1
|
Soares De Oliveira L, Ritter MJ. Thyroid hormone and the Liver. Hepatol Commun 2025; 9:e0596. [PMID: 39699315 PMCID: PMC11661762 DOI: 10.1097/hc9.0000000000000596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/20/2024] Open
Abstract
It is known that thyroid hormone can regulate hepatic metabolic pathways including cholesterol, de novo lipogenesis, fatty acid oxidation, lipophagy, and carbohydrate metabolism. Thyroid hormone action is mediated by the thyroid hormone receptor (THR) isoforms and their coregulators, and THRβ is the main isoform expressed in the liver. Dysregulation of thyroid hormone levels, as seen in hypothyroidism, has been associated with dyslipidemia and metabolic dysfunction-associated fatty liver disease. Given the beneficial effects of thyroid hormone in liver metabolism and the advances illuminating the use of thyroid hormone analogs such as resmetirom as therapeutic agents in the treatment of metabolic dysfunction-associated fatty liver disease, this review aims to further explore the relationship between TH, the liver, and metabolic dysfunction-associated fatty liver disease. Herein, we summarize the current clinical therapies and highlight future areas of research.
Collapse
|
2
|
Soares De Oliveira L, Kaserman JE, Van Der Spek AH, Lee NJ, Undeutsch HJ, Werder RB, Wilson AA, Hollenberg AN. Thyroid hormone receptor beta (THRβ1) is the major regulator of T3 action in human iPSC-derived hepatocytes. Mol Metab 2024; 90:102057. [PMID: 39481850 PMCID: PMC11615914 DOI: 10.1016/j.molmet.2024.102057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/08/2024] [Accepted: 10/24/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE Thyroid hormone (TH) action is mediated by thyroid hormone receptor (THR) isoforms. While THRβ1 is likely the main isoform expressed in liver, its role in human hepatocytes is not fully understood. METHODS To elucidate the role of THRβ1 action in human hepatocytes we used CRISPR/Cas9 editing to knock out THRβ1 in induced pluripotent stem cells (iPSC). Following directed differentiation to the hepatic lineage, iPSC-derived hepatocytes were then interrogated to determine the role of THRβ1 in ligand-independent and -dependent functions. RESULTS We found that the loss of THRβ1 promoted alterations in proliferation rate and metabolic pathways regulated by T3, including gluconeogenesis, lipid oxidation, fatty acid synthesis, and fatty acid uptake. We observed that key genes involved in liver metabolism are regulated through both T3 ligand-dependent and -independent THRβ1 signaling mechanisms. Finally, we demonstrate that following THRβ1 knockout, several key metabolic genes remain T3 responsive suggesting they are THRα targets. CONCLUSIONS These results highlight that iPSC-derived hepatocytes are an effective platform to study mechanisms regulating TH signaling in human hepatocytes.
Collapse
Affiliation(s)
- Lorraine Soares De Oliveira
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Joseph E Kaserman
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA; Department of Medicine, Section of Pulmonary and Critical Care Medicine, Chobanian & Avedisian School of Medicine, Boston Medical Center, MA 02118, USA
| | - Anne H Van Der Spek
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam 1081 HV, Netherlands
| | - Nora J Lee
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Hendrik J Undeutsch
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Rhiannon B Werder
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Andrew A Wilson
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA; Department of Medicine, Section of Pulmonary and Critical Care Medicine, Chobanian & Avedisian School of Medicine, Boston Medical Center, MA 02118, USA.
| | - Anthony N Hollenberg
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA.
| |
Collapse
|
3
|
Katz LS, Argmann C, Lambertini L, Scott DK. T3 and glucose increase expression of phosphoenolpyruvate carboxykinase (PCK1) leading to increased β-cell proliferation. Mol Metab 2022; 66:101646. [PMID: 36455788 PMCID: PMC9731891 DOI: 10.1016/j.molmet.2022.101646] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Thyroid hormone (T3) and high glucose concentrations are critical components of β-cell maturation and function. In the present study, we asked whether T3 and glucose signaling pathways coordinately regulate transcription of genes important for β-cell function and proliferation. METHODS RNA-seq analysis was performed on cadaveric human islets from five different donors in response to low and high glucose concentrations and in the presence or absence of T3. Gene expression was also studies in sorted human β-cells, mouse islets and Ins-1 cells by RT-qPCR. Silencing of the thyroid hormone receptors (THR) was conducted using lentiviruses. Proliferation was assessed by ki67 immunostaining in primary human/mouse islets. Chromatin immunoprecipitation and proximity ligation assay were preformed to validate interactions of ChREBP and THR. RESULTS We found glucose-mediated expression of carbohydrate response element binding protein alpha and beta (ChREBPα and ChREBPβ) mRNAs and their target genes are highly dependent on T3 concentrations in rodent and human β-cells. In β-cells, T3 and glucose coordinately regulate the expression of ChREBPβ and PCK1 (phosphoenolpyruvate carboxykinase-1) among other important genes for β-cell maturation. Additionally, we show the thyroid hormone receptor (THR) and ChREBP interact, and their relative response elements are located near to each other on mutually responsive genes. In FACS-sorted adult human β-cells, we found that high concentrations of glucose and T3 induced the expression of PCK1. Next, we show that overexpression of Pck1 together with dimethyl malate (DMM), a substrate precursor, significantly increased β-cell proliferation in human islets. Finally, using a Cre-Lox approach, we demonstrated that ChREBPβ contributes to Pck1-dependent β-cell proliferation in mouse β-cells. CONCLUSIONS We conclude that T3 and glucose act together to regulate ChREBPβ, leading to increased expression and activity of Pck1, and ultimately increased β-cell proliferation.
Collapse
Affiliation(s)
- Liora S Katz
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Carmen Argmann
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luca Lambertini
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
4
|
Su X, Chen X, Wang B. Relationship between the development of hyperlipidemia in hypothyroidism patients. Mol Biol Rep 2022; 49:11025-11035. [PMID: 36097119 DOI: 10.1007/s11033-022-07423-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022]
Abstract
As shown in the previous studies, hypothyroidism (HT) is identified to be closely associated with the elevated plasma levels of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), triglyceride (TG), and with the decreased plasma levels of high density lipoprotein cholesterol (HDL-C). On the other hand, the thyroid hormone (TH), which has been considered as a vital hormone produced and released by the thyroid gland, are well-established to regulate the metabolism of plasma TC; whereas other evidence proposed that the thyroid-stimulating hormone (TSH) also regulated the plasma cholesterol metabolism independently of the TH, which further promotes the progression of hyperlipidemia. Nevertheless, the potential mechanism is still not illustrated. It is worth noting that several studies has found that the progression of HT-induced hyperlipidemia might be associated with the down-regulated plasma levels of TH and the up-regulated plasma levels of TSH, revealing that HT could promote hyperlipidemia and its related cardio-metabolic disorders. Otherwise, multiple novel identified plasma proteins, such as proprotein convertase subtilisin/kexin type 9 (PCSK9), angiopoietin-like protein (ANGPTLs), and fibroblast growth factors (FGFs), have also been demonstrated to embrace a vital function in modulating the progression of hyperlipidemia induced by HT. In the present comprehensive review, the recent findings which elucidated the association of HT and the progression of hyperlipidemia were summarized. Furthermore, other results which illustrated the underlying mechanisms by which HT facilitates the progression of hyperlipidemia and its cardio-metabolic disorders are also listed in the current review.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, 361000, Xiamen, Fujian, China
| | - Xiang Chen
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, 361000, Xiamen, Fujian, China.
| | - Bin Wang
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, 361000, Xiamen, Fujian, China.
| |
Collapse
|
5
|
Cross-Talk between the Cytokine IL-37 and Thyroid Hormones in Modulating Chronic Inflammation Associated with Target Organ Damage in Age-Related Metabolic and Vascular Conditions. Int J Mol Sci 2022; 23:ijms23126456. [PMID: 35742902 PMCID: PMC9224418 DOI: 10.3390/ijms23126456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic inflammation is considered to be the main mechanism contributing to the development of age-related metabolic and vascular conditions. The phases of chronic inflammation that mediate the progression of target organ damage in these conditions are poorly known, however. In particular, there is a paucity of data on the link between chronic inflammation and metabolic disorders. Based on some of our own results and recent developments in our understanding of age-related inflammation as a whole-body response, we discuss the hypothesis that cross-talk between the cytokine IL-37 and thyroid hormones could be the key regulatory mechanism that justifies the metabolic effects of chronic tissue-related inflammation. The cytokine IL-37 is emerging as a strong natural suppressor of the chronic innate immune response. The effect of this cytokine has been identified in reversing metabolic costs of chronic inflammation. Thyroid hormones are known to regulate energy metabolism. There is a close link between thyroid function and inflammation in elderly individuals. Nonlinear associations between IL-37 and thyroid hormones, considered within the wider clinical context, can improve our understanding of the phases of chronic inflammation that are associated with target organ damage in age-related metabolic and vascular conditions.
Collapse
|
6
|
Liu H, Peng D. Update on dyslipidemia in hypothyroidism: the mechanism of dyslipidemia in hypothyroidism. Endocr Connect 2022; 11:e210002. [PMID: 35015703 PMCID: PMC8859969 DOI: 10.1530/ec-21-0002] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/11/2022] [Indexed: 11/18/2022]
Abstract
Hypothyroidism is often associated with elevated serum levels of total cholesterol, LDL-C and triglycerides. Thyroid hormone (TH) affects the production, clearance and transformation of cholesterol, but current research shows that thyroid-stimulating hormone (TSH) also participates in lipid metabolism independently of TH. Therefore, the mechanism of hypothyroidism-related dyslipidemia is associated with the decrease of TH and the increase of TSH levels. Some newly identified regulatory factors, such as proprotein convertase subtilisin/kexin type 9, angiogenin-like proteins and fibroblast growth factors are the underlying causes of dyslipidemia in hypothyroidism. HDL serum concentration changes were not consistent, and its function was reportedly impaired. The current review focuses on the updated understanding of the mechanism of hypothyroidism-related dyslipidemia.
Collapse
Affiliation(s)
- Huixing Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Correspondence should be addressed to D Peng:
| |
Collapse
|
7
|
Hyperlipidemia and hypothyroidism. Clin Chim Acta 2022; 527:61-70. [DOI: 10.1016/j.cca.2022.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 12/16/2022]
|
8
|
Mendoza A, Tang C, Choi J, Acuña M, Logan M, Martin AG, Al-Sowaimel L, Desai BN, Tenen DE, Jacobs C, Lyubetskaya A, Fu Y, Liu H, Tsai L, Cohen DE, Forrest D, Wilson AA, Hollenberg AN. Thyroid hormone signaling promotes hepatic lipogenesis through the transcription factor ChREBP. Sci Signal 2021; 14:eabh3839. [PMID: 34784250 DOI: 10.1126/scisignal.abh3839] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thyroid hormone (TH) action is essential for hepatic lipid synthesis and oxidation. Analysis of hepatocyte-specific thyroid receptor β1 (TRβ1) knockout mice confirmed a role for TH in stimulating de novo lipogenesis and fatty acid oxidation through its nuclear receptor. Specifically, TRβ1 and its principal corepressor NCoR1 in hepatocytes repressed de novo lipogenesis, whereas the TH-mediated induction of lipogenic genes depended on the transcription factor ChREBP. Mice with a hepatocyte-specific deficiency in ChREBP lost TH-mediated stimulation of the lipogenic program, which, in turn, impaired the regulation of fatty acid oxidation. TH regulated ChREBP activation and recruitment to DNA, revealing a mechanism by which TH regulates specific signaling pathways. Regulation of the lipogenic pathway by TH through ChREBP was conserved in hepatocytes derived from human induced pluripotent stem cells. These results demonstrate that TH signaling in the liver acts simultaneously to enhance both lipogenesis and fatty acid oxidation.
Collapse
Affiliation(s)
- Arturo Mendoza
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Catherine Tang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Jinyoung Choi
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Mariana Acuña
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Maya Logan
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Adriana G Martin
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Lujain Al-Sowaimel
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Bhavna N Desai
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Danielle E Tenen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Christopher Jacobs
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Anna Lyubetskaya
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Yulong Fu
- Laboratory of Endocrinology and Receptor Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hong Liu
- Laboratory of Endocrinology and Receptor Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Linus Tsai
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - David E Cohen
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew A Wilson
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| |
Collapse
|
9
|
Li L, Sakiyama H, Eguchi H, Yoshihara D, Fujiwara N, Suzuki K. Activation of the mitogen-activated protein kinase ERK1/2 signaling pathway suppresses the expression of ChREBPα and β in HepG2 cells. FEBS Open Bio 2021; 11:2008-2018. [PMID: 34051057 PMCID: PMC8255832 DOI: 10.1002/2211-5463.13208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/07/2021] [Accepted: 05/26/2021] [Indexed: 11/25/2022] Open
Abstract
The carbohydrate response element‐binding protein (ChREBP), a glucose‐responsive transcription factor that plays a critical role in the glucose‐mediated induction of genes involved in hepatic glycolysis and lipogenesis, exists as two isoforms: ChREBPα and ChREBPβ. However, the mechanism responsible for regulating the expression of both ChREBPα and β, as well as the mechanism that determines which specific isoform is more responsive to different stimuli, remains unclear. To address this issue, we compared the effects of several stimuli, including oxidative stress, on the mRNA and protein expression levels of ChREBPα and β in the hepatocyte cell line, HepG2. We found that H2O2 stimulation suppressed the expression of both mRNA and protein in HepG2 cells, but the mRNA expression level of ChREBPβ was < 1% of that for ChREBPα levels. In addition, the reduction in both ChREBPα and β mRNA levels was reversed by PD98059, a selective and cell permeable inhibitor of the MEK/ERK pathway. Additionally, the administration of 12‐O‐tetradecanoylphorbol 13‐acetate (TPA) and staurosporine (STS), activators of extracellular‐signal‐regulated kinase (ERK) signaling, also resulted in a decrease in the levels of both ChREBPα and β mRNA in HepG2 cells through ERK signaling. These collective data suggest that oxidative stress, including STS treatment, suppresses the expression of ChREBPα and β via the activation of ERK signaling in HepG2 cells. Such a decrease in the levels of expression of ChREBPα and β could result in the suppression of hepatic glycolysis and lipogenesis, and this would be expected to prevent further oxidative stress.
Collapse
Affiliation(s)
- Lan Li
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Haruhiko Sakiyama
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hironobu Eguchi
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Daisaku Yoshihara
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Noriko Fujiwara
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Keiichiro Suzuki
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
10
|
Shi YN, Liu YJ, Xie Z, Zhang WJ. Fructose and metabolic diseases: too much to be good. Chin Med J (Engl) 2021; 134:1276-1285. [PMID: 34010200 PMCID: PMC8183764 DOI: 10.1097/cm9.0000000000001545] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Excessive consumption of fructose, the sweetest of all naturally occurring carbohydrates, has been linked to worldwide epidemics of metabolic diseases in humans, and it is considered an independent risk factor for cardiovascular diseases. We provide an overview about the features of fructose metabolism, as well as potential mechanisms by which excessive fructose intake is associated with the pathogenesis of metabolic diseases both in humans and rodents. To accomplish this aim, we focus on illuminating the cellular and molecular mechanisms of fructose metabolism as well as its signaling effects on metabolic and cardiovascular homeostasis in health and disease, highlighting the role of carbohydrate-responsive element-binding protein in regulating fructose metabolism.
Collapse
Affiliation(s)
- Ya-Nan Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
| | - Ya-Jin Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
| | - Zhifang Xie
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China
| | - Weiping J. Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
11
|
Ritter MJ, Amano I, Hollenberg AN. Thyroid Hormone Signaling and the Liver. Hepatology 2020; 72:742-752. [PMID: 32343421 DOI: 10.1002/hep.31296] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/30/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Thyroid hormone (TH) plays a critical role in maintaining metabolic homeostasis throughout life. It is well known that the liver and thyroid are intimately linked, with TH playing important roles in de novo lipogenesis, beta-oxidation (fatty acid oxidation), cholesterol metabolism, and carbohydrate metabolism. Indeed, patients with hypothyroidism have abnormal lipid panels with higher levels of low-density lipoprotein levels, triglycerides (triacylglycerol; TAG), and apolipoprotein B levels. Even in euthyroid patients, lower serum-free thyroxine levels are associated with higher total cholesterol levels, LDL, and TAG levels. In addition to abnormal serum lipids, the risk of nonalcoholic fatty liver disease (NAFLD) increases with lower free thyroxine levels. As free thyroxine rises, the risk of NAFLD is reduced. This has led to numerous animal studies and clinical trials investigating TH analogs and TH receptor agonists as potential therapies for NAFLD and hyperlipidemia. Thus, TH plays an important role in maintaining hepatic homeostasis, and this continues to be an important area of study. A review of TH action and TH actions on the liver will be presented here.
Collapse
Affiliation(s)
- Megan J Ritter
- Division of Endocrinology, Weill Cornell Medicine, New York, NY
| | - Izuki Amano
- Division of Endocrinology, Weill Cornell Medicine, New York, NY.,Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | |
Collapse
|
12
|
Huang P, Huang FZ, Liu HZ, Zhang TY, Yang MS, Sun CZ. LncRNA MEG3 functions as a ceRNA in regulating hepatic lipogenesis by competitively binding to miR-21 with LRP6. Metabolism 2019; 94:1-8. [PMID: 30711569 DOI: 10.1016/j.metabol.2019.01.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/23/2019] [Accepted: 01/30/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Hepatic lipogenesis dysregulation is essential for the development of non-alcoholic fatty liver disease (NAFLD). Emerging evidence indicates the importance of the involvement of long non-coding RNAs (LncRNAs) in lipogenesis. However, the specific mechanism underlying this process is not clear. OBJECTIVE This study aimed to investigate the functional implication of LncRNA MEG3 (MEG3) in fatty degeneration of hepatocytes and in the pathogenesis of NAFLD. METHODS The expression of MEG3 was analysed in in vitro and in vivo models of NAFLD, which were established by free fatty acid (FFA)-challenged HepG2 cells and high-fat diet-fed mice, respectively. Endogenous MEG3 was over-expressed by a specific pcDNA3.1-MEG3 to evaluate the regulatory function of MEG3 on triglyceride (TG)- and lipogenesis-related genes. Bioinformatic analysis was used to predict the target genes and binding sites, and the targeted regulatory relationship was verified with a dual luciferase assay. Finally, the possible pathway that regulates MEG3 was also evaluated. RESULTS We found that the downregulation of MEG3 in vitro and in vivo models of NAFLD was negatively correlated with lipogenesis-related genes and that overexpression of MEG3 reversed FFA-induced lipid accumulation in HepG2 cells. miR-21 was upregulated in the FFA-challenged HepG2 cells and was physically associated with MEG3 in the process of lipogenesis. Our mechanistic studies demonstrated that MEG3 competitively binds to miR-21 with LRP6, followed by the inhibition of the mTOR pathway, which induces intracellular lipid accumulation. CONCLUSION Our data are the first to document the working model of MEG3 functions as a potential hepatocyte lipid degeneration suppressor. MEG3 helps to alleviate lipid over-deposition, probably by binding to miR-21 to regulate the expression of LRP6. Our results suggest the potency of MEG3 as a biomarker for NAFLD and as a therapeutic target for treatment.
Collapse
Affiliation(s)
- Peng Huang
- Department of General Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, China; Department of General Surgery, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha 410008, China
| | - Fei-Zhou Huang
- Department of General Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, China
| | - Huai-Zheng Liu
- Emergency Department, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, China
| | - Tian-Yi Zhang
- Emergency Department, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, China
| | - Ming-Shi Yang
- Emergency Department, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, China
| | - Chuan-Zheng Sun
- Emergency Department, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, China.
| |
Collapse
|
13
|
Jayanthi R, Srinivasan AR. Biochemical isthmus [nexus] between type 2 diabetes mellitus and thyroid status-an update. Diabetes Metab Syndr 2019; 13:1173-1177. [PMID: 31336461 DOI: 10.1016/j.dsx.2019.01.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/18/2019] [Indexed: 11/29/2022]
Abstract
Both Type 1 [T1DM] and Type 2 diabetes mellitus [T2DM] share a nexus with altered thyroid status. In recent times, evidences point to the link between thyroid hormones andT2DM in particular. Several lines of evidences suggest an array of biochemical and molecular events. Gene polymorphism, disturbances in gene expression and regulation, enhanced and bizarre absorption of dietary glucose from intestine, decreased utilization of glucose by tissues and aberrations in hepatic handling of glucose with the onus on Gluconeogenesis are some of the projected mechanisms. Insulin resistance, a progressive condition is the hallmark in T2DM. Hypothyroidism as well as hyperthyroidism have been associated with insulin resistance which are synonymous with impaired glucose metabolism in T2DM. A multitude of basic, clinical and molecular studies provide an insight into thyroid comorbidity in T2DM, though there are a few instances to suggest equivocal link denoting cause-effect relationship. In biochemical pharmacology, as fortified by pharmacogenomics, modalities have now been proposed, through drug trials, to underline the utility of specifically designed thyroid hormone analogues in addressing metabolic syndrome, DM and associated cardiovascular pathology. A thorough understanding of the physiological, biochemical and molecular mechanisms would certainly open newer vistas in the perspectives of T2DM with special reference to alterations in thyroid status.
Collapse
Affiliation(s)
- Rajendran Jayanthi
- Department of Biochemistry, Mahatma Gandhi Medical College and Research Institute, Sri Balaji Vidyapeeth, Pillaiyarkuppam, Pondicherry, 607 402, India
| | - Abu Raghavan Srinivasan
- Department of Biochemistry, Mahatma Gandhi Medical College and Research Institute, Sri Balaji Vidyapeeth, Pillaiyarkuppam, Pondicherry, 607 402, India.
| |
Collapse
|
14
|
Matsuoka S, Bariuan JV, Nakagiri S, Abd Eldaim MA, Okamatsu-Ogura Y, Kimura K. Linking pathways and processes: Retinoic acid and glucose. MOLECULAR NUTRITION: CARBOHYDRATES 2019:247-264. [DOI: 10.1016/b978-0-12-849886-6.00013-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
15
|
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
|
16
|
Integrating Thyroid Hormone Signaling in Hypothalamic Control of Metabolism: Crosstalk Between Nuclear Receptors. Int J Mol Sci 2018; 19:ijms19072017. [PMID: 29997323 PMCID: PMC6073315 DOI: 10.3390/ijms19072017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/06/2018] [Accepted: 07/06/2018] [Indexed: 12/18/2022] Open
Abstract
The obesity epidemic is well recognized as a significant global health issue. A better understanding of the energy homeostasis mechanisms could help to identify promising anti-obesity therapeutic strategies. It is well established that the hypothalamus plays a pivotal role governing energy balance. The hypothalamus consists of tightly interconnected and specialized neurons that permit the sensing and integration of several peripheral inputs, including metabolic and hormonal signals for an appropriate physiological response. Current evidence shows that thyroid hormones (THs) constitute one of the key endocrine factors governing the regulation and the integration of metabolic homeostasis at the hypothalamic level. THs modulate numerous genes involved in the central control of metabolism, as TRH (Thyrotropin-Releasing Hormone) and MC4R (Melanocortin 4 Receptor). THs act through their interaction with thyroid hormone receptors (TRs). Interestingly, TH signaling, especially regarding metabolic regulations, involves TRs crosstalk with other metabolically linked nuclear receptors (NRs) including PPAR (Peroxisome proliferator-activated receptor) and LXR (Liver X receptor). In this review, we will summarize current knowledge on the important role of THs integration of metabolic pathways in the central regulation of metabolism. Particularly, we will shed light on the crosstalk between TRs and other NRs in controlling energy homeostasis. This could be an important track for the development of attractive therapeutic compounds.
Collapse
|
17
|
O-GlcNAc site-mapping of liver X receptor-α and O-GlcNAc transferase. Biochem Biophys Res Commun 2018; 499:354-360. [PMID: 29577901 DOI: 10.1016/j.bbrc.2018.03.164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 01/17/2023]
Abstract
The Liver X Receptor α (LXRα) belongs to the nuclear receptor superfamily and plays an essential role in regulating cholesterol, lipid and glucose metabolism and inflammatory responses. We have previously shown that LXRα is post-translationally modified by O-linked β-N-acetyl-glucosamine (O-GlcNAc) with increased transcriptional activity. Moreover, we showed that LXRα associates with O-GlcNAc transferase (OGT) in vitro and in vivo in mouse liver. In this study, we report that human LXRα is O-GlcNAc modified in its N-terminal domain (NTD) by identifying a specific O-GlcNAc site S49 and a novel O-GlcNAc modified peptide 20LWKPGAQDASSQAQGGSSCILRE42. However, O-GlcNAc site-mutations did not modulate LXRα transactivation of selected target gene promoters in vitro. Peptide array and co-immunoprecipitation assays demonstrate that LXRα interacts with OGT in its NTD and ligand-binding domain (LBD) in a ligand-independent fashion. Moreover, we map two new O-GlcNAc sites in the longest OGT isoform (ncOGT): S437 in the tetratricopeptide repeat (TPR) 13 domain and T1043 in the far C-terminus, and a new O-GlcNAc modified peptide (amino acids 826-832) in the intervening region (Int-D) within the catalytic domain. We also map four new O-GlcNAc sites in the short isoform sOGT: S391, T393, S399 and S437 in the TPRs 11-13 domain. Future studies will reveal the biological role of identified O-GlcNAc sites in LXRα and OGT.
Collapse
|
18
|
Sugar sensing by ChREBP/Mondo-Mlx-new insight into downstream regulatory networks and integration of nutrient-derived signals. Curr Opin Cell Biol 2017; 51:89-96. [PMID: 29278834 DOI: 10.1016/j.ceb.2017.12.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/17/2017] [Accepted: 12/13/2017] [Indexed: 12/13/2022]
Abstract
Animals regulate their physiology with respect to nutrient status, which requires nutrient sensing pathways. Simple carbohydrates, sugars, are sensed by the basic-helix-loop-helix leucine zipper transcription factors ChREBP/Mondo, together with their heterodimerization partner Mlx, which are well-established activators of sugar-induced lipogenesis. Loss of ChREBP/Mondo-Mlx in mouse and Drosophila leads to sugar intolerance, that is, inability to survive on sugar containing diet. Recent evidence has revealed that ChREBP/Mondo-Mlx responds to sugar and fatty acid-derived metabolites through several mechanisms and cross-connects with other nutrient sensing pathways. ChREBP/Mondo-Mlx controls several downstream transcription factors and hormones, which mediate not only readjustment of metabolic pathways, but also control feeding behavior, intestinal digestion, and circadian rhythm.
Collapse
|
19
|
Jois T, Sleeman MW. The regulation and role of carbohydrate response element-binding protein in metabolic homeostasis and disease. J Neuroendocrinol 2017; 29. [PMID: 28370553 DOI: 10.1111/jne.12473] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
Abstract
The transcription factor carbohydrate response element-binding protein (ChREBP) is a member of the basic helix-loop-helix leucine zipper transcription factor family. Under high-glucose conditions, it has a role in regulating the expression of key genes involved in various pathways, including glycolysis, gluconeogenesis and lipogenesis. It does this by forming a tetrameric complex made up of two ChREBP/Mlx heterodimers, which enables it to bind to the carbohydrate response element (ChoRE) in the promoter region of its target genes to regulate transcription. Because ChREBP plays a key role in glucose signalling and metabolism, and aberrations in glucose homeostasis are often present in metabolic diseases, this transcription factor presents itself as an enticing target with respect to further understanding metabolic disease mechanisms and potentially uncovering new therapeutic targets.
Collapse
Affiliation(s)
- T Jois
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - M W Sleeman
- Department of Physiology, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
20
|
Senese R, Cioffi F, de Lange P, Leanza C, Iannucci LF, Silvestri E, Moreno M, Lombardi A, Goglia F, Lanni A. Both 3,5-Diiodo-L-Thyronine and 3,5,3'-Triiodo-L-Thyronine Prevent Short-term Hepatic Lipid Accumulation via Distinct Mechanisms in Rats Being Fed a High-Fat Diet. Front Physiol 2017; 8:706. [PMID: 28959215 PMCID: PMC5603695 DOI: 10.3389/fphys.2017.00706] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/31/2017] [Indexed: 12/14/2022] Open
Abstract
3,3′,5-triiodo-L-thyronine (T3) improves hepatic lipid accumulation by increasing lipid catabolism but it also increases lipogenesis, which at first glance appears contradictory. Recent studies have shown that 3,5-diiodothyronine (T2), a natural thyroid hormone derivative, also has the capacity to stimulate hepatic lipid catabolism, however, little is known about its possible effects on lipogenic gene expression. Because genes classically involved in hepatic lipogenesis such as SPOT14, acetyl-CoA-carboxylase (ACC), and fatty acid synthase (FAS) contain thyroid hormone response elements (TREs), we studied their transcriptional regulation, focusing on TRE-mediated effects of T3 compared to T2 in rats receiving high-fat diet (HFD) for 1 week. HFD rats showed a marked lipid accumulation in the liver, which was significantly reduced upon simultaneous administration of either T3 or T2 with the diet. When administered to HFD rats, T2, in contrast with T3, markedly downregulated the expression of the above-mentioned genes. T2 downregulated expression of the transcription factors carbohydrate-response element-binding protein (ChREBP) and sterol regulatory element binding protein-1c (SREBP-1c) involved in activation of transcription of these genes, which explains the suppressed expression of their target genes involved in lipogenesis. T3, however, did not repress expression of the TRE-containing ChREBP gene but repressed SREBP-1c expression. Despite suppression of SREBP-1c expression by T3 (which can be explained by the presence of nTRE in its promoter), the target genes were not suppressed, but normalized to HFD reference levels or even upregulated (ACC), partly due to the presence of TREs on the promoters of these genes and partly to the lack of suppression of ChREBP. Thus, T2 and T3 probably act by different molecular mechanisms to achieve inhibition of hepatic lipid accumulation.
Collapse
Affiliation(s)
- Rosalba Senese
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| | - Federica Cioffi
- Dipartimento di Scienze e Tecnologie, Università degli Studi del SannioBenevento, Italy
| | - Pieter de Lange
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| | - Cristina Leanza
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| | - Liliana F Iannucci
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| | - Elena Silvestri
- Dipartimento di Scienze e Tecnologie, Università degli Studi del SannioBenevento, Italy
| | - Maria Moreno
- Dipartimento di Scienze e Tecnologie, Università degli Studi del SannioBenevento, Italy
| | - Assunta Lombardi
- Dipartimento di Biologia, Università degli Studi di Napoli Federico IINaples, Italy
| | - Fernando Goglia
- Dipartimento di Scienze e Tecnologie, Università degli Studi del SannioBenevento, Italy
| | - Antonia Lanni
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| |
Collapse
|
21
|
Methylcytosine dioxygenase TET3 interacts with thyroid hormone nuclear receptors and stabilizes their association to chromatin. Proc Natl Acad Sci U S A 2017; 114:8229-8234. [PMID: 28716910 DOI: 10.1073/pnas.1702192114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Thyroid hormone receptors (TRs) are members of the nuclear hormone receptor superfamily that act as ligand-dependent transcription factors. Here we identified the ten-eleven translocation protein 3 (TET3) as a TR interacting protein increasing cell sensitivity to T3. The interaction between TET3 and TRs is independent of TET3 catalytic activity and specifically allows the stabilization of TRs on chromatin. We provide evidence that TET3 is required for TR stability, efficient binding of target genes, and transcriptional activation. Interestingly, the differential ability of different TRα1 mutants to interact with TET3 might explain their differential dominant activity in patients carrying TR germline mutations. So this study evidences a mode of action for TET3 as a nonclassical coregulator of TRs, modulating its stability and access to chromatin, rather than its intrinsic transcriptional activity. This regulatory function might be more general toward nuclear receptors. Indeed, TET3 interacts with different members of the superfamily and also enhances their association to chromatin.
Collapse
|
22
|
Damiano F, Rochira A, Gnoni A, Siculella L. Action of Thyroid Hormones, T3 and T2, on Hepatic Fatty Acids: Differences in Metabolic Effects and Molecular Mechanisms. Int J Mol Sci 2017; 18:ijms18040744. [PMID: 28362337 PMCID: PMC5412329 DOI: 10.3390/ijms18040744] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 12/28/2022] Open
Abstract
The thyroid hormones (THs) 3,3′,5,5′-tetraiodo-l-thyronine (T4) and 3,5,3′-triiodo-l-thyronine (T3) influence many metabolic pathways. The major physiological function of THs is to sustain basal energy expenditure, by acting primarily on carbohydrate and lipid catabolism. Beyond the mobilization and degradation of lipids, at the hepatic level THs stimulate the de novo fatty acid synthesis (de novo lipogenesis, DNL), through both the modulation of gene expression and the rapid activation of cell signalling pathways. 3,5-Diiodo-l-thyronine (T2), previously considered only a T3 catabolite, has been shown to mimic some of T3 effects on lipid catabolism. However, T2 action is more rapid than that of T3, and seems to be independent of protein synthesis. An inhibitory effect on DNL has been documented for T2. Here, we give an overview of the mechanisms of THs action on liver fatty acid metabolism, focusing on the different effects exerted by T2 and T3 on the regulation of the DNL. The inhibitory action on DNL exerted by T2 makes this compound a potential and attractive drug for the treatment of some metabolic diseases and cancer.
Collapse
Affiliation(s)
- Fabrizio Damiano
- Laboratory of Biochemistry and Molecular Biology, Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| | - Alessio Rochira
- Laboratory of Biochemistry and Molecular Biology, Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| | - Antonio Gnoni
- Department of Basic Medical Sciences, Section of Medical Biochemistry, University of Bari Aldo Moro, 70125 Bari, Italy.
| | - Luisa Siculella
- Laboratory of Biochemistry and Molecular Biology, Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| |
Collapse
|
23
|
Regulation of hepatic lipogenesis by the zinc finger protein Zbtb20. Nat Commun 2017; 8:14824. [PMID: 28327662 PMCID: PMC5364431 DOI: 10.1038/ncomms14824] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/03/2017] [Indexed: 12/13/2022] Open
Abstract
Hepatic de novo lipogenesis (DNL) converts carbohydrates into triglycerides and is known to influence systemic lipid homoeostasis. Here, we demonstrate that the zinc finger protein Zbtb20 is required for DNL. Mice lacking Zbtb20 in the liver exhibit hypolipidemia and reduced levels of liver triglycerides, along with impaired hepatic lipogenesis. The expression of genes involved in glycolysis and DNL, including that of two ChREBP isoforms, is decreased in livers of knockout mice. Zbtb20 binds to and enhances the activity of the ChREBP-α promoter, suggesting that altered metabolic gene expression is mainly driven by ChREBP. In addition, ChREBP-β overexpression largely restores hepatic expression of genes involved in glucose and lipid metabolism, and increases plasma and liver triglyceride levels in knockout mice. Finally, we show that Zbtb20 ablation protects from diet-induced liver steatosis and improves hepatic insulin resistance. We suggest ZBTB20 is an essential regulator of hepatic lipogenesis and may be a therapeutic target for the treatment of fatty liver disease. De novo lipogenesis is tightly controlled by hormonal and nutritional signals and plays an important role in energy homoeostasis. Here, Liu et al. show that zinc finger protein ZBTB20 regulates the expression of key glycolytic and lipogenic genes by modulating ChREBP expression and transcriptional activity.
Collapse
|
24
|
The transcription factor carbohydrate-response element-binding protein (ChREBP): A possible link between metabolic disease and cancer. Biochim Biophys Acta Mol Basis Dis 2016; 1863:474-485. [PMID: 27919710 DOI: 10.1016/j.bbadis.2016.11.029] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/24/2016] [Accepted: 11/29/2016] [Indexed: 12/19/2022]
Abstract
Carbohydrate-response element-binding protein (ChREBP) has been identified as a transcription factor that binds to carbohydrate response element in the promoter of pyruvate kinase, liver and red blood cells. ChREBP is activated by metabolites derived from glucose and suppressed by adenosine monophosphate (AMP), ketone bodies and cyclic cAMP. ChREBP regulates gene transcription related to glucose and lipid metabolism. Findings from knockout mice and human subjects suggest that ChREBP helps to induce hepatic steatosis, dyslipidemia, and glucose intolerance. Moreover, in tumor cells, ChREBP promotes aerobic glycolysis through p53 inhibition, resulting in tumor cell proliferation. Anti-diabetic and anti-lipidemic drugs such as atorvastatin, metformin, bile acid sequestrants, docosahexaenoic acid and eicosapentaenoic acid may affect ChREBP transactivity. Secretory proteins such as fibroblast growth factor 21 and ANGPTL8 (Betatrophin) may be promising candidates for biologic markers reflecting ChREBP transactivity. Thus, ChREBP is associated with metabolic diseases and cancers, and may be a link between them.
Collapse
|
25
|
Once and for all, LXRα and LXRβ are gatekeepers of the endocrine system. Mol Aspects Med 2016; 49:31-46. [DOI: 10.1016/j.mam.2016.04.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/08/2016] [Accepted: 04/10/2016] [Indexed: 01/08/2023]
|
26
|
Identification of HNF-4α as a key transcription factor to promote ChREBP expression in response to glucose. Sci Rep 2016; 6:23944. [PMID: 27029511 PMCID: PMC4814918 DOI: 10.1038/srep23944] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/16/2016] [Indexed: 01/08/2023] Open
Abstract
Transcription factor carbohydrate responsive element binding protein (ChREBP) promotes glycolysis and lipogenesis in metabolic tissues and cancer cells. ChREBP-α and ChREBP-β, two isoforms of ChREBP transcribed from different promoters, are both transcriptionally induced by glucose. However, the mechanism by which glucose increases ChREBP mRNA levels remains unclear. Here we report that hepatocyte nuclear factor 4 alpha (HNF-4α) is a key transcription factor for glucose-induced ChREBP-α and ChREBP-β expression. Ectopic HNF-4α expression increased ChREBP transcription while knockdown of HNF-4α greatly reduced ChREBP mRNA levels in liver cancer cells and mouse primary hepatocytes. HNF-4α not only directly bound to an E-box-containing region in intron 12 of the ChREBP gene, but also promoted ChREBP-β transcription by directly binding to two DR1 sites and one E-box-containing site of the ChREBP-β promoter. Moreover, HNF-4α interacted with ChREBP-α and synergistically promoted ChREBP-β transcription. Functionally, HNF-4α suppression reduced glucose-dependent ChREBP induction. Increased nuclear abundance of HNF-4α and its binding to cis-elements of ChREBP gene in response to glucose contributed to glucose-responsive ChREBP transcription. Taken together, our results not only revealed the novel mechanism by which HNF-4α promoted ChREBP transcription in response to glucose, but also demonstrated that ChREBP-α and HNF-4α synergistically increased ChREBP-β transcription.
Collapse
|
27
|
Update on the molecular biology of dyslipidemias. Clin Chim Acta 2016; 454:143-85. [DOI: 10.1016/j.cca.2015.10.033] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/24/2015] [Accepted: 10/30/2015] [Indexed: 12/20/2022]
|
28
|
Singh BK, Sinha RA, Zhou J, Tripathi M, Ohba K, Wang ME, Astapova I, Ghosh S, Hollenberg AN, Gauthier K, Yen PM. Hepatic FOXO1 Target Genes Are Co-regulated by Thyroid Hormone via RICTOR Protein Deacetylation and MTORC2-AKT Protein Inhibition. J Biol Chem 2016; 291:198-214. [PMID: 26453307 PMCID: PMC4697156 DOI: 10.1074/jbc.m115.668673] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/01/2015] [Indexed: 12/21/2022] Open
Abstract
MTORC2-AKT is a key regulator of carbohydrate metabolism and insulin signaling due to its effects on FOXO1 phosphorylation. Interestingly, both FOXO1 and thyroid hormone (TH) have similar effects on carbohydrate and energy metabolism as well as overlapping transcriptional regulation of many target genes. Currently, little is known about the regulation of MTORC2-AKT or FOXO1 by TH. Accordingly, we performed hepatic transcriptome profiling in mice after FOXO1 knockdown in the absence or presence of TH, and we compared these results with hepatic FOXO1 and THRB1 (TRβ1) ChIP-Seq data. We identified a subset of TH-stimulated FOXO1 target genes that required co-regulation by FOXO1 and TH. TH activation of FOXO1 was directly linked to an increase in SIRT1-MTORC2 interaction and RICTOR deacetylation. This, in turn, led to decreased AKT and FOXO1 phosphorylation. Moreover, TH increased FOXO1 nuclear localization, DNA binding, and target gene transcription by reducing AKT-dependent FOXO1 phosphorylation in a THRB1-dependent manner. These events were associated with TH-mediated oxidative phosphorylation and NAD(+) production and suggested that downstream metabolic effects by TH can post-translationally activate other transcription factors. Our results showed that RICTOR/MTORC2-AKT can integrate convergent hormonal and metabolic signals to provide coordinated and sensitive regulation of hepatic FOXO1-target gene expression.
Collapse
Affiliation(s)
- Brijesh K Singh
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program and
| | - Rohit A Sinha
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program and
| | - Jin Zhou
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program and
| | - Madhulika Tripathi
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program and the Stroke Trial Unit, National Neuroscience Institute Singapore, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Kenji Ohba
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program and
| | - Mu-En Wang
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program and the Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Inna Astapova
- the Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115, and
| | - Sujoy Ghosh
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program and Centre for Computational Biology, Duke-National University of Singapore Graduate Medical School, Singapore 169857, Singapore
| | - Anthony N Hollenberg
- the Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115, and
| | - Karine Gauthier
- the Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46, Allée d'Italie 69364, Lyon Cedex 07, France
| | - Paul M Yen
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program and
| |
Collapse
|
29
|
Bindesbøll C, Fan Q, Nørgaard RC, MacPherson L, Ruan HB, Wu J, Pedersen TÅ, Steffensen KR, Yang X, Matthews J, Mandrup S, Nebb HI, Grønning-Wang LM. Liver X receptor regulates hepatic nuclear O-GlcNAc signaling and carbohydrate responsive element-binding protein activity. J Lipid Res 2015; 56:771-85. [PMID: 25724563 DOI: 10.1194/jlr.m049130] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Liver X receptor (LXR)α and LXRβ play key roles in hepatic de novo lipogenesis through their regulation of lipogenic genes, including sterol regulatory element-binding protein (SREBP)-1c and carbohydrate responsive element-binding protein (ChREBP). LXRs activate lipogenic gene transcription in response to feeding, which is believed to be mediated by insulin. We have previously shown that LXRs are targets for glucose-hexosamine-derived O-linked β-N-acetylglucosamine (O-GlcNAc) modification enhancing their ability to regulate SREBP-1c promoter activity in vitro. To elucidate insulin-independent effects of feeding on LXR-mediated lipogenic gene expression in vivo, we subjected control and streptozotocin-treated LXRα/β(+/+) and LXRα/β(-/-) mice to a fasting-refeeding regime. We show that under hyperglycemic and hypoinsulinemic conditions, LXRs maintain their ability to upregulate the expression of glycolytic and lipogenic enzymes, including glucokinase (GK), SREBP-1c, ChREBPα, and the newly identified shorter isoform ChREBPβ. Furthermore, glucose-dependent increases in LXR/retinoid X receptor-regulated luciferase activity driven by the ChREBPα promoter was mediated, at least in part, by O-GlcNAc transferase (OGT) signaling in Huh7 cells. Moreover, we show that LXR and OGT interact and colocalize in the nucleus and that loss of LXRs profoundly reduced nuclear O-GlcNAc signaling and ChREBPα promoter binding activity in vivo. In summary, our study provides evidence that LXRs act as nutrient and glucose metabolic sensors upstream of ChREBP by modulating GK expression, nuclear O-GlcNAc signaling, and ChREBP expression and activity.
Collapse
Affiliation(s)
- Christian Bindesbøll
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, N-0316 Oslo, Norway
| | - Qiong Fan
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, N-0316 Oslo, Norway
| | - Rikke C Nørgaard
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, N-0316 Oslo, Norway
| | - Laura MacPherson
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Hai-Bin Ruan
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT 06519 Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06519
| | - Jing Wu
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT 06519 Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06519
| | - Thomas Å Pedersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Knut R Steffensen
- Division of Clinical Chemistry Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, C174, SE-141 86 Stockholm, Sweden
| | - Xiaoyong Yang
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT 06519 Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06519 Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06519
| | - Jason Matthews
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, N-0316 Oslo, Norway Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Hilde I Nebb
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, N-0316 Oslo, Norway
| | - Line M Grønning-Wang
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, N-0316 Oslo, Norway
| |
Collapse
|
30
|
Liu KL, Lo M, Canaple L, Gauthier K, del Carmine P, Beylot M. Vascular Function of the Mesenteric Artery Isolated from Thyroid Hormone Receptor-E Knockout Mice. J Vasc Res 2014; 51:350-9. [DOI: 10.1159/000368195] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/14/2014] [Indexed: 11/19/2022] Open
|
31
|
Abstract
NCoR1 (nuclear receptor corepressor) and SMRT (silencing mediator of retinoid and thyroid hormone receptors; NCoR2) are well-recognized coregulators of nuclear receptor (NR) action. However, their unique roles in the regulation of thyroid hormone (TH) signaling in specific cell types have not been determined. To accomplish this we generated mice that lacked function of either NCoR1, SMRT, or both in the liver only and additionally a global SMRT knockout model. Despite both corepressors being present in the liver, deletion of SMRT in either euthyroid or hypothyroid animals had little effect on TH signaling. In contrast, disruption of NCoR1 action confirmed that NCoR1 is the principal mediator of TH sensitivity in vivo. Similarly, global disruption of SMRT, unlike the global disruption of NCoR1, did not affect TH levels. While SMRT played little role in TH-regulated pathways, when disrupted in combination with NCoR1, it greatly accentuated the synthesis and storage of hepatic lipid. Taken together, these data demonstrate that corepressor specificity exists in vivo and that NCoR1 is the principal regulator of TH action. However, both corepressors collaborate to control hepatic lipid content, which likely reflects their cooperative activity in regulating the action of multiple NRs including the TH receptor (TR).
Collapse
|
32
|
Billon C, Canaple L, Fleury S, Deloire A, Beylot M, Dombrowicz D, Del Carmine P, Samarut J, Gauthier K. TRα protects against atherosclerosis in male mice: identification of a novel anti-inflammatory property for TRα in mice. Endocrinology 2014; 155:2735-45. [PMID: 24797634 DOI: 10.1210/en.2014-1098] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hypothyroidism is associated with an increased occurrence of atherosclerosis, suggesting some protective role for thyroid hormones (THs). Hypercholesterolemia is one of the major risk factor to develop this disease. Here, we show that the well-known TH cholesterol lowering effect was dependent on TH nuclear receptor (TR)β liver activity. But most importantly, TRα was also shown to contribute of slowing down atherosclerosis progression via an independent mechanism. Introduction of TRα(0/0) deletion in the ApoE(-/-) background accelerated the appearance of plaques. Earlier cholesterol accumulation was detected in aorta macrophages, likely due to impaired cholesterol efflux. The IL-1β inflammatory cytokine was elevated in serum and macrophages in correlation with an activation of the AKT/nuclear factor κB pathway in these cells. Inhibition of AKT prevented inflammation and restored normal cholesterol efflux. Similar low-grade inflammation was identified in TRα(0/0) male mice. Thus, the mere absence of TRα is associated with elevated levels of cytokines likely responsible for cholesterol accumulation and atherosclerosis. This TRα protective activity should be relevant for other inflammatory pathologies.
Collapse
Affiliation(s)
- Cyrielle Billon
- Institut de Génomique Fonctionnelle de Lyon (C.B., L.C., A.D., J.S., K.G.), Université de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique. Institut National de la Recherche Agronomique, Ecole Normale Supérieure de Lyon, 69364 Lyon, France; Inserm Unité 1011 (S.F., D.D.), University of Lille Nord de France and Institut Pasteur de Lille, 59000 Lille, France; and Inserm Equipe Région-Inserm 22/Equipe Associée 4173 (M.B.) and Anira-ANIPHY (P.d.C.), Faculté Rockefeller, Université Lyon 1, 69008 Lyon, France
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Karagianni P, Talianidis I. Transcription factor networks regulating hepatic fatty acid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:2-8. [PMID: 24814048 DOI: 10.1016/j.bbalip.2014.05.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 02/06/2023]
Abstract
Tight regulation of lipid levels is critical for cellular and organismal homeostasis, not only in terms of energy utilization and storage, but also to prevent potential toxicity. The liver utilizes a set of hepatic transcription factors to regulate the expression of genes implicated in all aspects of lipid metabolism including catabolism, transport, and synthesis. In this article, we will review the main transcriptional mechanisms regulating the expression of genes involved in hepatic lipid metabolism. The principal regulatory pathways are composed of simple modules of transcription factor crosstalks, which correspond to building blocks of more complex regulatory networks. These transcriptional networks contribute to the regulation of proper lipid homeostasis in parallel to posttranslational mechanisms and end product-mediated modulation of lipid metabolizing enzymes. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.
Collapse
Affiliation(s)
| | - Iannis Talianidis
- Biomedical Sciences Research Center Alexander Fleming, 16672 Vari, Greece.
| |
Collapse
|
34
|
Oosterveer MH, Schoonjans K. Hepatic glucose sensing and integrative pathways in the liver. Cell Mol Life Sci 2014; 71:1453-67. [PMID: 24196749 PMCID: PMC11114046 DOI: 10.1007/s00018-013-1505-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/17/2013] [Accepted: 10/18/2013] [Indexed: 12/21/2022]
Abstract
The hepatic glucose-sensing system is a functional network of enzymes and transcription factors that is critical for the maintenance of energy homeostasis and systemic glycemia. Here we review the recent literature on its components and metabolic actions. Glucokinase (GCK) is generally considered as the initial postprandial glucose-sensing component, which acts as the gatekeeper for hepatic glucose metabolism and provides metabolites that activate the transcription factor carbohydrate response element binding protein (ChREBP). Recently, liver receptor homolog 1 (LRH-1) has emerged as an upstream regulator of the central GCK-ChREBP axis, with a critical role in the integration of hepatic intermediary metabolism in response to glucose. Evidence is also accumulating that O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) and acetylation can act as glucose-sensitive modifications that may contribute to hepatic glucose sensing by targeting regulatory proteins and the epigenome. Further elucidation of the components and functional roles of the hepatic glucose-sensing system may contribute to the future treatment of liver diseases associated with deregulated glucose sensors.
Collapse
Affiliation(s)
- Maaike H. Oosterveer
- Department of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Kristina Schoonjans
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
35
|
Zhao RL, Sun B, Liu Y, Li JH, Xiong WL, Liang DC, Guo G, Zuo AJ, Zhang JY. Cloning and identification of a novel thyroid hormone receptor β isoform expressed in the pituitary gland. Mol Cell Biochem 2014; 389:141-50. [PMID: 24481752 DOI: 10.1007/s11010-013-1935-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 12/18/2013] [Indexed: 12/11/2022]
Abstract
We have previously identified a novel Trβ isoform (TrβΔ) in the rat, in which a novel exon N (108 bps) was found between exon 3 and exon 4 of TrβΔ, which represents the only difference between TrβΔ and Trβ1. In this study, we searched for an elongated Trβ2-like subtype with one additional exon N. We successfully isolated the entire mRNA/cDNA of a novel elongated Trβ2 isoform via PCR in the rat pituitary gland. The mRNA/cDNA was only 108 bps (exon N) longer than that Trβ2, and the extension of the sequence was between exon 3 and 4 of Trβ. The whole sequence of this novel Trβ isoform has been published in NCBI GenBank (HM043807.1); it is named TRbeta2Delta (Trβ2Δ). In adult rat pituitary tissue, quantitative real-time RT-PCR analysis showed that the mRNA levels of Trβ2Δ and Trβ2 were roughly equal (P > 0.05). We cloned, expressed, and purified the His-Trβ2Δ protein [recombinant TRβ2Δ (rTRβ2Δ)]. SDS-PAGE and western blotting revealed that the molecular weight of rTRβ2Δ was 58.2 kDa. Using a radioligand binding assay and an electrophoretic mobility shift assay, rTRβ2Δ-bound T3 with high affinity and recognized thyroid hormone response element (TRE) binding sites. Finally, in vitro transfection experiments further confirmed that rTRβ2Δ binding T3 significantly promotes the transcription of target genes via the TRE. Here, we have provided evidence suggesting that rTRβ2Δ is a novel functional TR isoform.
Collapse
Affiliation(s)
- Rong-Lan Zhao
- Institute of Endocrinology, Metabolic Disease Hospital, Tianjin Medical University, Key Laboratory of Hormone and Development, National Health Ministry of China, Tianjin, 300070, People's Republic of China,
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Singh BK, Sinha RA, Zhou J, Xie SY, You SH, Gauthier K, Yen PM. FoxO1 deacetylation regulates thyroid hormone-induced transcription of key hepatic gluconeogenic genes. J Biol Chem 2013; 288:30365-30372. [PMID: 23995837 PMCID: PMC3798501 DOI: 10.1074/jbc.m113.504845] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 08/20/2013] [Indexed: 11/06/2022] Open
Abstract
Hepatic gluconeogenesis is a concerted process that integrates transcriptional regulation with hormonal signals. A major regulator is thyroid hormone (TH), which acts through its nuclear receptor (TR) to induce the expression of the hepatic gluconeogenic genes, phosphoenolpyruvate carboxykinase (PCK1) and glucose-6-phosphatase (G6PC). Forkhead transcription factor FoxO1 also is an important regulator of these genes; however, its functional interactions with TR are not known. Here, we report that TR-mediated transcriptional activation of PCK1 and G6PC in human hepatic cells and mouse liver was FoxO1-dependent and furthermore required FoxO1 deacetylation by the NAD(+)-dependent deacetylase, SirT1. siRNA knockdown of FoxO1 decreased, whereas overexpression of FoxO1 increased, TH-dependent transcriptional activation of PCK1 and G6PC in cultured hepatic cells. FoxO1 siRNA knockdown also decreased TH-mediated transcription in vivo. Additionally, TH was unable to induce FoxO1 deacetylation or hepatic PCK1 gene expression in TH receptor β-null (TRβ(-/-)) mice. Moreover, TH stimulated FoxO1 recruitment to the PCK1 and G6PC gene promoters in a SirT1-dependent manner. In summary, our results show that TH-dependent deacetylation of a second metabolically regulated transcription factor represents a novel mechanism for transcriptional integration of nuclear hormone action with cellular energy status.
Collapse
Affiliation(s)
- Brijesh Kumar Singh
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore 169857
| | - Rohit Anthony Sinha
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore 169857
| | - Jin Zhou
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore 169857
| | - Sherwin Ying Xie
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore 169857
| | - Seo-Hee You
- the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, the Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, and
| | - Karine Gauthier
- the Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46, allée d'Italie 69364 Lyon Cedex 07, France
| | - Paul Michael Yen
- From the Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore 169857,.
| |
Collapse
|
37
|
Ikeda Y, Tsuchiya H, Hama S, Kajimoto K, Kogure K. Resistin affects lipid metabolism during adipocyte maturation of 3T3-L1 cells. FEBS J 2013; 280:5884-95. [PMID: 24034627 DOI: 10.1111/febs.12514] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 08/27/2013] [Accepted: 09/02/2013] [Indexed: 01/01/2023]
Abstract
Resistin, an adipose-tissue-specific secretory factor, aggravates metabolic syndrome through impairment of glucose metabolism. Previously, we demonstrated that resistin expression was induced in both 3T3-L1 cells and primary pre-adipocytes derived from Zucker obese rats during the process of differentiation and maturation (Ikeda Y, Hama S, Kajimoto K, Okuno T, Tsuchiya H & Kogure K (2011) Biol Pharm Bull 34, 865-870). However, the biological function of resistin in adipocytes is poorly understood. In the present study, we examined the effects of resistin knockdown on the biological features of 3T3-L1 cells. We found that lipid content was significantly decreased in 3T3-L1 cells transfected with anti-resistin small interfering RNA (siRNA) after adipocyte differentiation. While expression of peroxisome proliferator activated receptor γ and CCAAT/enhancer-binding protein α was not affected, protein expression and transcriptional activity levels of carbohydrate response element binding protein (ChREBP), which upregulates transcription of lipogenic genes, decreased after anti-resistin siRNA treatment. Moreover, gene expression of fatty acid synthase and acetyl-CoA carboxylase 2, which are known to be regulated by ChREBP, were also suppressed by resistin knockdown. In contrast, activity of the fatty acid β-oxidation-regulating protein carnitine palmitoyltransferase 1 increased. These results suggest that resistin knockdown induces suppression of lipid production and activation of fatty acid β-oxidation. Consequently, resistin may affect lipid metabolism during adipocyte maturation.
Collapse
Affiliation(s)
- Yoshito Ikeda
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Japan
| | | | | | | | | |
Collapse
|
38
|
The liver X receptor: A master regulator of the gut–liver axis and a target for non alcoholic fatty liver disease. Biochem Pharmacol 2013; 86:96-105. [DOI: 10.1016/j.bcp.2013.03.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/21/2013] [Accepted: 03/21/2013] [Indexed: 12/15/2022]
|
39
|
Gathercole LL, Morgan SA, Tomlinson JW. Hormonal Regulation of Lipogenesis. VITAMINS & HORMONES 2013; 91:1-27. [DOI: 10.1016/b978-0-12-407766-9.00001-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
40
|
Wang C. The Relationship between Type 2 Diabetes Mellitus and Related Thyroid Diseases. J Diabetes Res 2013; 2013:390534. [PMID: 23671867 PMCID: PMC3647563 DOI: 10.1155/2013/390534] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 03/15/2013] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) has an intersecting underlying pathology with thyroid dysfunction. The literature is punctuated with evidence indicating a contribution of abnormalities of thyroid hormones to type 2 DM. The most probable mechanism leading to T2DM in thyroid dysfunction could be attributed to perturbed genetic expression of a constellation of genes along with physiological aberrations leading to impaired glucose utilization and disposal in muscles, overproduction of hepatic glucose output, and enhanced absorption of splanchnic glucose. These factors contribute to insulin resistance. Insulin resistance is also associated with thyroid dysfunction. Hyper- and hypothyroidism have been associated with insulin resistance which has been reported to be the major cause of impaired glucose metabolism in T2DM. The state-of-art evidence suggests a pivotal role of insulin resistance in underlining the relation between T2DM and thyroid dysfunction. A plethora of preclinical, molecular, and clinical studies have evidenced an undeniable role of thyroid malfunctioning as a comorbid disorder of T2DM. It has been investigated that specifically designed thyroid hormone analogues can be looked upon as the potential therapeutic strategies to alleviate diabetes, obesity, and atherosclerosis. These molecules are in final stages of preclinical and clinical evaluation and may pave the way to unveil a distinct class of drugs to treat metabolic disorders.
Collapse
Affiliation(s)
- Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, Shanghai 201399, China
- *Chaoxun Wang:
| |
Collapse
|
41
|
Diaz-Moralli S, Ramos-Montoya A, Marin S, Fernandez-Alvarez A, Casado M, Cascante M. Target metabolomics revealed complementary roles of hexose- and pentose-phosphates in the regulation of carbohydrate-dependent gene expression. Am J Physiol Endocrinol Metab 2012; 303:E234-42. [PMID: 22569070 DOI: 10.1152/ajpendo.00675.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Carbohydrate response element-binding protein (ChREBP) is a transcription factor that mediates glucose signaling in mammalian liver, leading to the expression of different glycolytic and lipogenic genes, such as pyruvate kinase (L-PK) and fatty acid synthase (FAS). The current model for ChREBP activation in response to sugar phosphates holds that glucose metabolization to xylulose 5-phosphate (X-5-P) triggers the activation of protein phosphatase 2A, which dephosphorylates ChREBP and leads to its nuclear translocation and activation. However, evidence indicates that glucose 6-phosphate (G-6-P) is the most likely signal metabolite for the glucose-induced transcription of these genes. The glucose derivative that is responsible for carbohydrate-dependent gene expression remains to be identified. The difficulties in measuring G-6-P and X-5-P concentrations simultaneously and in changing them independently have hindered such identification. To discriminate between these possibilities, we adapted a liquid chromatography mass spectrometry method to identify and quantify sugar phosphates in human hepatocarcinoma cells (Hep G2) and rat hepatocytes in response to different carbon sources and in the presence/absence of a glucose-6-phosphate dehydrogenase inhibitor. We also used this method to demonstrate that these cells could not metabolize 2-deoxyglucose beyond 2-deoxyglucose-6-phosphate. The simultaneous quantification of sugar phosphates and FAS and L-PK expression levels demonstrated that both X-5-P and G-6-P play a role in the modulation of gene expression. In conclusion, this report presents for the first time a single mechanism that incorporates the effects of X-5-P and G-6-P on the enhancement of the expression of carbohydrate-responsive genes.
Collapse
Affiliation(s)
- Santiago Diaz-Moralli
- Faculty of Biology, Department of Biochemistry and Molecular Biology, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
42
|
Oosterveer MH, Mataki C, Yamamoto H, Harach T, Moullan N, van Dijk TH, Ayuso E, Bosch F, Postic C, Groen AK, Auwerx J, Schoonjans K. LRH-1-dependent glucose sensing determines intermediary metabolism in liver. J Clin Invest 2012; 122:2817-26. [PMID: 22772466 DOI: 10.1172/jci62368] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 05/30/2012] [Indexed: 12/19/2022] Open
Abstract
Liver receptor homolog 1 (LRH-1), an established regulator of cholesterol and bile acid homeostasis, has recently emerged as a potential drug target for liver disease. Although LRH-1 activation may protect the liver against diet-induced steatosis and insulin resistance, little is known about how LRH-1 controls hepatic glucose and fatty acid metabolism under physiological conditions. We therefore assessed the role of LRH-1 in hepatic intermediary metabolism. In mice with conditional deletion of Lrh1 in liver, analysis of hepatic glucose fluxes revealed reduced glucokinase (GCK) and glycogen synthase fluxes as compared with those of wild-type littermates. These changes were attributed to direct transcriptional regulation of Gck by LRH-1. Impaired glucokinase-mediated glucose phosphorylation in LRH-1-deficient livers was also associated with reduced glycogen synthesis, glycolysis, and de novo lipogenesis in response to acute and prolonged glucose exposure. Accordingly, hepatic carbohydrate response element-binding protein activity was reduced in these animals. Cumulatively, these data identify LRH-1 as a key regulatory component of the hepatic glucose-sensing system required for proper integration of postprandial glucose and lipid metabolism.
Collapse
Affiliation(s)
- Maaike H Oosterveer
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
Havula E, Hietakangas V. Glucose sensing by ChREBP/MondoA-Mlx transcription factors. Semin Cell Dev Biol 2012; 23:640-7. [PMID: 22406740 DOI: 10.1016/j.semcdb.2012.02.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 02/24/2012] [Indexed: 01/02/2023]
Abstract
The paralogous transcription factors ChREBP and MondoA, together with their common binding partner Mlx, have emerged as key mediators of intracellular glucose sensing. By regulating target genes involved in glycolysis and lipogenesis, they mediate metabolic adaptation to changing glucose levels. As disturbed glucose homeostasis plays a central role in human metabolic diseases and as cancer cells often display altered glucose metabolism, better understanding of cellular glucose sensing will likely uncover new therapeutic opportunities. Here we review the regulation, function and evolutionary conservation of the ChREBP/MondoA-Mlx glucose sensing system and discuss possible directions for future research.
Collapse
Affiliation(s)
- Essi Havula
- Institute of Biotechnology, University of Helsinki, Viikinkaari 1, 00014 Helsinki, Finland
| | | |
Collapse
|
45
|
Abstract
Thyroid hormones regulate cholesterol and lipoprotein metabolism, whereas thyroid disorders, including overt and subclinical hypothyroidism, considerably alter lipid profile and promote cardiovascular disease. Good evidence shows that high thyroid-stimulating hormone (TSH) is associated with a nonfavorable lipid profile, although TSH has no cutoff threshold for its association with lipids. Thyromimetics represent a new class of hypolipidemic drugs: their imminent application in patients with severe dyslipidemias, combined or not with statins, will improve the lipid profile, potentially accelerate energy expenditure and, as a consequence, vitally lessen the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Leonidas H Duntas
- Endocrine Unit, Evgenidion Hospital, University of Athens, 20 Papadiamantopoulou Street, 11528 Athens, Greece.
| | | |
Collapse
|
46
|
Strollo F, Carucci I, Morè M, Marico G, Strollo G, Masini MA, Gentile S. Free Triiodothyronine and Cholesterol Levels in Euthyroid Elderly T2DM Patients. Int J Endocrinol 2012; 2012:420370. [PMID: 22973308 PMCID: PMC3438739 DOI: 10.1155/2012/420370] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 07/05/2012] [Accepted: 07/23/2012] [Indexed: 12/16/2022] Open
Abstract
Thyroid function regulates lipid metabolism. Despite the fact that T2DM is more prevalent in the elderly, often associates with thyroid dysfunction and increases cardiovascular risk both per se and via high TC and LDL-C levels, the association of the latter with FT(3) and FT(4) levels has not yet been fully investigated in T2DM. While trying to fill this gap in 296 elderly outpatients with T2DM, we found that TC and LDL-C correlated negatively with FT(4) and positively with FT(3). When divided according to treatment by oral hypoglycaemic agents (OHA) and insulin (IT), they reacted differently with respect to investigated associations: in the OHA's TC and LDL-C correlated negatively with FT(4) and showed no association with FT(3), whereas, in the IT's TC and LDL-C correlated positively with FT(3) and negatively with FT(4). When controlled for possible confounding factors, these associations did not change in the IT's but were missing in the OHA's. Recent literature reports upon complex hypothalamic and peripheral interactions between T2DM and thyroid, and suggests T(3) to enhance cholesterol synthesis and to have a role in insulin resistance states. Further investigations are needed to understand the intimate mechanisms of lipid metabolism in T2DM with respect to thyroid function.
Collapse
Affiliation(s)
- F. Strollo
- Endocrine Unit, INRCA, Via Cassia 1167, 00189 Rome, Italy
| | - I. Carucci
- Endocrine Unit, INRCA, Via Cassia 1167, 00189 Rome, Italy
| | - M. Morè
- Endocrine Unit, INRCA, Via Cassia 1167, 00189 Rome, Italy
| | - G. Marico
- Department of Clinical and Experimental Medicine, Second University of Naples, Via Pansini 5, 80131 Naples, Italy
| | - G. Strollo
- Endocrinology and Diabetes Service, FBF St. Peter's Hospital, Via Cassia 600, 001879 Rome, Italy
| | - M. A. Masini
- DipTeRis, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy
| | - S. Gentile
- Department of Clinical and Experimental Medicine, Second University of Naples, Via Pansini 5, 80131 Naples, Italy
- *S. Gentile:
| |
Collapse
|
47
|
Brenta G. Why can insulin resistance be a natural consequence of thyroid dysfunction? J Thyroid Res 2011; 2011:152850. [PMID: 21941681 PMCID: PMC3175696 DOI: 10.4061/2011/152850] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/05/2011] [Indexed: 01/06/2023] Open
Abstract
Evidence for a relationship between T4 and T3 and glucose metabolism appeared over 100 years ago when the influence of thyroid hormone excess in the deterioration of glucose metabolism was first noticed. Since then, it has been known that hyperthyroidism is associated with insulin resistance. More recently, hypothyroidism has also been linked to decreased insulin sensitivity. The explanation to this apparent paradox may lie in the differential effects of thyroid hormones at the liver and peripheral tissues level.
The purpose of this paper is to explore the effects of thyroid hormones in glucose metabolism and analyze the mechanisms whereby alterations of thyroid hormones lead to insulin resistance.
Collapse
Affiliation(s)
- Gabriela Brenta
- Department of Endocrinology, Dr. César Milstein Hospital, La Rioja 951, C1221ACI, Buenos Aires, Argentina
| |
Collapse
|
48
|
Guo S, Zheng F, Qiu X, Yang N. ChREBP gene polymorphisms are associated with coronary artery disease in Han population of Hubei province. Clin Chim Acta 2011; 412:1854-60. [PMID: 21726544 DOI: 10.1016/j.cca.2011.06.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 05/18/2011] [Accepted: 06/19/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND ChREBP regulates lipogenesis and glucose utilization in the liver. Current reports suggest a contradictive association between rs3812316 of this gene and triglyceride level. We hypothesized the polymorphisms in ChREBP gene were associated with CAD in Chinese population. METHODS The ChREBP gene polymorphisms were analyzed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) methods in 200 controls and 310 CAD patients. Serum lipids and glucose concentrations were measured in all subjects. Haplotypes were constructed based on rs3812316, rs7798357 and rs1051921. All the data were analyzed using SPSS14.0, PLINK1.07 and SHEsis software. RESULTS The rare allele G of rs3812316 was significantly lower in the CAD group after adjusting for age, sex, BMI, SBP and DBP (OR(a)=0.589, 95%CI=0.361-0.961, P=0.034). No significant differences between cases and controls were found in genotype or allele distributions of rs7798357, rs17145750 and rs1051921. Haplotype CGC was significant higher in CAD group (P<0.01, OR=2.364, 95%CI=1.608-3.474), while haplotypes GGC, CGT, CCC were significant lower in CAD group (P<0.05). CONCLUSIONS The rs3812316 and the haplotypes in ChREBP gene appeared to be related to high susceptibility to CAD.
Collapse
Affiliation(s)
- Shuren Guo
- Center for Gene Diagnosis, Zhongnan Hospital, Wuhan University, Hubei, China
| | | | | | | |
Collapse
|
49
|
Song Y, Yao X, Ying H. Thyroid hormone action in metabolic regulation. Protein Cell 2011; 2:358-68. [PMID: 21614672 DOI: 10.1007/s13238-011-1046-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 04/24/2011] [Indexed: 12/19/2022] Open
Abstract
Thyroid hormone plays pivotal roles in growth, differentiation, development and metabolic homeostasis via thyroid hormone receptors (TRs) by controlling the expression of TR target genes. The transcriptional activity of TRs is modulated by multiple factors including various TR isoforms, diverse thyroid hormone response elements, different heterodimeric partners, coregulators, and the cellular location of TRs. In the present review, we summarize recent advance in understanding the molecular mechanisms of thyroid hormone action obtained from human subject research, thyroid hormone mimetics application, TR isoform-specific knock-in mouse models, and mitochondrion study with highlights in metabolic regulations. Finally, as future perspectives, we share our thoughts about current challenges and possible approaches to promote our knowledge of thyroid hormone action in metabolism.
Collapse
Affiliation(s)
- Yiyun Song
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | | | | |
Collapse
|
50
|
Poupeau A, Postic C. Cross-regulation of hepatic glucose metabolism via ChREBP and nuclear receptors. Biochim Biophys Acta Mol Basis Dis 2011; 1812:995-1006. [PMID: 21453770 DOI: 10.1016/j.bbadis.2011.03.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 03/21/2011] [Accepted: 03/22/2011] [Indexed: 01/17/2023]
Abstract
There is a worldwide epidemic of obesity and type 2 diabetes, two major public health concerns associated with alterations in both insulin and glucose signaling pathways. Glucose is not only an energy source but also controls the expression of key genes involved in energetic metabolism, through the glucose-signaling transcription factor, Carbohydrate Responsive Element Binding Protein (ChREBP). ChREBP has emerged as a central regulator of de novo fatty acid synthesis (lipogenesis) in response to glucose under both physiological and physiopathological conditions. Glucose activates ChREBP by regulating its entry from the cytosol to the nucleus, thereby promoting its binding to carbohydrate responsive element (ChoRE) in the promoter regions of glycolytic (L-PK) and lipogenic genes (ACC and FAS). We have previously reported that the inhibition of ChREBP in liver of obese ob/ob mice improves the metabolic alterations linked to obesity, fatty liver and insulin-resistance. Therefore, regulating ChREBP activity could be an attractive target for lipid-lowering therapies in obesity and diabetes. However, before this is possible, a better understanding of the mechanism(s) regulating its activity is needed. In this review, we summarize recent findings on the role and regulation of ChREBP and particularly emphasize on the cross-regulations that may exist between key nuclear receptors (LXR, TR, HNF4α) and ChREBP for the control of hepatic glucose metabolism. These novel molecular cross-talks may open the way to new pharmacological opportunities. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.
Collapse
|