1
|
Ritter MJ, Amano I, Hollenberg AN. Transcriptional Cofactors for Thyroid Hormone Receptors. Endocrinology 2025; 166:bqae164. [PMID: 39679543 PMCID: PMC11702866 DOI: 10.1210/endocr/bqae164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
Thyroid hormone (TH) is essential throughout life. Its actions are mediated primarily by the thyroid hormone receptor (THR), which is a nuclear receptor. Classically, the THRs act as inducible transcription factors. In the absence of TH, a corepressor complex is recruited to the THR to limit TH-related gene expression. In the presence of TH, the corepressor complex is dismissed and a coactivator complex is recruited to facilitate TH-related gene expression. These coregulators can interact with multiple nuclear receptors and are also key in maintaining normal physiologic function. The nuclear receptor corepressor 1 (NCOR1) and the nuclear receptor corepressor 2 (NCOR2) have been the most extensively studied corepressors of the THR involved in histone deacetylation. The steroid receptor coactivator/p160 (SRC) family and in particular, SRC-1, plays a key role in histone acetylation associated with the THR. The Mediator Complex is also required for pretranscription machinery assembly. This mini-review focuses on how these transcriptional cofactors influence TH-action and signaling, primarily via histone modifications.
Collapse
Affiliation(s)
- Megan J Ritter
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Izuki Amano
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Anthony N Hollenberg
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
2
|
Abstract
Thyroid hormones (THs) are key hormones that regulate development and metabolism in mammals. In man, the major target tissues for TH action are the brain, liver, muscle, heart, and adipose tissue. Defects in TH synthesis, transport, metabolism, and nuclear action have been associated with genetic and endocrine diseases in man. Over the past few years, there has been renewed interest in TH action and the therapeutic potential of THs and thyromimetics to treat several metabolic disorders such as hypercholesterolemia, dyslipidaemia, non-alcoholic fatty liver disease (NAFLD), and TH transporter defects. Recent advances in the development of tissue and TH receptor isoform-targeted thyromimetics have kindled new hope for translating our fundamental understanding of TH action into an effective therapy. This review provides a concise overview of the historical development of our understanding of TH action, its physiological and pathophysiological effects on metabolism, and future therapeutic applications to treat metabolic dysfunction.
Collapse
Affiliation(s)
- Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore.
- Div. Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
3
|
Cho YW, Fu Y, Huang CCJ, Wu X, Ng L, Kelley KA, Vella KR, Berg AH, Hollenberg AN, Liu H, Forrest D. Thyroid hormone-regulated chromatin landscape and transcriptional sensitivity of the pituitary gland. Commun Biol 2023; 6:1253. [PMID: 38081939 PMCID: PMC10713718 DOI: 10.1038/s42003-023-05546-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Thyroid hormone (3,5,3'-triiodothyronine, T3) is a key regulator of pituitary gland function. The response to T3 is thought to hinge crucially on interactions of nuclear T3 receptors with enhancers but these sites in pituitary chromatin remain surprisingly obscure. Here, we investigate genome-wide receptor binding in mice using tagged endogenous thyroid hormone receptor β (TRβ) and analyze T3-regulated open chromatin using an anterior pituitary-specific Cre driver (Thrbb2Cre). Strikingly, T3 regulates histone modifications and chromatin opening primarily at sites that maintain TRβ binding regardless of T3 levels rather than at sites where T3 abolishes or induces de novo binding. These sites associate more frequently with T3-activated than T3-suppressed genes. TRβ-deficiency blunts T3-regulated gene expression, indicating that TRβ confers transcriptional sensitivity. We propose a model of gene activation in which poised receptor-enhancer complexes facilitate adjustable responses to T3 fluctuations, suggesting a genomic basis for T3-dependent pituitary function or pituitary dysfunction in thyroid disorders.
Collapse
Affiliation(s)
- Young-Wook Cho
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yulong Fu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chen-Che Jeff Huang
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuefeng Wu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lily Ng
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kevin A Kelley
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Kristen R Vella
- Division of Endocrinology, Diabetes and Metabolism, Weill Department of Medicine Weill Cornell Medicine, New York, New York, 10065, USA
| | - Anders H Berg
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, California, 90048, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Weill Department of Medicine Weill Cornell Medicine, New York, New York, 10065, USA
| | - Hong Liu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
4
|
Yang R, Cao J, Speakman JR, Zhao Z. Limits to sustained energy intake. XXXIII. Thyroid hormones play important roles in milk production but do not define the heat dissipation limit in Swiss mice. J Exp Biol 2023; 226:jeb245393. [PMID: 37767758 DOI: 10.1242/jeb.245393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 09/22/2023] [Indexed: 09/29/2023]
Abstract
The limits to sustained energy intake set physiological upper boundaries that affect many aspects of human and animal performance. The mechanisms underlying these limits, however, remain unclear. We exposed Swiss mice to either supplementary thyroid hormones (THs) or the inhibitor methimazole during lactation at 21 or 32.5°C, and measured food intake, resting metabolic rate (RMR), milk energy output (MEO), serum THs and mammary gland gene expression of females, and litter size and mass of their offspring. Lactating females developed hyperthyroidism following exposure to supplementary THs at 21°C, but they did not significantly change body temperature, asymptotic food intake, RMR or MEO, and litter and mass were unaffected. Hypothyroidism, induced by either methimazole or 32.5°C exposure, significantly decreased asymptotic food intake, RMR and MEO, resulting in significantly decreased litter size and litter mass. Furthermore, gene expression of key genes in the mammary gland was significantly decreased by either methimazole or heat exposure, including gene expression of THs and prolactin receptors, and Stat5a and Stat5b. This suggests that endogenous THs are necessary to maintain sustained energy intake and MEO. Suppression of the thyroid axis seems to be an essential aspect of the mechanism by which mice at 32.5°C reduce their lactation performance to avoid overheating. However, THs do not define the upper limit to sustained energy intake and MEO at peak lactation at 21°C. Another, as yet unknown, factor prevents supplementary thyroxine exerting any stimulatory metabolic impacts on lactating mice at 21°C.
Collapse
Affiliation(s)
- Rui Yang
- College of Life and Environmental Science, Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China
| | - Jing Cao
- College of Life and Environmental Science, Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China
| | - John R Speakman
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK
| | - Zhijun Zhao
- College of Life and Environmental Science, Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China
| |
Collapse
|
5
|
Abstract
Background: Thyroid hormone (TH) has actions in every tissue of the body and is essential for normal development, as well as having important actions in the adult. The earliest markers of TH action that were identified and monitored clinically, even before TH could be measured in serum, included oxygen consumption, basal metabolic rate, serum cholesterol, and deep tendon reflex time. Cellular, rodent, amphibian, zebrafish, and human models have been used to study TH action. Summary: Early studies of the mechanism of TH action focused on saturable-specific triiodothyronine (T3) nuclear binding and direct actions of T3 that altered protein expression. Additional effects of TH were recognized on mitochondria, stimulation of ion transport, especially the sodium potassium ATPase, augmentation of adrenergic signaling, role as a neurotransmitter, and direct plasma membrane effects. The cloning of the thyroid hormone receptor (THR) genes in 1986 and report of the THR crystal structure in 1995 produced rapid progress in understanding the mechanism of TH nuclear action, as well as the development of modified THR ligands. These findings revealed nuances of TH signaling, including the role of nuclear receptor coactivators and corepressors, repression of positively stimulated genes by the unliganded receptor, THR isoform-specific actions of TRα (THRA) and TRβ (THRB), and THR binding DNA as a heterodimer with retinoid-x-receptor (RXR) for genes positively regulated by TH. The identification of genetic disorders of TH transport and signaling, especially Resistance to Thyroid Hormone (RTH) and monocarboxylate transporter 8 (Mct8) defects, has been highly informative with respect to the mechanism of TH action. Conclusions: The impact of THR isoform, post-translational modifications, receptor cofactors, DNA response element, and selective TH tissue uptake, on TH action, have clinical implications for diagnosing and treating thyroid disease. Additionally, these findings have led to the development of novel TH and TH analogue therapies for metabolic, neurological, and cardiovascular diseases.
Collapse
Affiliation(s)
- Gregory A Brent
- Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
6
|
Abstract
The present review traces the road leading to discovery of L-thyroxine, thyroid hormone (3,5,3´-triiodo-L-thyronine, T3) and its cognate nuclear receptors. Thyroid hormone is a pleio-tropic regulator of growth, differentiation, and tissue homeostasis in higher organisms. The major site of the thyroid hormone action is predominantly a cell nucleus. T3 specific binding sites in the cell nuclei have opened a new era in the field of the thyroid hormone receptors (TRs) discovery. T3 actions are mediated by high affinity nuclear TRs, TRalpha and TRbeta, which function as T3-activated transcription factors playing an essential role as transcription-modulating proteins affecting the transcriptional responses in target genes. Discovery and characterization of nuclear retinoid X receptors (RXRs), which form with TRs a heterodimer RXR/TR, positioned RXRs at the epicenter of molecular endocrinology. Transcriptional control via nuclear RXR/TR heterodimer represents a direct action of thyroid hormone. T3 plays a crucial role in the development of brain, it exerts significant effects on the cardiovascular system, skeletal muscle contractile function, bone development and growth, both female and male reproductive systems, and skin. It plays an important role in maintaining the hepatic, kidney and intestine homeostasis and in pancreas, it stimulates the beta-cell proliferation and survival. The TRs cross-talk with other signaling pathways intensifies the T3 action at cellular level. The role of thyroid hormone in human cancers, acting via its cognate nuclear receptors, has not been fully elucidated yet. This review is aimed to describe the history of T3 receptors, starting from discovery of T3 binding sites in the cell nuclei to revelation of T3 receptors as T3-inducible transcription factors in relation to T3 action at cellular level. It also focuses on milestones of investigation, comprising RXR/TR dimerization, cross-talk between T3 receptors, and other regulatory pathways within the cell and mainly on genomic action of T3. This review also focuses on novel directions of investigation on relationships between T3 receptors and cancer. Based on the update of available literature and the author's experimental experience, it is devoted to clinicians and medical students.
Collapse
|
7
|
Galton VA, Larsen PR, Berry MJ. The Deiodinases: Their Identification and Cloning of Their Genes. Endocrinology 2021; 162:6101156. [PMID: 33449107 PMCID: PMC7864002 DOI: 10.1210/endocr/bqab005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Indexed: 01/03/2023]
Abstract
In this minireview, we provide a historical outline of the events that led to the identification and characterization of the deiodinases, the recognition that deiodination plays a major role in thyroid hormone action, and the cloning of the 3 deiodinase genes. The story starts in 1820, when it was first determined that elemental iodine was important for normal thyroid function. Almost 100 years later, it was found that the primary active principle of the gland, T4, contains iodine. Once radioactive iodine became available in the 1940s, it was demonstrated that the metabolism of T4 included deiodination, but at the time it was assumed to be merely a degradative process. However, this view was questioned after the discovery of T3 in 1952. We discuss in some detail the events of the next 20 years, which included some failures followed by the successful demonstration that deiodination is indeed essential to normal thyroid hormone action. Finally, we describe how the 3 deiodinases were identified and characterized and their genes cloned.
Collapse
Affiliation(s)
- Valerie Anne Galton
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Correspondence: Valerie Anne Galton, Geisel School of Medicine at Dartmouth, Department of Molecular and Systems Biology, Remsen Building, 66 College Street, Hanover, NH 03755, USA.
| | - P Reed Larsen
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Marla J Berry
- Pacific Biosciences Research Center, SOEST, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
8
|
Barreto-Chaves MLM, Senger N, Fevereiro MR, Parletta AC, Takano APC. Impact of hyperthyroidism on cardiac hypertrophy. Endocr Connect 2020; 9:R59-R69. [PMID: 32101527 PMCID: PMC7159257 DOI: 10.1530/ec-19-0543] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 02/25/2020] [Indexed: 12/14/2022]
Abstract
The cardiac growth process (hypertrophy) is a crucial phenomenon conserved across a wide array of species and is critically involved in the maintenance of cardiac homeostasis. This process enables an organism to adapt to changes in systemic demand and occurs due to a plethora of responses, depending on the type of signal or stimuli received. The growth of cardiac muscle cells in response to environmental conditions depends on the type, strength and duration of stimuli, and results in adaptive physiological responses or non-adaptive pathological responses. Thyroid hormones (TH) have a direct effect on the heart and induce a cardiac hypertrophy phenotype, which may evolve to heart failure. In this review, we summarize the literature on TH function in the heart by presenting results from experimental studies. We discuss the mechanistic aspects of TH associated with cardiac myocyte hypertrophy, increased cardiac myocyte contractility and electrical remodeling, as well as the associated signaling pathways. In addition to classical crosstalk with the sympathetic nervous system (SNS), emerging work pointing to the new endocrine interaction between TH and the renin-angiotensin system (RAS) is also explored. Given the inflammatory potential of the angiotensin II peptide, this new interaction may open the door for new therapeutic approaches which target the key mechanisms responsible for TH-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- M L M Barreto-Chaves
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - N Senger
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - M R Fevereiro
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - A C Parletta
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - A P C Takano
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Morris JC, Galton VA. The isolation of thyroxine (T4), the discovery of 3,5,3'-triiodothyronine (T3), and the identification of the deiodinases that generate T3 from T4: An historical review. Endocrine 2019; 66:3-9. [PMID: 31256344 DOI: 10.1007/s12020-019-01990-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/21/2019] [Indexed: 10/26/2022]
Affiliation(s)
- John C Morris
- Divisions of Endocrinology and Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Valerie Anne Galton
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| |
Collapse
|
10
|
Galton VA. The ups and downs of the thyroxine pro-hormone hypothesis. Mol Cell Endocrinol 2017; 458:105-111. [PMID: 28130114 DOI: 10.1016/j.mce.2017.01.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/20/2016] [Accepted: 01/19/2017] [Indexed: 12/25/2022]
Abstract
Thyroxine (T4) is the major thyroid hormone in the thyroid gland and the circulation. However, it is widely accepted on the basis of abundant evidence that 3,5,3'-triiodothyronine (T3) is responsible for most, if not all, of the physiological effects of TH in extrathyroidal tissues, and T4 functions as the pro-hormone. Whether T4 has any intrinsic activity per se or is merely a pro-hormone that must be converted to T3 in order to exert any TH action has yet to be resolved. Although there are some physiological actions of T4 that are mediated by receptors at the cell membrane (non-genomic effects), the vast majority of the physiological effects of the THs identified to date involve the binding of T3 to specific nuclear receptors to regulate gene expression (genomic effects). This review examines how the role of T4 in genomic TH action has been viewed and debated during the hundred years since it was first isolated in 1914.
Collapse
Affiliation(s)
- Valerie Anne Galton
- Department of Physiology and Neurobiology, The Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH 03756, USA.
| |
Collapse
|
11
|
Bargi-Souza P, Goulart-Silva F, Nunes MT. Novel aspects of T 3 actions on GH and TSH synthesis and secretion: physiological implications. J Mol Endocrinol 2017; 59:R167-R178. [PMID: 28951438 DOI: 10.1530/jme-17-0068] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 09/26/2017] [Indexed: 12/27/2022]
Abstract
Thyroid hormones (THs) classically regulate the gene expression by transcriptional mechanisms. In pituitary, the encoding genes for growth hormone (GH) and thyroid-stimulating hormone (TSH) are examples of genes regulated by triiodothyronine (T3) in a positive and negative way, respectively. Recent studies have shown a rapid adjustment of GH and TSH synthesis/secretion induced by T3 posttranscriptional actions. In somatotrophs, T3 promotes an increase in Gh mRNA content, poly(A) tail length and binding to the ribosome, associated with a rearrangement of actin cytoskeleton. In thyrotrophs, T3 reduces Tshb mRNA content, poly(A) tail length and its association with the ribosome. In parallel, it promotes a redistribution of TSH secretory granules to more distal regions of the cell periphery, indicating a rapid effect of T3 inhibition of TSH secretion. T3 was shown to affect the content of tubulin and the polymerization of actin and tubulin cytoskeletons in the whole anterior pituitary gland, and to increase intracellular alpha (CGA) content. This review summarizes genomic and non-genomic/posttranscriptional actions of TH on the regulation of several steps of GH and TSH synthesis and secretion. These distinct mechanisms induced by T3 can occur simultaneously, even though non-genomic effects are promptly elicited and precede the genomic actions, coexisting in a functional network within the cells.
Collapse
Affiliation(s)
| | | | - M T Nunes
- Department of Physiology and Biophysics of the Institute of Biomedical SciencesUniversity of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
12
|
Martinez B, Scheibner M, Soñanez-Organis JG, Jaques JT, Crocker DE, Ortiz RM. Increased sensitivity of thyroid hormone-mediated signaling despite prolonged fasting. Gen Comp Endocrinol 2017; 252:36-47. [PMID: 28743556 PMCID: PMC5580341 DOI: 10.1016/j.ygcen.2017.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 07/20/2017] [Accepted: 07/21/2017] [Indexed: 11/24/2022]
Abstract
Thyroid hormones (TH) can increase cellular metabolism. Food deprivation in mammals is typically associated with reduced thyroid gland responsiveness, in an effort to suppress cellular metabolism and abate starvation. However, in prolonged-fasted, elephant seal pups, cellular TH-mediated proteins are up-regulated and TH levels are maintained with fasting duration. The function and contribution of the thyroid gland to this apparent paradox is unknown and physiologically perplexing. Here we show that the thyroid gland remains responsive during prolonged food deprivation, and that its function and production of TH increase with fasting duration in elephant seals. We discovered that our modeled plasma TH data in response to exogenous thyroid stimulating hormone predicted cellular signaling, which was corroborated independently by the enzyme expression data. The data suggest that the regulation and function of the thyroid gland in the northern elephant seal is atypical for a fasted animal, and can be better described as, "adaptive fasting". Furthermore, the modeling data help substantiate the in vivo responses measured, providing unique insight on hormone clearance, production rates, and thyroid gland responsiveness. Because these unique endocrine responses occur simultaneously with a nearly strict reliance on the oxidation of lipid, these findings provide an intriguing model to better understand the TH-mediated reliance on lipid metabolism that is not otherwise present in morbidly obese humans. When coupled with cellular, tissue-specific responses, these data provide a more integrated assessment of thyroidal status that can be extrapolated for many fasting/food deprived mammals.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Molecular and Cellular Biology, University of California Merced, 5200 North Lake Road, Merced, CA 95343, USA.
| | - Michael Scheibner
- Department of Physics, University of California Merced, 5200 North Lake Road, Merced, CA 95343, USA
| | - José G Soñanez-Organis
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Lázaro Cárdenas del Río No. 100, Francisco Villa, Navojoa, Sonora 85880, Mexico
| | - John T Jaques
- Texas A&M Veterinary Diagnostic Laboratory, 1 Sippel Road, College Station, TX 77843, USA
| | - Daniel E Crocker
- Department of Biology, Sonoma State University, 1801 E. Cotati Avenue, Rohnert Park, CA 94928, USA
| | - Rudy M Ortiz
- Department of Molecular and Cellular Biology, University of California Merced, 5200 North Lake Road, Merced, CA 95343, USA
| |
Collapse
|
13
|
Kim N, Gu JY, Yoo HJ, Han SE, Kim YI, Nam-Goong IS, Kim ES, Kim HK. Contact system activation and high thrombin generation in hyperthyroidism. Eur J Endocrinol 2017; 176:583-589. [PMID: 28137735 DOI: 10.1530/eje-16-0835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/17/2017] [Accepted: 01/30/2017] [Indexed: 11/08/2022]
Abstract
BACKGROUND Hyperthyroidism is associated with increased thrombotic risk. As contact system activation through formation of neutrophil extracellular traps (NET) has emerged as an important trigger of thrombosis, we hypothesized that the contact system is activated along with active NET formation in hyperthyroidism and that their markers correlate with disease severity. SUBJECTS AND METHODS In 61 patients with hyperthyroidism and 40 normal controls, the levels of coagulation factors (fibrinogen, and factor VII, VIII, IX, XI and XII), D-dimer, thrombin generation assay (TGA) markers, NET formation markers (histone-DNA complex, double-stranded DNA and neutrophil elastase) and contact system markers (activated factor XII (XIIa), high-molecular-weight kininogen (HMWK), prekallikrein and bradykinin) were measured. RESULTS Patients with hyperthyroidism showed higher levels of fibrinogen (median (interquartile range), 315 (280-344) vs 262 (223-300), P = 0.001), D-dimer (103.8 (64.8-151.5) vs 50.7 (37.4-76.0), P < 0.001), peak thrombin (131.9 (102.2-159.4) vs 31.6 (14.8-83.7), P < 0.001) and endogenous thrombin potential (649 (538-736) vs 367 (197-1147), P = 0.021) in TGA with 1 pM tissue factor, neutrophil elastase (1.10 (0.39-2.18) vs 0.23 (0.20-0.35), P < 0.001), factor XIIa (66.9 (52.8-87.0) vs 73.0 (57.1-86.6), P < 0.001), HMWK (6.11 (4.95-7.98) vs 3.83 (2.60-5.68), P < 0.001), prekallikrein (2.15 (1.00-6.36) vs 1.41 (0.63-2.22), P = 0.026) and bradykinin (152.4 (137.6-180.4) vs 118.3 (97.1-137.9), P < 0.001) than did normal controls. In age- and sex-adjusted logistic regression analysis, fibrinogen, factor VIII, IX and XIIa, D-dimer, peak thrombin, neutrophil elastase, HMWK and bradykinin showed significant odds ratios representing hyperthyroidism's contribution to coagulation and contact system activation. Free T4 was significantly correlated with factors VIII and IX, D-dimer, double-stranded DNA and bradykinin. CONCLUSION This study demonstrated that contact system activation and abundant NET formation occurred in the high thrombin generation state in hyperthyroidism and were correlated with free T4 level.
Collapse
Affiliation(s)
- Namhee Kim
- Department of Laboratory Medicine and Cancer Research InstituteSeoul National University College of Medicine, Seoul, Korea
| | - Ja-Yoon Gu
- Department of Laboratory Medicine and Cancer Research InstituteSeoul National University College of Medicine, Seoul, Korea
| | - Hyun Ju Yoo
- Department of Laboratory Medicine and Cancer Research InstituteSeoul National University College of Medicine, Seoul, Korea
| | - Se Eun Han
- Department of Internal MedicineUlsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Young Il Kim
- Department of Internal MedicineUlsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Il Sung Nam-Goong
- Department of Internal MedicineUlsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Eun Sook Kim
- Department of Internal MedicineUlsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Hyun Kyung Kim
- Department of Laboratory Medicine and Cancer Research InstituteSeoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Martinez B, Soñanez-Organis JG, Godoy-Lugo JA, Horin LJ, Crocker DE, Ortiz RM. Thyroid hormone-stimulated increases in PGC-1α and UCP2 promote life history-specific endocrine changes and maintain a lipid-based metabolism. Am J Physiol Regul Integr Comp Physiol 2016; 312:R189-R196. [PMID: 27903512 DOI: 10.1152/ajpregu.00395.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/31/2016] [Accepted: 11/18/2016] [Indexed: 12/12/2022]
Abstract
Thyroid hormones (THs) regulate metabolism, but are typically suppressed during times of stressful physiological conditions, including fasting. Interestingly, prolonged fasting in northern elephant seal pups is associated with reliance on a lipid-based metabolism and increased levels of circulating THs that are partially attributed to active secretion as opposed to reduced clearance. This apparent paradox is coupled with complementary increases in cellular TH-mediated activity, suggesting that in mammals naturally adapted to prolonged fasting, THs are necessary to support metabolism. However, the functional relevance of this physiological paradox has remained largely unexplored, especially as it relates to the regulation of lipids. To address the hypothesis that TSH-mediated increase in THs contributes to lipid metabolism, we infused early and late-fasted pups with TSH and measured several key genes in adipose and muscle, and plasma hormones associated with regulation of lipid metabolism. TSH infusion increased the mRNA expressions of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) more than 6.5-fold at 60 min in muscle, and expression of uncoupling protein 2 (UCP2) more than 27-fold during the early fast at 60 min, in adipose. Additionally, during the late fast period, the protein content of adipose CD36 increased 1.1-fold, and plasma nonesterified fatty acid (NEFA) concentrations increased 25% at 120 min, with NEFA levels returning to baseline after 24 h. We show that the TSH-induced increases in THs in fasting pups are functional and likely contribute to the maintenance of a lipid-based metabolism.
Collapse
Affiliation(s)
- Bridget Martinez
- Molecular and Cellular Biology, University of California Merced, Merced, California;
| | - José G Soñanez-Organis
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Francisco Villa, Navojoa Sonora, México
| | - José Arquimides Godoy-Lugo
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Francisco Villa, Navojoa Sonora, México
| | - Lillian J Horin
- W. M. Keck Science Department, Pitzer College, Claremont, California; and
| | - Daniel E Crocker
- Department of Biology, Sonoma State University, Rohnert Park, California
| | - Rudy M Ortiz
- Molecular and Cellular Biology, University of California Merced, Merced, California
| |
Collapse
|
15
|
Czarnecka AM, Matak D, Szymanski L, Czarnecka KH, Lewicki S, Zdanowski R, Brzezianska-Lasota E, Szczylik C. Triiodothyronine regulates cell growth and survival in renal cell cancer. Int J Oncol 2016; 49:1666-78. [PMID: 27632932 DOI: 10.3892/ijo.2016.3668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/25/2016] [Indexed: 11/05/2022] Open
Abstract
Triiodothyronine plays an important role in the regulation of kidney cell growth, differentiation and metabolism. Patients with renal cell cancer who develop hypothyreosis during tyrosine kinase inhibitor (TKI) treatment have statistically longer survival. In this study, we developed cell based model of triiodothyronine (T3) analysis in RCC and we show the different effects of T3 on renal cell cancer (RCC) cell growth response and expression of the thyroid hormone receptor in human renal cell cancer cell lines from primary and metastatic tumors along with human kidney cancer stem cells. Wild-type thyroid hormone receptor is ubiquitously expressed in human renal cancer cell lines, but normalized against healthy renal proximal tube cell expression its level is upregulated in Caki-2, RCC6, SKRC-42, SKRC-45 cell lines. On the contrary the mRNA level in the 769-P, ACHN, HKCSC, and HEK293 cells is significantly decreased. The TRβ protein was abundant in the cytoplasm of the 786-O, Caki-2, RCC6, and SKRC-45 cells and in the nucleus of SKRC-42, ACHN, 769-P and cancer stem cells. T3 has promoting effect on the cell proliferation of HKCSC, Caki-2, ASE, ACHN, SK-RC-42, SMKT-R2, Caki-1, 786-0, and SK-RC-45 cells. Tyrosine kinase inhibitor, sunitinib, directly inhibits proliferation of RCC cells, while thyroid hormone receptor antagonist 1-850 (CAS 251310‑57-3) has less significant inhibitory impact. T3 stimulation does not abrogate inhibitory effect of sunitinib. Renal cancer tumor cells hypostimulated with T3 may be more responsive to tyrosine kinase inhibition. Moreover, some tumors may be considered as T3-independent and present aggressive phenotype with thyroid hormone receptor activated independently from the ligand. On the contrary proliferation induced by deregulated VHL and or c-Met pathways may transgress normal T3 mediated regulation of the cell cycle.
Collapse
Affiliation(s)
- Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Damian Matak
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Lukasz Szymanski
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Karolina H Czarnecka
- Department of Molecular Bases of Medicine, Medical University of Lodz, Lodz, Poland
| | - Slawomir Lewicki
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Robert Zdanowski
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | | | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
16
|
Tinnikov AA, Samuels HH. A novel cell lysis approach reveals that caspase-2 rapidly translocates from the nucleus to the cytoplasm in response to apoptotic stimuli. PLoS One 2013; 8:e61085. [PMID: 23596516 PMCID: PMC3626589 DOI: 10.1371/journal.pone.0061085] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 03/07/2013] [Indexed: 11/22/2022] Open
Abstract
Unlike other caspases, caspase-2 appears to be a nuclear protein although immunocytochemical studies have suggested that it may also be localized to the cytosol and golgi. Where and how caspase-2 is activated in response to apoptotic signals is not clear. Earlier immunocytochemistry studies suggest that caspase-2 is activated in the nucleus and through cleavage of BID leads to increased mitochondrial permeability. More recent studies using bimolecular fluorescence complementation found that caspase-2 oligomerization that leads to activation only occurs in the cytoplasm. Thus, apoptotic signals may lead to activation of caspase-2 which may already reside in the cytoplasm or lead to release of nuclear caspase-2 to the extra-nuclear cytoplasmic compartment. It has not been possible to study release of nuclear caspase-2 to the cytoplasm by cell fractionation studies since cell lysis is known to release nuclear caspase-2 to the extra-nuclear fraction. This is similar to what is known about unliganded nuclear estrogen receptor-α (ERα ) when cells are disrupted. In this study we found that pre-treatment of cells with N-ethylmaleimide (NEM), which alkylates cysteine thiol groups in proteins, completely prevents redistribution of caspase-2 and ERα from the nucleus to the extra-nuclear fraction when cells are lysed. Using this approach we provide evidence that apoptotic signals rapidly leads to a shift of caspase-2 from the nucleus to the extra-nuclear fraction, which precedes the detection of apoptosis. These findings are consistent with a model where apoptotic signals lead to a rapid shift of caspase-2 from the nucleus to the cytoplasm where activation occurs.
Collapse
Affiliation(s)
- Alexander A. Tinnikov
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Herbert H. Samuels
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
17
|
Burris TP, Solt LA, Wang Y, Crumbley C, Banerjee S, Griffett K, Lundasen T, Hughes T, Kojetin DJ. Nuclear receptors and their selective pharmacologic modulators. Pharmacol Rev 2013; 65:710-78. [PMID: 23457206 PMCID: PMC11060414 DOI: 10.1124/pr.112.006833] [Citation(s) in RCA: 206] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Nuclear receptors are ligand-activated transcription factors and include the receptors for steroid hormones, lipophilic vitamins, sterols, and bile acids. These receptors serve as targets for development of myriad drugs that target a range of disorders. Classically defined ligands that bind to the ligand-binding domain of nuclear receptors, whether they are endogenous or synthetic, either activate receptor activity (agonists) or block activation (antagonists) and due to the ability to alter activity of the receptors are often termed receptor "modulators." The complex pharmacology of nuclear receptors has provided a class of ligands distinct from these simple modulators where ligands display agonist/partial agonist/antagonist function in a tissue or gene selective manner. This class of ligands is defined as selective modulators. Here, we review the development and pharmacology of a range of selective nuclear receptor modulators.
Collapse
Affiliation(s)
- Thomas P Burris
- The Scripps Research Institute, 130 Scripps Way 2A1, Jupiter, FL 33458, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Affiliation(s)
- Valerie Anne Galton
- Department of Physiology and Neurobiology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, USA.
| |
Collapse
|
19
|
Pramfalk C, Pedrelli M, Parini P. Role of thyroid receptor β in lipid metabolism. Biochim Biophys Acta Mol Basis Dis 2010; 1812:929-37. [PMID: 21194564 DOI: 10.1016/j.bbadis.2010.12.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 12/08/2010] [Accepted: 12/20/2010] [Indexed: 12/19/2022]
Abstract
Thyroid hormones (THs) exert their actions by binding to thyroid hormone receptors (TRs) and thereby affect tissue differentiation, development, and metabolism in most tissues. TH-deficiency creates a less favorable lipid profile (e.g. increased plasma cholesterol levels), whereas TH-excess is associated with both positive (e.g. reduced plasma cholesterol levels) and negative (e.g. increased heart rate) effects. TRs are encoded by two genes, THRA and THRB, which, by alternative splicing, generate several isoforms (e.g. TRα1, TRα2, TRβ1, and TRβ2). TRα, the major TR in the heart, is crucial for heart rate and for cardiac contractility and relaxation, whereas TRβ1, the major TR in the liver, is important for lipid metabolism. Selective modulation of TRβ1 is thus considered as a potential therapeutic target to treat dyslipidemia without cardiac side effects. Several selective TH analogs have been tested in preclinical studies with promising results, but only a few of these compounds have so far been tested in clinical studies. This review focuses on the role of THs, TRs, and selective and non-selective TH analogs in lipid metabolism. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.
Collapse
Affiliation(s)
- Camilla Pramfalk
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, S-141 86 Stockholm, Sweden
| | | | | |
Collapse
|
20
|
Hall JA, Ribich S, Christoffolete MA, Simovic G, Correa-Medina M, Patti ME, Bianco AC. Absence of thyroid hormone activation during development underlies a permanent defect in adaptive thermogenesis. Endocrinology 2010; 151:4573-82. [PMID: 20660060 PMCID: PMC2940501 DOI: 10.1210/en.2010-0511] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 06/10/2010] [Indexed: 11/19/2022]
Abstract
Type 2 deiodinase (D2), which is highly expressed in brown adipose tissue (BAT), is an enzyme that amplifies thyroid hormone signaling in individual cells. Mice with inactivation of the D2 pathway (D2KO) exhibit dramatically impaired thermogenesis in BAT, leading to hypothermia during cold exposure and a greater susceptibility to diet-induced obesity. This was interpreted as a result of defective acute activation of BAT D2. Here we report that the adult D2KO BAT has a permanent thermogenic defect that stems from impaired embryonic BAT development. D2KO embryos have normal serum T3 but due to lack of D2-generated T3 in BAT, this tissue exhibits decreased expression of genes defining BAT identity [i.e. UCP1, PGC-1alpha and Dio2 (nonfunctional)], which results in impaired differentiation and oxidative capacity. Coinciding with a reduction of these T3-responsive genes, there is oxidative stress that in a cell model of brown adipogenesis can be linked to decreased insulin signaling and decreased adipogenesis. This discovery highlights the importance of deiodinase-controlled thyroid hormone signaling in BAT development, where it has important metabolic repercussions for energy homeostasis in adulthood.
Collapse
MESH Headings
- Acclimatization/genetics
- Acclimatization/physiology
- Adipocytes/cytology
- Adipocytes/metabolism
- Adipogenesis/genetics
- Adipogenesis/physiology
- Adipose Tissue, Brown/embryology
- Adipose Tissue, Brown/growth & development
- Adipose Tissue, Brown/metabolism
- Animals
- Blotting, Western
- Cell Differentiation/genetics
- Cell Differentiation/physiology
- Cells, Cultured
- Embryo, Mammalian/embryology
- Embryo, Mammalian/metabolism
- Embryo, Mammalian/physiology
- Female
- Gene Expression Regulation, Developmental
- Iodide Peroxidase/genetics
- Iodide Peroxidase/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oxygen Consumption/genetics
- Oxygen Consumption/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Temperature
- Thermogenesis/genetics
- Thermogenesis/physiology
- Thyroid Hormones/blood
- Thyroid Hormones/metabolism
- Time Factors
- Iodothyronine Deiodinase Type II
Collapse
Affiliation(s)
- Jessica A Hall
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The processes and pathways mediating the intermediary metabolism of carbohydrates, lipids, and proteins are all affected by thyroid hormones (THs) in almost all tissues. Particular attention has been devoted by scientists to the effects of THs on lipid metabolism. Among others, effects related to cholesterol, lipid handling, and cardiac performance have been the subject of study. Many reports are present in the literature concerning the calorigenic effect of THs, with most of them aimed at identifying the molecular basis of this effect. However, at the moment the mechanism(s) underlying the metabolic effects of THs remain to be elucidated. THs exert most of their effects though TH receptors (TRs). However, some effects of THs cannot be explained by a nuclear-mediated pathway, and recently an increasing number of nonnuclear actions have been described, which can provide a regulatory system of which the effects differ from those mediated on the transcriptional level by TRs. Some of the TH derivatives (naturally occurring metabolites and analogs) possess biological activities. TH-related biological effects have been described for physiological products such as tetraiodothyroacetic acid (Tetrac) and triiodothyroacetic acid (Triac) (via oxidative deamination and decarboxylation of thyroxine [T4] and triiodothyronine [T3] alanine chain), 3,3',5'-triiodothyronine (rT3) (via T4 and T3 deiodination), 3,3'-diiodothyronine (3,3'-T2) and 3,5-diiodothyronine (T2) (via T4, T3, and rT3 deiodination), and 3-iodothyronamine (T1AM) and thyronamine (T0AM) (via T4 and T3 deiodination and amino acid decarboxylation), as well as for TH structural analogs, such as 3,5,3'-triiodothyropropionic acid (Triprop), 3,5-dibromo-3-pyridazinone-l-thyronine (L-940901), N-[3,5-dimethyl-4-(4'-hydroxy-3'-isopropylphenoxy)-phenyl]-oxamic acid (CGS 23425), 3,5-dimethyl-4[(4'-hydroxy-3'-isopropylbenzyl)-phenoxy] acetic acid (GC-1), 3,5-dichloro-4[(4-hydroxy-3-isopropylphenoxy)phenyl] acetic acid (KB-141), and 3,5-diiodothyropropionic acid (DITPA). Most of these compounds have interesting properties: counteracting lipid accumulation, reducing cholesterol level, and increasing lipid metabolism without cardiotoxic effects. Hopefully, further studies on basic mechanisms of such compounds will be harbingers of more knowledge on the metabolic effects of TH derivatives and on their possible clinical application.
Collapse
Affiliation(s)
- Maria Moreno
- Dipartimento di Scienze Biologiche ed Ambientali, Università degli Studi del Sannio, Via Port'Arsa, Benevento, Italy
| | | | | | | | | | | |
Collapse
|
22
|
Abstract
Thyroid hormones (THs) have important effects on cellular development, growth, and metabolism. They bind to thyroid hormone receptors (TRs), TRalpha and TRbeta, which belong to the nuclear hormone receptor superfamily. These receptors also bind to enhancer elements in the promoters of target genes, and can regulate both positive and negative transcription. Recent emerging evidence has characterized some of the molecular mechanisms by which THs regulate transcription as co-repressors, and co-activators have been identified and their effects on histone acetylation examined. THs also have rapid effects that do not require transcription. These can occur via TRs or other cellular proteins, and typically occur outside the nucleus. It appears that THs regulate multiple cellular functions using a diverse array of receptors and signaling systems. TR isoform- or pathway-specific drugs might provide the therapeutic benefits of TH action such as decreasing obesity or lowering cholesterol levels without some of the side effects of hyperthyroidism.
Collapse
Affiliation(s)
- Alexis Oetting
- Laboratory of Gene Regulation and Development, National Institute of Child Health and Development, National Institute of Health, Bethesda, MD, USA
| | | |
Collapse
|
23
|
|
24
|
|
25
|
Siddiqi A, Parsons MP, Lewis JL, Monson JP, Williams GR, Burrin JM. TR expression and function in human bone marrow stromal and osteoblast-like cells. J Clin Endocrinol Metab 2002; 87:906-14. [PMID: 11836340 DOI: 10.1210/jcem.87.2.8226] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormones influence both bone formation and bone resorption. In vitro studies demonstrate direct effects of thyroid hormones on cells of the osteoblast lineage. Transcriptional regulation by thyroid hormones is mediated by ligand-dependent transcription factors called TRs. The three main T(3)-binding TR isoforms are TRalpha1, TRbeta1, and TRbeta2. TRs have been identified in cells of the osteoblast lineage, but it is still not known whether TR isoform expression differs in primary cultures of human osteoblasts. We used immunocytochemistry, Western blotting, nuclear binding assays, and transient transfection studies to examine the expression of functional TR isoforms in primary cultures of osteoblasts (hOb) derived from explants of trabecular bone, in human bone marrow stromal cells (hBMS), which are believed to be the source of osteoblast progenitor cells, and for comparison in the transformed human osteosarcoma cell lines MG63 and SaOs-2. TRalpha1, TRbeta1, and TRbeta2 proteins were expressed in all cells, although expression was greatest in MG63 > hBMS > SaOs-2 > hOb. Differences between isoforms were also apparent, with TRalpha1> TRbeta1 > TRbeta2 in all cell types. Incubation with [(125)I]T(3) confirmed reversible T(3) binding to cell nuclei. Specific binding was greatest in MG63 > hBMS > SaOs-2 > hOb. Finally, endogenous TR activity was determined in transfections using a thyroid hormone response element derived from the rat GH gene linked to the luciferase reporter gene. In MG63 and hBMS cells T(3) treatment increased luciferase activity 5.5 +/- 0.7-fold (P < 0.05), confirming the presence of endogenous receptors. In SaOs-2 and hOb cells, T(3) treatment had no effect on thyroid hormone response element-thymidine kinase-luciferase expression, suggesting that in these cells TR expression was too low to be detected. These results indicate that three main TR isoforms are expressed in cells of the human osteoblast lineage, but that expression and endogenous TR activity are predominantly present in hBMS cells. Whether there are distinct mechanisms of thyroid hormone action mediated by TRalpha1, TRbeta1, and TRbeta2 in hOb and hBMS cells remains to be shown.
Collapse
Affiliation(s)
- Ayesha Siddiqi
- Department of Endocrinology, St. Bartholomew's and the Royal London School of Medicine and Dentistry, West Smithfield, London, United Kingdom EC1A 7BE
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
Thyroid hormones (THs) play critical roles in the differentiation, growth, metabolism, and physiological function of virtually all tissues. TH binds to receptors that are ligand-regulatable transcription factors belonging to the nuclear hormone receptor superfamily. Tremendous progress has been made recently in our understanding of the molecular mechanisms that underlie TH action. In this review, we present the major advances in our knowledge of the molecular mechanisms of TH action and their implications for TH action in specific tissues, resistance to thyroid hormone syndrome, and genetically engineered mouse models.
Collapse
Affiliation(s)
- P M Yen
- Molecular Regulation and Neuroendocrinology Section, Clinical Endocrinology Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
27
|
Manna PR, Roy P, Clark BJ, Stocco DM, Huhtaniemi IT. Interaction of thyroid hormone and steroidogenic acute regulatory (StAR) protein in the regulation of murine Leydig cell steroidogenesis. J Steroid Biochem Mol Biol 2001; 76:167-77. [PMID: 11384875 DOI: 10.1016/s0960-0760(00)00156-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The steroidogenic acute regulatory (StAR) protein, a novel phosphoprotein, is a crucial factor involved in intramitochondrial cholesterol transportation, the rate-limiting step in steroidogenesis. The present investigations were undertaken to elucidate involvement of thyroid hormone and StAR protein in the regulation of steroidogenesis in mouse Leydig cells. Treatment of cells with triiodothyronine (T(3)) coordinately augmented the levels of StAR protein, StAR mRNA, and steroid production, and these responses were progressively dependent on expression of steroidogenic factor 1 (SF-1). With regard to steroidogenesis and StAR expression, the T(3) response requires both on-going mRNA and protein synthesis. In addition, the effects of T(3) were acutely modulated at the steroidogenic machinery and luteinizing hormone receptor (LHR) function, while these levels were suppressed following longer periods of exposure to T(3). Furthermore, the inhibition of SF-1 expression by DAX-1 markedly abolished T(3)-mediated StAR expression in a time frame, which was consistent with decreased steroid biosynthesis. Specific involvement of SF-1 was further confirmed by assessing the 5'-flanking region of the mouse StAR gene, which identified a region between -254 and -110 bp that was essential for T(3) function. Importantly, it was found that the SF-1 binding site at position -135 bp of the 5'-flanking region was greatly involved in T(3)-mediated reporter activity. Electrophoretic mobility shift assays (EMSA) also demonstrated involvement of SF-1 in T(3) function. The relevance of T(3)-mediated LHR function was investigated in mice rendered hypo-and hyperthyroid, which accounted for up-regulation in the former and down-regulation in the latter group, respectively. These findings demonstrate a key role of thyroid hormone in maintaining mouse Leydig cell function, where thyroid hormone and StAR protein coordinately regulate steroid hormone biosynthesis.
Collapse
Affiliation(s)
- P R Manna
- Department of Physiology, Institute of Biomedicine, University of Turku, kiinamyllynkatu 10, FIN-20520, Turku, Finland
| | | | | | | | | |
Collapse
|
28
|
Manna PR, Kero J, Tena-Sempere M, Pakarinen P, Stocco DM, Huhtaniemi IT. Assessment of mechanisms of thyroid hormone action in mouse Leydig cells: regulation of the steroidogenic acute regulatory protein, steroidogenesis, and luteinizing hormone receptor function. Endocrinology 2001; 142:319-31. [PMID: 11145595 DOI: 10.1210/endo.142.1.7900] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recently, we demonstrated that triiodothyronine (T(3)) stimulated steroid hormone biosynthesis and steroidogenic acute regulatory (StAR) protein expression in mLTC-1 mouse Leydig tumor cells through the mediation of steroidogenic factor 1 (SF-1). We now report a dual response mechanism of T(3) on steroidogenesis and StAR expression, and on LH receptor (LHR) expression and binding in mLTC-1 cells. T(3) acutely (8 h), induced a 260% increase in StAR messenger RNA (mRNA) expression over the basal level which was coincident with an increase in progesterone (P) production. In contrast, chronic stimulation with T(3) (beyond 8 h), resulted in an attenuation of StAR expression and P production. This attenuation was most likely caused by a decrease in cholesterol delivery to the inner mitochondrial membrane as demonstrated by incubations with the hydrophilic steroid precursors, 22R hydroxycholesterol and pregnenolone, which restored P synthesis. In similar studies, chronic treatment with T(3) increased the levels of cytochrome P450scc mRNA by 83%, whereas those of cytochrome P450 17alpha-hydroxylase and 3ss-hydroxysteroid dehydrogenase decreased. The diminished response in steroidogenesis following chronic T(3) exposure was not a result of alterations in StAR mRNA stability, but rather was due to inhibition of transcription of the StAR gene. Similar acute stimulatory and chronic inhibitory responses to T(3) were found when LHR mRNA expression and LHR ligand binding were examined. Transfections with an LHR or StAR promoter/luciferase reporter construct demonstrated that a 173-bp fragment of the LHR promoter containing an SF-1 binding motif was involved in T(3) response, as was the SF-1 recognition site at -135 bp in the StAR promoter. Furthermore, the importance of SF-1 in T(3) function was also verified employing mutation in the bases of SF-1 sequences using electrophoretic mobility shift assays. The potential physiological relevance of these findings was demonstrated when similar responses were obtained in mice rendered hypo and hyperthyroid. Collectively, these observations further characterize the thyroid-gonadal connection and provide insights into the mechanisms for a dual regulatory role of thyroid hormone in Leydig cell functions.
Collapse
Affiliation(s)
- P R Manna
- Department of Physiology, Institute of Biomedicine, University of Turku, FIN-20520 Turku, Finland
| | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Koenig RJ. Thyroid Hormone Receptors. Compr Physiol 1999. [DOI: 10.1002/cphy.cp070523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Brent GA. Regulation of Gene Expression by Thyroid Hormones: Relation to Growth and Development. Compr Physiol 1999. [DOI: 10.1002/cphy.cp070524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
32
|
Abstract
The past 25 years have witnessed dramatic changes in our concepts of thyroid hormone action. Progress in this area was made possible by the recognition of the central role of triiodothyronine in mediating thyroid hormone action and the recognition of specific nuclear receptors in target tissues as demonstrated by displacement studies. The cloning of the receptors and receptor variants has enabled investigators to undertake detailed analyses of the biochemical events which underlie the physiological and pathological action of thyroid hormone.
Collapse
Affiliation(s)
- J H Oppenheimer
- Department of Medicine, University of Minnesota, Minneapolis 55466, USA
| |
Collapse
|
33
|
Hsieh JC, Shimizu Y, Minoshima S, Shimizu N, Haussler CA, Jurutka PW, Haussler MR. Novel nuclear localization signal between the two DNA-binding zinc fingers in the human vitamin D receptor. J Cell Biochem 1998. [DOI: 10.1002/(sici)1097-4644(19980701)70:1<94::aid-jcb10>3.0.co;2-b] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
34
|
Motomura K, Brent GA. Mechanisms of thyroid hormone action. Implications for the clinical manifestation of thyrotoxicosis. Endocrinol Metab Clin North Am 1998; 27:1-23. [PMID: 9534024 DOI: 10.1016/s0889-8529(05)70294-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Serum thyroid hormone concentrations alone do not explain the variability and severity of the range of symptoms observed in thyrotoxic patients. Despite gaps in our understanding of the links between the clinical manifestations of thyrotoxicosis and the underlying mechanisms, much has been learned. A limited number of markers directly reflect T3 action. The future elucidation of T3 targets that mediate these effects should ultimately lead to additional clinical markers of tissue-specific T3 action. The availability of such tests should allow for more specific treatment of individual patients.
Collapse
Affiliation(s)
- K Motomura
- Department of Medicine, University of California-Los Angeles School of Medicine, USA
| | | |
Collapse
|
35
|
Bogazzi F, Bartalena L, Brogioni S, Burelli A, Grasso L, Dell'Unto E, Manetti L, Martino E. L-thyroxine directly affects expression of thyroid hormone-sensitive genes: regulatory effect of RXRbeta. Mol Cell Endocrinol 1997; 134:23-31. [PMID: 9406846 DOI: 10.1016/s0303-7207(97)00156-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
L-thyroxine (T4) has been considered mainly a prohormone, the hormonal action of which is related to its conversion to 3,5,3'-triiodothyronine (T3) in peripheral tissues. In this study we investigated in transient transfection assays whether T4 might directly affect the expression of thyroid hormone (TH) sensitive genes. The reporter construct ME-TRE-TK-CAT or TSH-TRE-TK-CAT containing the nucleotide sequence of the TH response element (TRE) of either malic enzyme (ME) or TSHbeta genes, was transfected with either TH receptor (TR) alpha alone or in combination with retinoid X receptor (RXR) beta into NIH3T3 cells. Addition of 100 nM T4 to the culture medium in the presence of TRalpha increased the basal level of ME-TRE-TK-CAT expression by 4.5-fold. T4 action was due to a direct interaction with TRalpha and not to its conversion to T3, since T4 effect persisted in the presence of 5'-deiodinase inhibitors (propylthiouracil, iopanoic acid) effectively preventing T3 generation, as assessed by the absence of T3 by HPLC in the cellular extracts of transfected cells. In a dose-response study half-maximal stimulation by T4 was achieved at a concentration of 100 nM, whereas 50% of maximal induction was produced by 1 nM T3 and 6 nM triiodothyroacetic acid (TRIAC). Coexpression of RXRbeta greatly enhanced the transcriptional activity of the ME-TRE-TK-CAT gene when either T3, T4 or TRIAC was added to the culture medium of NIH3T3 cells, but established a hormonal hierarchy in the reporter activation different than that observed in the presence of TRalpha alone (TRIAC > T3 > or = T4, instead of T3 > TRIAC > T4). T4 at a concentration of 100 nM could activate the TH/TR-dependent down-regulation mediated by the negative TSH-TRE, although at a lower level than that obtained with similar concentrations of T3 (35 and 55% inhibition, respectively). Our results demonstrate that, in addition to the action mediated through its monodeiodination to T3, T4 exerts a direct effect on genes that are either positively or negatively regulated by TH. Moreover, RXRbeta, forming heterodimers with TRs, appeared to exert a central role in modulating the sensitivity of TH-responsive genes to different iodothyronines.
Collapse
Affiliation(s)
- F Bogazzi
- Istituto di Endocrinologia, Università di Pisa, Presidio Ospedaliero di Cisanello, Italy
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
During brain development, before the apparatus of neurotransmission has been set into place, many neurotransmitters act as growth regulators. In adult brain, their role in neurotransmission comes to the fore but neuronal plasticity and other growth-related processes are their continuing responsibility. This has been clearly demonstrated for catecholamines. Previous as well as recent evidence now indicates that thyroid hormones may participate in the developing and adult brain through similar mechanisms. Immunohistochemical mapping of brain triiodothyronine (antibody specificity established by numerous appropriate tests) demonstrated that the hormone was concentrated in both noradrenergic centers and noradrenergic projection sites. In the centers (locus coeruleus and lateral tegmental system) triiodothyronine staining, like that of tyrosine hydroxylase, was heavily concentrated in cytosol and cell processes. By contrast, in noradrenergic targets, label was most prominent in cell nuclei. Combined biochemical and morphologic data allows a construct of thyroid hormone circuitry to unfold: The locus coeruleus is conveniently located just beneath the ependyma of the 4th ventricle. Thyroxine, entering the brain via the choroid plexus, is preferentially delivered to subependymal brain structures. High concentrations of locus coeruleus norepinephrine promote active conversion of thyroxine to triiodothyronine, leading to the preeminence of the locus coeruleus as a site of triiodothyronine concentration. Results of treatment with the locus coeruleus neurotoxin DSP-4 established that axonal transport accounts for delivery of both triiodothyronine and norepinephrine from locus coeruleus to noradrenergic terminal fields. The apparatus for transduction of thyronergic and noradrenergic signals at both membrane and nuclear sites resides in the postsynaptic target cells. Upon internalization of hormone in post-synaptic target cells, genomic effects of triiodothyronine, norepinephrine, and/or their second messengers are possible and expected. The evidence establishes a direct morphologic connection between central thyronergic and noradrenergic systems, supporting earlier proposals that triiodothyronine or its proximate metabolites may serve as cotransmitters with norepinephrine in the adrenergic nervous system.
Collapse
Affiliation(s)
- M B Dratman
- Department of Medicine, MCP Hanneman School of Medicine, Allegheny University, and Medical Research Service, Veterans Affairs Medical Center, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
37
|
Fierro-Renoy JF, Szuchet S, Falcone M, Macchia E, DeGroot L. Three different thyroid hormone receptor isoforms are detected in a pure culture of ovine oligodendrocytes. Glia 1995; 14:322-8. [PMID: 8530188 DOI: 10.1002/glia.440140408] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Thyroid hormones are important for the normal development of the central nervous system. In humans, the period around the end of the intrauterine life and the first few months of neonatal life is critically dependent on the presence of normal amounts of thyroid hormone. There are significant events occurring during this time; myelination is one. Myelin is synthesized by oligodendrocytes. A panel of site-specific polyclonal antibodies against alpha-1 thyroid hormone receptor (TR), alpha-2 variant TR, and beta-1 TR isoforms has been employed to investigate the presence of TR isoforms in a pure culture of ovine oligodendrocytes by the avidin-biotin peroxidase immunocytochemical method. Strong nuclear staining was obtained with all the anti-TR antibodies; no reaction products were detected in the cytoplasm or cellular processes. By contrast, an anti-myelin basic protein antibody gave strong cytoplasmic and process staining; no nuclear staining was seen. These latter results served to 1) confirm that the cells under study are oligodendrocytes; and 2) prove that the nuclear staining with anti-TR antibodies is specific. Preimmune sera were totally negative. Scatchard analysis of [125I] T3 binding by isolated oligodendrocyte nuclei demonstrated the existence of high-affinity--low-capacity T3 binding sites with a Ka of approximately 6 x 10(-9) M and a maximal binding capacity of approximately 20 fmol/100 micrograms of DNA. Our results demonstrate that differentiated oligodendrocytes express alpha-1 and alpha-2 variant and beta-1 isoforms of TR at the protein level and support the notion of a direct impact of thyroid hormones on oligodendrocytes in their regulation of myelin synthesis.
Collapse
Affiliation(s)
- J F Fierro-Renoy
- Department of Medicine, University of Chicago, Illinois 60637, USA
| | | | | | | | | |
Collapse
|
38
|
Walter IB, Droz B. Nuclear and cytoplasmic triiodothyronine-binding sites in primary sensory neurons and Schwann cells: radioautographic study during development. J Neuroendocrinol 1995; 7:127-36. [PMID: 7767325 DOI: 10.1111/j.1365-2826.1995.tb00675.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The effects of the thyroid hormones on target cells are mediated through nuclear T3 receptors. In the peripheral nervous system, nuclear T3 receptors were previously detected with the monoclonal antibody 2B3 mAb in all the primary sensory neurons throughout neuronal life and in peripheral glia at the perinatal period only (Eur. J. Neurosci. 5, 319, 1993). To determine whether these nuclear T3 receptors correspond to functional ones able to bind T3, cryostat sections and in vitro cell cultures of dorsal root ganglion (DRG) or sciatic nerve were incubated with 0.1 nM [125I]-labeled T3, either alone to visualize the total T3-binding sites or added with a 10(3) fold excess of unlabeled T3 to estimate the part due to the non-specific T3-binding. After glutaraldehyde fixation, radioautography showed that the specific T3-binding sites were largely prevalent. The T3-binding capacity of peripheral glia in DRG and sciatic nerve was restricted to the perinatal period in vivo and to Schwann cells cultured in vitro. In all the primary sensory neurons, specific T3-binding sites were disclosed in foetal as well as adult rats. The detection of the T3-binding sites in the nucleus indicated that the nuclear T3 receptors are functional. Moreover the concomitant presence of both T3-binding sites and T3 receptors alpha isoforms in the perikaryon of DRG neurons infers that: 1) [125I]-labeled T3 can be retained on the T3-binding 'E' domain of nascent alpha 1 isoform molecules newly-synthesized on the perikaryal ribosomes; 2) the alpha isoforms translocated to the nucleus are modified by posttranslational changes and finally recognized by 2B3 mAb as nuclear T3 receptor. In conclusion, the radioautographic visualization of the T3-binding sites in peripheral neurons and glia confirms that the nuclear T3 receptors are functional and contributes to clarify the discordant intracellular localization provided by the immunocytochemical detection of nuclear T3 receptors and T3 receptor alpha isoforms.
Collapse
Affiliation(s)
- I B Walter
- Institut d'Histologie et d'Embryologie, Faculté de Médecine, Université de Lausanne, Switzerland
| | | |
Collapse
|
39
|
Duchamp C, Burton KA, Herpin P, Dauncey MJ. Perinatal ontogeny of porcine nuclear thyroid hormone receptors and its modulation by thyroid status. THE AMERICAN JOURNAL OF PHYSIOLOGY 1994; 267:E687-93. [PMID: 7977719 DOI: 10.1152/ajpendo.1994.267.5.e687] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Induction of nuclear thyroid hormone receptors (TRs) represents a key point in the control of growth, development, differentiation, and metabolism of most tissues. The influence of thyroid status on the ontogeny of hepatic and skeletal muscle TRs has been investigated in perinatal pigs. Plasma concentrations of total and free 3,5,3'-triiodothyronine (T3) increased markedly from 80 days of fetal life (80 f) to 2 days of postnatal life. Test piglets obtained from sows fed a high glucosinolate rapeseed diet had lower T3 and thyroxine levels than controls at 110 f and showed a higher postnatal surge in T3. Maximal T3 binding capacity (Bmax, pmol T3/mg DNA, means +/- SE) in liver increased from 0.07 +/- 0.01 at 80 f to 0.37 +/- 0.02 at birth and then plateaued. In longissimus dorsi muscle, Bmax values were much higher than in liver and increased from 0.90 +/- 0.02 at 80 f to 1.37 +/- 0.13 at birth and then declined to 1.09 +/- 0.11 at 2 days of age. Long-term fetal hypothyroidism affected the ontogenic profile of both liver and muscle receptors but in opposite directions; Bmax values were reduced in liver but increased in muscle. Postnatally, lower muscle Bmax values occurred in parallel with transient higher levels of circulating T3. Apparent binding affinities were slightly different in liver and muscle during fetal life, and there was an effect of age in muscle. In conclusion, as far as the receptor is concerned, fetal muscle can potentially respond to thyroid hormones much earlier in development than the liver.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- C Duchamp
- Department of Cellular Physiology, Babraham Institute, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
40
|
Bandyopadhyay A, Bhattacharya S. Purification of putative thyroid hormone receptor from the ovarian nuclei of fresh water perch, Anabas testudineus. FISH PHYSIOLOGY AND BIOCHEMISTRY 1994; 13:387-398. [PMID: 24197075 DOI: 10.1007/bf00003418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/26/1994] [Indexed: 06/02/2023]
Abstract
Perch ovarian putative T3 (3,5,3'-triiodo-L-thyroxine) receptor was purified to 580-fold by extracting the ovarian nuclear preparation with 0.4 M KCl, gel filtration on Sephadex G-25, DEAE-Sephacel chromatography and FPLC Superose 6 chromatography. To monitor the T3 binding protein at each purification step, aliquots from each peak protein fractions were incubated with (125)I-T3 (0.16 pmol to 3.2 nmol) in the absence or presence of 500-fold excess of unlabelled T3. Maximum binding capacity (Bmax) obtained from the Scatchard plot analysis was estimated to determine the extent of purification at each step. Purified putative T3 receptors showed a single band in polyacrylamide gel electrophoresis (PAGE) indicating homogeneity of the putative receptor protein. The molecular weight of the putative T3 receptor protein, as determined on a FPLC Superose 6 column, was 50 kD. Treatment of putative T3 receptor protein with β-mercaptoethanol followed by SDS-PAGE resulted in two subunits of 26 and 31 kD. Purification increased the specific activity of the receptor, but did not alter its affinity. Analogue specificity of the purified receptor corresponded to that of the crude nuclear preparation. Triiodothyroacetic acid (Triac) and T3 equally competed in inhibiting radiolabelled T3 binding while thyroxine (T4) was a poor competitor. T3 receptor antiserum crossreacted with the receptor protein. (125)I-labelled receptor protein binding with its antiserum was inhibited by increasing logarithmic concentrations of unlabelled receptor. In contrast to earlier reports on hepatic T3 receptor, which is a monomer, present investigation demonstrated T3 binding protein in the perch ovary to be a heterodimer held together by disulphide bond.
Collapse
Affiliation(s)
- A Bandyopadhyay
- Department of Zoology, Visva-Bharati University, Santiniketan-731 235, W. Bengal, India
| | | |
Collapse
|
41
|
Zhu Y, Goodridge AG, Stapleton SR. Zinc, vanadate and selenate inhibit the tri-iodothyronine-induced expression of fatty acid synthase and malic enzyme in chick-embryo hepatocytes in culture. Biochem J 1994; 303 ( Pt 1):213-6. [PMID: 7945243 PMCID: PMC1137578 DOI: 10.1042/bj3030213] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Insulin regulates the expression of genes involved in a variety of metabolic processes. In chick-embryo hepatocytes in culture, insulin amplifies the tri-iodothyronine (T3)-induced enzyme activity, and the level and rate of transcription of mRNA for both fatty acid synthase (FAS) and malic enzyme (ME). Insulin alone, however, has little or no effect on the expression of these genes. In chick-embryo hepatocytes, the mechanism by which insulin regulates the expression of these or other genes is not known. Several recent studies have compared the effects of zinc, vanadate and selenate on insulin-sensitive processes in an attempt to probe the mechanism of insulin action. Because zinc, vanadate and selenate mimic the effects of insulin on several processes, they have been termed insulin-mimetics. We have studied the effect of zinc, vanadate and selenate on the T3-induced expression of both FAS and ME. Like insulin, these agents had little or no effect on the basal activities for FAS and ME in chick-embryo hepatocytes in culture for 48 h. Unlike insulin, however, zinc, vanadate and selenate inhibited the T3-induced activities and mRNA levels of both FAS and ME. Maximal inhibition was achieved at concentrations of 50 microM zinc or vanadate, or 20 microM selenate. Zinc and vanadate also inhibited the T3-induced transcription of the FAS and ME genes. Although the mechanism of this inhibition is unknown, our results indicate that it is not mediated through inhibition of binding of T3 to its nuclear receptor nor through a general toxic effect. Thus zinc, vanadate and selenate are not insulin-mimetics under all conditions, and their effects on other insulin-sensitive processes may be fortuitous and unrelated to actions or components of the insulin signalling pathway.
Collapse
Affiliation(s)
- Y Zhu
- Department of Chemistry, Western Michigan University, Kalamazoo 49008
| | | | | |
Collapse
|
42
|
Atkinson BG. Metamorphosis: Model systems for studying gene expression in postembryonic development. ACTA ACUST UNITED AC 1994. [DOI: 10.1002/dvg.1020150402] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
43
|
Rodríguez M, Jolin T. Triiodothyronine receptor complex in developing rat brain and pituitary. THE AMERICAN JOURNAL OF PHYSIOLOGY 1993; 264:E804-9. [PMID: 8498502 DOI: 10.1152/ajpendo.1993.264.5.e804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In vitro saturation analysis combined with nuclear 3,5,3'-triiodothyronine (T3) quantification was used to examine the changes in T3 binding parameters in rat pituitary and cerebrocortical nuclei from fetal day 14 to postnatal day 20. T3 receptors were first detectable in neuronal, glial, and pituitary nuclei on fetal days 14, 17, and 18, respectively. Thereafter T3 receptor concentrations in neuronal, glial, and pituitary nuclei increased throughout the developmental period studied, reaching maximal levels during neonatal life (1,129, 1,025, and 635 fmol/mg DNA, respectively). T3 levels in pituitary, neuronal, and glial nuclei also increased during development there being a 35-, 34-, and 120-fold rise between fetal days 16-18 and the 20th postnatal day. Endogenous T3 receptor occupancy throughout the experimental period increased six- to ninefold in the three types of nuclei. The presence of T3 receptor complex in the pituitary and cerebrocortical nuclei during perinatal development lends further support to the hypothesis that T3 may be an important factor in determining the differentiation and development of these cells.
Collapse
Affiliation(s)
- M Rodríguez
- Instituto de Investigaciones Biomédicas, Madrid, Spain
| | | |
Collapse
|
44
|
Ligand-dependent, Pit-1/growth hormone factor-1 (GHF-1)-independent transcriptional stimulation of rat growth hormone gene expression by thyroid hormone receptors in vitro. Mol Cell Biol 1993. [PMID: 8441408 DOI: 10.1128/mcb.13.3.1719] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The expression of the rat growth hormone (rGH) gene in the anterior pituitary gland is modulated by Pit-1/GHF-1, a pituitary-specific transcription factor, and by other more widely distributed factors, such as the thyroid hormone receptors (TRs), Sp1, and the glucocorticoid receptor. Thyroid hormone (T3)-mediated transcriptional stimulation of rGH gene expression has been extensively studied in vivo and in vitro including the measurements of (i) rGH mRNA by blot hybridization, (ii) transcriptional rate of rGH gene by nuclear run-on, and (iii) reporter gene expression in which a chimeric plasmid containing 5'-flanking sequences of the rGH gene linked to a reporter gene has been transfected either stably or transiently into pituitary and/or nonpituitary cells. From these studies, it has been suggested that the Pit-1/GHF-1 binding site is necessary for full T3 action. We developed a cell-free in vitro transcription system to examine further the roles of the TRs and Pit-1/GHF-1 in rGH gene activation. Using GH3 nuclear extract as a source of TRs and Pit-1/GHF-1, this in vitro transcription assay showed that T3 stimulation of rGH promoter activity is dependent on the addition of T3 to the GH3 nuclear extract. This transcriptional stimulation was augmented with increasing concentrations of ligand and was T3, but not T4 or reverse T3, specific. T3-mediated stimulation of rGH promoter activity was completely abolished by preincubation of the nuclear extract with rGH-thyroid hormone response element (-200 to -160) but not with Pit-1/GHF-1 (-137 to -65) oligonucleotides. Further, neither deletion of both Pit-1/GHF-1 binding sites nor mutation of the proximal Pit-1/GHF-1 binding site from the rGH promoter abrogated the T3 effect. These results provide evidence that T3-stimulated rGH promoter activity is independent of Pit-1/GHF-1 and raise the possibility that the stimulation of rGH gene expression by T3 might involve direct interaction of TRs with the general transcriptional apparatus.
Collapse
|
45
|
Suen CS, Chin WW. Ligand-dependent, Pit-1/growth hormone factor-1 (GHF-1)-independent transcriptional stimulation of rat growth hormone gene expression by thyroid hormone receptors in vitro. Mol Cell Biol 1993; 13:1719-27. [PMID: 8441408 PMCID: PMC359484 DOI: 10.1128/mcb.13.3.1719-1727.1993] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The expression of the rat growth hormone (rGH) gene in the anterior pituitary gland is modulated by Pit-1/GHF-1, a pituitary-specific transcription factor, and by other more widely distributed factors, such as the thyroid hormone receptors (TRs), Sp1, and the glucocorticoid receptor. Thyroid hormone (T3)-mediated transcriptional stimulation of rGH gene expression has been extensively studied in vivo and in vitro including the measurements of (i) rGH mRNA by blot hybridization, (ii) transcriptional rate of rGH gene by nuclear run-on, and (iii) reporter gene expression in which a chimeric plasmid containing 5'-flanking sequences of the rGH gene linked to a reporter gene has been transfected either stably or transiently into pituitary and/or nonpituitary cells. From these studies, it has been suggested that the Pit-1/GHF-1 binding site is necessary for full T3 action. We developed a cell-free in vitro transcription system to examine further the roles of the TRs and Pit-1/GHF-1 in rGH gene activation. Using GH3 nuclear extract as a source of TRs and Pit-1/GHF-1, this in vitro transcription assay showed that T3 stimulation of rGH promoter activity is dependent on the addition of T3 to the GH3 nuclear extract. This transcriptional stimulation was augmented with increasing concentrations of ligand and was T3, but not T4 or reverse T3, specific. T3-mediated stimulation of rGH promoter activity was completely abolished by preincubation of the nuclear extract with rGH-thyroid hormone response element (-200 to -160) but not with Pit-1/GHF-1 (-137 to -65) oligonucleotides. Further, neither deletion of both Pit-1/GHF-1 binding sites nor mutation of the proximal Pit-1/GHF-1 binding site from the rGH promoter abrogated the T3 effect. These results provide evidence that T3-stimulated rGH promoter activity is independent of Pit-1/GHF-1 and raise the possibility that the stimulation of rGH gene expression by T3 might involve direct interaction of TRs with the general transcriptional apparatus.
Collapse
Affiliation(s)
- C S Suen
- Department of Medicine, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, Massachusetts
| | | |
Collapse
|
46
|
Roncero C, Goodridge A. Hexanoate and octanoate inhibit transcription of the malic enzyme and fatty acid synthase genes in chick embryo hepatocytes in culture. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(18)42128-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
47
|
Overexpression of the alpha-thyroid hormone receptor in avian cell lines. Effects on expression of the malic enzyme gene are selective and cell-specific. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(19)49838-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
48
|
MacLatchy DL, Eales JG. Intra- and extra-cellular sources of T3 binding to putative thyroid hormone receptors in liver, kidney, and gill nuclei of immature rainbow trout, Oncorhynchus mykiss. THE JOURNAL OF EXPERIMENTAL ZOOLOGY 1992; 262:22-9. [PMID: 1583450 DOI: 10.1002/jez.1402620105] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The sources of extracellular and intracellular 3,5,3'-triiodo-L-thyronine (T3) binding to putative thyroid hormone receptors in liver, kidney, and gill nuclei were determined in vivo for immature rainbow trout at 12 degrees C. Both [131I]T3 and [125I]T4 were injected intraperitoneally, the plasma and tissues were examined at isotopic equilibrium at 20 h, and the proportions of intracellular [125I]T3 and extracellular [131I]T3 saturably bound in the nucleus were determined. Comparable total amounts of T3 were saturably bound in the nuclei of liver (7.2), kidney (8.0), and gill (9.7 moles x 10(-13) .mg DNA-1), but the percentage of nuclear T3 generated within the target cell was greater for gill (76%) than for liver (50%) and kidney (28%). Both gill and liver possess a low Km T4 5'monodeiodinase which could be responsible for the high proportion of the nuclear T3 generated within those tissues.
Collapse
Affiliation(s)
- D L MacLatchy
- Department of Zoology, University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
49
|
|
50
|
Halperin Y, Shapiro LE, Surks MI. Role of L-thyroxine in nuclear thyroid hormone receptor occupancy and growth hormone production in cultured GC cells. J Clin Invest 1991; 88:1291-9. [PMID: 1918379 PMCID: PMC295598 DOI: 10.1172/jci115433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The contribution of L-thyroxine (T4) to nuclear thyroid receptor occupancy was studied in GC cells incubated with concentrations of 3,5,3'-triiodo-L-thyronine (T3) and T4 that resulted in free iodothyronine levels similar to those in serum of euthyroid rats. T4 accounted for 5.4-10% of the occupied receptors: T3 derived from T4 [T3(T4)] and T3 added to medium accounted for the remainder of receptor occupancy. Incubation with increasing medium free T4 resulted in a progressive increase in the contribution of T4 and T3(T4) to receptor occupancy. In incubations with 3.6-fold increased medium free T4, T4 accounted for 20.4%, and T3(T4) for 40.3% of receptor occupancy. These occupancy data and the experimentally determined Ka of thyroid receptor for T3 and T4 allowed calculation of nuclear free iodothyronine concentrations. Nuclear free T3 was 3-6-fold greater than medium free T3 and nuclear [corrected] free T4 was 12-19-fold greater than medium free T4. When GC cells were incubated with decreased medium free T3 and physiological medium free T4, both nuclear receptor occupancy and growth hormone production decreased as well. However, a twofold increase in medium free T4, in the presence of decreased medium free T3, restored receptor occupancy and growth hormone production to or near control values. These findings establish a role for T4 in addition to T3(T4) in nuclear receptor occupancy and biological activity in rat anterior pituitary tissue both in physiologic conditions and when medium free T4 is raised. The findings may have relevance to the sick euthyroid thyroid syndrome in which free T4 may be increased in some patients who have decreased serum free T3.
Collapse
Affiliation(s)
- Y Halperin
- Department of Medicine, Montefiore Medical Center, Bronx, New York
| | | | | |
Collapse
|