined transcriptional factors. Hepatol Int 2013; 7:937-944. [DOI: 10.1007/s12072-013-9432-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
26
|
Roy P, Kumar B, Shende A, Singh A, Meena A, Ghosal R, Ranganathan M, Bandyopadhyay A. A genome-wide screen indicates correlation between differentiation and expression of metabolism related genes. PLoS One 2013; 8:e63670. [PMID: 23717462 PMCID: PMC3661535 DOI: 10.1371/journal.pone.0063670] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 04/04/2013] [Indexed: 01/07/2023] Open
Abstract
Differentiated tissues may be considered as materials with distinct properties. The differentiation program of a given tissue ensures that it acquires material properties commensurate with its function. It may be hypothesized that some of these properties are acquired through production of tissue-specific metabolites synthesized by metabolic enzymes. To establish correlation between metabolism and organogenesis we have carried out a genome-wide expression study of metabolism related genes by RNA in-situ hybridization. 23% of the metabolism related genes studied are expressed in a tissue-restricted but not tissue-exclusive manner. We have conducted the screen on whole mount chicken (Gallus gallus) embryos from four distinct developmental stages to correlate dynamic changes in expression patterns of metabolic enzymes with spatio-temporally unique developmental events. Our data strongly suggests that unique combinations of metabolism related genes, and not specific metabolic pathways, are upregulated during differentiation. Further, expression of metabolism related genes in well established signaling centers that regulate different aspects of morphogenesis indicates developmental roles of some of the metabolism related genes. The database of tissue-restricted expression patterns of metabolism related genes, generated in this study, should serve as a resource for systematic identification of these genes with tissue-specific functions during development. Finally, comprehensive understanding of differentiation is not possible unless the downstream genes of a differentiation cascade are identified. We propose, metabolic enzymes constitute a significant portion of these downstream target genes. Thus our study should help elucidate different aspects of tissue differentiation.
Collapse
Affiliation(s)
- Priti Roy
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, U.P., India
| | - Brijesh Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, U.P., India
| | - Akhilesh Shende
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, U.P., India
| | - Anupama Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, U.P., India
| | - Anil Meena
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, U.P., India
| | - Ritika Ghosal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, U.P., India
| | - Madhav Ranganathan
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, U.P., India
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, U.P., India
- * E-mail:
| |
Collapse
|
27
|
Transcriptional regulation of PCFT by KLF4, HNF4α, CDX2 and C/EBPα: implication in its site-specific expression in the small intestine. Biochem Biophys Res Commun 2013; 431:158-63. [PMID: 23313509 DOI: 10.1016/j.bbrc.2013.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 01/03/2013] [Indexed: 01/10/2023]
Abstract
Proton-coupled folate transporter (PCFT), which is responsible for the intestinal uptake of folates and analogs, is expressed only in the proximal region in the small intestine. The present study was to examine its transcriptional regulation, which may be involved in such a unique expression profile and potentially in its alteration, using dual-luciferase reporter assays in human embryonic kidney (HEK) 293 cells. The luciferase activity derived from the reporter construct containing the 5'-flanking sequence of -1695/+96 of the human PCFT gene was enhanced most extensively by the introduction of Krüppel-like factor 4 (KLF4). The KLF4-induced luciferase activity was further enhanced by hepatocyte nuclear factor 4α (HNF4α) synergistically. To the contrary, caudal-type homeobox transcription factor 2 (CDX2) and CCAAT/enhancer-binding protein α (C/EBPα) extensively suppressed the luciferase activity induced by KLF4 alone and also that induced by KLF4 and HNF4α. Western blot analysis using the rat small intestine indicated uniform expression of KLF4 along the intestinal tract, proximal-oriented expression of HNF4α, distal-oriented expression of CDX2 and C/EBPα. These results suggest that the activity of PCFT promoter is basically induced by KLF4 and the gradiented expression profile of PCFT may be at least in part accounted for by those of HNF4α, CDX2 and C/EBPα.
Collapse
|
28
|
Thyroid hormone receptors: the challenge of elucidating isotype-specific functions and cell-specific response. Biochim Biophys Acta Gen Subj 2012; 1830:3900-7. [PMID: 22704954 DOI: 10.1016/j.bbagen.2012.06.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 06/04/2012] [Accepted: 06/05/2012] [Indexed: 02/06/2023]
Abstract
BACKGROUND Thyroid hormone receptors TRα1, TRβ1 and TRβ2 are broadly expressed and exert a pleiotropic influence on many developmental and homeostatic processes. Extensive genetic studies in mice precisely defined their respective function. SCOPE OF REVIEW The purpose of the review is to discuss two puzzling issues: MAJOR CONCLUSIONS Mouse genetics support a balanced contribution of expression pattern and receptor intrinsic properties in defining the receptor respective functions. The molecular mechanisms sustaining cell specific response remain hypothetical and based on studies performed with other nuclear receptors. GENERAL SIGNIFICANCE The isoform-specificity and cell-specificity questions have many implications for clinical research, drug development, and endocrine disruptor studies. This article is part of a Special Issue entitled Thyroid hormone signalling.
Collapse
|
29
|
Kanamoto N, Tagami T, Ueda-Sakane Y, Sone M, Miura M, Yasoda A, Tamura N, Arai H, Nakao K. Forkhead box A1 (FOXA1) and A2 (FOXA2) oppositely regulate human type 1 iodothyronine deiodinase gene in liver. Endocrinology 2012; 153:492-500. [PMID: 22067325 DOI: 10.1210/en.2011-1310] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Type 1 iodothyronine deiodinase (D1), a selenoenzyme that catalyzes the bioactivation of thyroid hormone, is expressed mainly in the liver. Its expression and activity are modulated by several factors, but the precise mechanism of its transcriptional regulation remains unclear. In the present study, we have analyzed the promoter of human D1 gene (hDIO1) to identify factors that prevalently increase D1 activity in the human liver. Deletion and mutation analyses demonstrated that a forkhead box (FOX)A binding site and an E-box site within the region between nucleotides -187 and -132 are important for hDIO1 promoter activity in the liver. EMSA demonstrated that FOXA1 and FOXA2 specifically bind to the FOXA binding site and that upstream stimulatory factor (USF) specifically binds to the E-box element. Overexpression of FOXA2 decreased hDIO1 promoter activity, and short interfering RNA-mediated knockdown of FOXA2 increased the expression of hDIO1 mRNA. In contrast, overexpression of USF1/2 increased hDIO1 promoter activity. Short interfering RNA-mediated knockdown of FOXA1 decreased the expression of hDIO1 mRNA, but knockdown of both FOXA1 and FOXA2 restored it. The response of the hDIO1 promoter to USF was greatly attenuated in the absence of FOXA1. Taken together, these results indicate that a balance of FOXA1 and FOXA2 expression modulates hDIO1 expression in the liver.
Collapse
Affiliation(s)
- Naotetsu Kanamoto
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bolotin E, Liao H, Chi Ta T, Yang C, Hwang-Verslues W, Evans JR, Jiang T, Sladek FM. Integrated approach for the identification of human hepatocyte nuclear factor 4alpha target genes using protein binding microarrays. Hepatology 2010; 51:642-53. [PMID: 20054869 PMCID: PMC3581146 DOI: 10.1002/hep.23357] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
UNLABELLED Hepatocyte nuclear factor 4 alpha (HNF4alpha), a member of the nuclear receptor superfamily, is essential for liver function and is linked to several diseases including diabetes, hemophilia, atherosclerosis, and hepatitis. Although many DNA response elements and target genes have been identified for HNF4alpha, the complete repertoire of binding sites and target genes in the human genome is unknown. Here, we adapt protein binding microarrays (PBMs) to examine the DNA-binding characteristics of two HNF4alpha species (rat and human) and isoforms (HNF4alpha2 and HNF4alpha8) in a high-throughput fashion. We identified approximately 1400 new binding sequences and used this dataset to successfully train a Support Vector Machine (SVM) model that predicts an additional approximately 10,000 unique HNF4alpha-binding sequences; we also identify new rules for HNF4alpha DNA binding. We performed expression profiling of an HNF4alpha RNA interference knockdown in HepG2 cells and compared the results to a search of the promoters of all human genes with the PBM and SVM models, as well as published genome-wide location analysis. Using this integrated approach, we identified approximately 240 new direct HNF4alpha human target genes, including new functional categories of genes not typically associated with HNF4alpha, such as cell cycle, immune function, apoptosis, stress response, and other cancer-related genes. CONCLUSION We report the first use of PBMs with a full-length liver-enriched transcription factor and greatly expand the repertoire of HNF4alpha-binding sequences and target genes, thereby identifying new functions for HNF4alpha. We also establish a web-based tool, HNF4 Motif Finder, that can be used to identify potential HNF4alpha-binding sites in any sequence.
Collapse
Affiliation(s)
- Eugene Bolotin
- Genetics, Genomics and Bioinformatics Graduate Program, University of California Riverside, Riverside, CA
| | - Hailing Liao
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, CA
| | - Tuong Chi Ta
- Cell, Molecular, and Developmental Biology Graduate Program, University of California Riverside, Riverside, CA
| | - Chuhu Yang
- Genetics, Genomics and Bioinformatics Graduate Program, University of California Riverside, Riverside, CA
| | - Wendy Hwang-Verslues
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, CA
| | - Jane R. Evans
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, CA
| | - Tao Jiang
- Department of Computer Science and Engineering, University of California Riverside, Riverside, CA,Institute for Integrated Genome Biology (also at UCR)
| | - Frances M. Sladek
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, CA,Institute for Integrated Genome Biology (also at UCR)
| |
Collapse
|
31
|
Sugrue ML, Vella KR, Morales C, Lopez ME, Hollenberg AN. The thyrotropin-releasing hormone gene is regulated by thyroid hormone at the level of transcription in vivo. Endocrinology 2010; 151:793-801. [PMID: 20032051 PMCID: PMC2817611 DOI: 10.1210/en.2009-0976] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The expression of the TRH gene in the paraventricular nucleus (PVH) of the hypothalamus is required for the normal production of thyroid hormone (TH) in rodents and humans. In addition, the regulation of TRH mRNA expression by TH, specifically in the PVH, ensures tight control of the set point of the hypothalamic-pituitary-thyroid axis. Although many studies have assumed that the regulation of TRH expression by TH is at the level of transcription, there is little data available to demonstrate this. We used two in vivo model systems to show this. In the first model system, we developed an in situ hybridization (ISH) assay directed against TRH heteronuclear RNA to measure TRH transcription directly in vivo. We show that in the euthyroid state, TRH transcription is present both in the PVH and anterior/lateral hypothalamus. In the hypothyroid state, transcription is activated in the PVH only and can be shut off within 5 h by TH. In the second model system, we employed transgenic mice that express the Cre recombinase under the control of the genomic region containing the TRH gene. Remarkably, TH regulates Cre expression in these mice in the PVH only. Taken together, these data affirm that TH regulates TRH at the level of transcription in the PVH only and that genomic elements surrounding the TRH gene mediate its regulation by T(3). Thus, it should be possible to identify the elements within the TRH locus that mediate its regulation by T(3) using in vivo approaches.
Collapse
Affiliation(s)
- Michelle L Sugrue
- Division of Endocrinology, Metabolism, and Diabetes, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, E/CLS-0738, Boston, Massachusetts 02215, USA
| | | | | | | | | |
Collapse
|
32
|
Inagaki T, Tachibana M, Magoori K, Kudo H, Tanaka T, Okamura M, Naito M, Kodama T, Shinkai Y, Sakai J. Obesity and metabolic syndrome in histone demethylase JHDM2a-deficient mice. Genes Cells 2009; 14:991-1001. [DOI: 10.1111/j.1365-2443.2009.01326.x] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
33
|
Stoytcheva ZR, Berry MJ. Transcriptional regulation of mammalian selenoprotein expression. Biochim Biophys Acta Gen Subj 2009; 1790:1429-40. [PMID: 19465084 DOI: 10.1016/j.bbagen.2009.05.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 05/05/2009] [Accepted: 05/18/2009] [Indexed: 01/23/2023]
Abstract
BACKGROUND Selenoproteins contain the twenty-first amino acid, selenocysteine, and are involved in cellular defenses against oxidative damage, important metabolic and developmental pathways, and responses to environmental challenges. Elucidating the mechanisms regulating selenoprotein expression at the transcriptional level is a key to understanding how these mechanisms are called into play to respond to the changing environment. METHODS This review summarizes published studies on transcriptional regulation of selenoprotein genes, focused primarily on genes whose encoded protein functions are at least partially understood. This is followed by in silico analysis of predicted regulatory elements in selenoprotein genes, including those in the aforementioned category as well as the genes whose functions are not known. RESULTS Our findings reveal regulatory pathways common to many selenoprotein genes, including several involved in stress-responses. In addition, tissue-specific regulatory factors are implicated in regulating many selenoprotein genes. CONCLUSIONS These studies provide new insights into how selenoprotein genes respond to environmental and other challenges, and the roles these proteins play in allowing cells to adapt to these changes. GENERAL SIGNIFICANCE Elucidating the regulatory mechanisms affecting selenoprotein expression is essential for understanding their roles in human diseases, and for developing diagnostic and potential therapeutic approaches to address dysregulation of members of this gene family.
Collapse
Affiliation(s)
- Zoia R Stoytcheva
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Suite 222, Honolulu, HI 96813, USA
| | | |
Collapse
|
34
|
The peroxisome proliferator-activated receptor gamma/retinoid X receptor alpha heterodimer targets the histone modification enzyme PR-Set7/Setd8 gene and regulates adipogenesis through a positive feedback loop. Mol Cell Biol 2009; 29:3544-55. [PMID: 19414603 DOI: 10.1128/mcb.01856-08] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Control of cell differentiation occurs through transcriptional mechanisms and through epigenetic modification. Using a chromatin immunoprecipitation-on-chip approach, we performed a genome-wide search for target genes of peroxisome proliferator-activated receptor gamma (PPAR gamma) and its partner protein retinoid X receptor alpha during adipogenesis. We show that these two receptors target several genes that encode histone lysine methyltransferase SET domain proteins. The histone H4 Lys 20 (H4K20) monomethyltransferase PR-Set7/Setd8 gene is upregulated by PPAR gamma during adipogenesis, and the knockdown of PR-Set7/Setd8 suppressed adipogenesis. Intriguingly, monomethylated H4K20 (H4K20me1) levels are robustly increased toward the end of differentiation. PR-Set7/Setd8 positively regulates the expression of PPAR gamma and its targets through H4K20 monomethylation. Furthermore, the activation of PPAR gamma transcriptional activity leads to the induction of H4K20me1 modification of PPAR gamma and its targets and thereby promotes adipogenesis. We also show that PPAR gamma targets PPAR gamma2 and promotes its gene expression through H4K20 monomethylation. Our results connect transcriptional regulation and epigenetic chromatin modulation through H4K20 monomethylation during adipogenesis through a feedback loop.
Collapse
|