1
|
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
|
2
|
Galton VA, Martinez ME, Dragon JA, St Germain DL, Hernandez A. The Intrinsic Activity of Thyroxine Is Critical for Survival and Growth and Regulates Gene Expression in Neonatal Liver. Thyroid 2021; 31:528-541. [PMID: 32791885 PMCID: PMC7994419 DOI: 10.1089/thy.2020.0508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Thyroxine (T4) is generally considered to be a prohormone that requires conversion to triiodothyronine (T3) to exert biological activity. Although evidence suggests that T4 has intrinsic activity, it is questionable if this activity has any physiological relevance. Methods: To answer this question, triple knockout (KO) mice (Triples) that cannot express the types 1 (D1) and 2 (D2) deiodinase and the Pax8 genes were generated. Thus, they lack a thyroid and cannot convert T4 to T3. Triples were injected on alternate days with either vehicle or physiological doses of T4, T3, or T3+T4 from postnatal days 2-14. They were euthanized at P15, and RNA-seq was employed to profile gene expression in the liver. In another experiment, Pax8KO mice were injected with T3, T4, or T4+T3, and growth rate and survival to P84 were determined. Results: The growth retardation of Triples was not improved by either T3 or T4 alone but was significantly improved by T4+T3. In the liver, T4 significantly regulated the expression of genes that were also regulated by T3, but the proportion of genes that were negatively regulated was higher in mice treated with T4 than in mice treated with T3. Treatment with T4+T3 identified genes that were regulated synergistically by T3 and T4, and genes that were regulated only by T4+T3. Analysis of these genes revealed enrichment in mechanisms related to cell proliferation and cholesterol physiology, suggesting a unique contribution of T4 to these biological functions. Pax8KO mice all survived to P84 when injected with T4 or T4+T3. However, survival rate with T3 was only 50% and 10% at 3.5 and 12 weeks of life, respectively. Conclusions: T4 has intrinsic activity in vivo and is critical for survival and growth. At a physiological level, T4 per se can upregulate or downregulate many T3 target genes in the neonatal liver. While most of these genes are also regulated by T3, subsets respond exclusively to T4 or demonstrate enhanced or normalized expression only in the presence of both hormones. These studies demonstrate for the first time a complex dependency on both T4 and T3 for normal mammalian growth and development.
Collapse
Affiliation(s)
- Valerie Anne Galton
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | | | - Julie A. Dragon
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | | | - Arturo Hernandez
- Maine Medical Center Research Institute, MaineHealth, Scarborough, Maine, USA
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| |
Collapse
|
3
|
Di Paolo V, Mangialardo C, Zacà C, Barberi M, Sereni E, Borini A, Centanni M, Coticchio G, Verga-Falzacappa C, Canipari R. Thyroid hormones T3 and T4 regulate human luteinized granulosa cells, counteracting apoptosis and promoting cell survival. J Endocrinol Invest 2020; 43:821-831. [PMID: 31894536 DOI: 10.1007/s40618-019-01169-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/19/2019] [Indexed: 01/10/2023]
Abstract
PURPOSE Fine and balanced regulation of cell proliferation and apoptosis are key to achieve ovarian follicle development from the primordial to the preovulatory stage and therefore assure female reproductive function. While gonadotropins are the major and most recognized regulators of follicle cell growth and function, other factors, both systemic and local, play equally important roles. This work is aimed at evaluating the effects of thyroid hormones (THs) on human granulosa luteinized (hGL) viability. METHODS Human GL cells derived from assisted reproduction treatments were exposed to T3 or T4. Cell viability was evaluated by MTT assay. Apoptosis was evaluated by the TUNEL assay and active caspase-3 staining. StAR, CYP19A1,Caspase-3, P53 and BAX mRNA were evaluated by real-time PCR. LC3-I/-II, AKT and pAKT were evaluated by western blot. RESULTS T3 and T4 promoted cell viability in a dose-dependent modality and modulate StAR and CYP19A1 expression. T3 and to a lesser extent T4 mitigated cell death induced by serum starvation by inhibition of caspase-3 activity and expression of P53 and BAX; and attenuate cell death experimentally induced by C2-ceramide. Cell death derived from starvation appeared to be involved in autophagic processes, as the levels of autophagic markers (LC3-II/LC3-I ratio) decreased when starved cells were exposed to T3 and T4. This effect was associated with an increase in pAkt levels. CONCLUSION From the present study, THs emerge as potent anti-apoptotic agents in hGL cells. This effect is achieved by inhibiting the apoptosis signalling pathway of BAX and caspase-3, while maintaining active the PI3K/AKT pathway.
Collapse
Affiliation(s)
- V Di Paolo
- DAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Via A. Scarpa 14, 00161, Rome, Italy
- Department of Haematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - C Mangialardo
- Department of Medical‑Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy
| | - C Zacà
- 9.Baby, Family and Fertility Center, Via Dante 15, 40125, Bologna, Italy
| | - M Barberi
- 9.Baby, Family and Fertility Center, Via Dante 15, 40125, Bologna, Italy
| | - E Sereni
- 9.Baby, Family and Fertility Center, Via Dante 15, 40125, Bologna, Italy
| | - A Borini
- 9.Baby, Family and Fertility Center, Via Dante 15, 40125, Bologna, Italy
| | - M Centanni
- Department of Medical‑Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy
| | - G Coticchio
- 9.Baby, Family and Fertility Center, Via Dante 15, 40125, Bologna, Italy
| | - C Verga-Falzacappa
- Department of Medical‑Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy
| | - R Canipari
- DAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Via A. Scarpa 14, 00161, Rome, Italy.
| |
Collapse
|
4
|
Canipari R, Mangialardo C, Di Paolo V, Alfei F, Ucci S, Russi V, Santaguida MG, Virili C, Segni M, Misiti S, Centanni M, Verga Falzacappa C. Thyroid hormones act as mitogenic and pro survival factors in rat ovarian follicles. J Endocrinol Invest 2019; 42:271-282. [PMID: 29934772 DOI: 10.1007/s40618-018-0912-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 06/11/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE Thyroid disorders are clinically associated with impaired fertility in women, and these abnormalities can be improved by restoring the euthyroid state. The exact mechanisms of thyroid effect on female fertility are not well known; however, it is conceivable that thyroid hormones (THs) might act on ovarian physiology via receptors in granulosa cells. This work is aimed at evaluating the effects of THs on non-tumoral granulosa cells and follicles. METHODS Freshly isolated rat ovarian follicles and granulosa cells were exposed to T3 or T4 (THs). Cell growth and viability were evaluated by cell counting and the MTT assay, respectively, follicle growth was evaluated by volume measurements. Apoptosis was evaluated by the TUNEL assay and active Caspase 3 staining. rGROV cells were exposed to T3, and apoptosis was induced by serum deprivation. Bcl2, Bcl-2-associated X protein (BAX), Akt and pAkt expression were evaluated by western blot. RESULTS T3 induced a 40% increase in follicle volume (after 7 days). This increase was presumably due to the observed decrease (33%) in the apoptotic rate of the granulosa cell population. Both T3 and T4 caused a dose-dependent increase in rat granulosa cell number and viability. In addition, THs decreased the cell apoptotic rate in a dose-dependent manner. In both conditions, T3 appeared to be more efficient. In rGROV cells, 100 nM T3 induced cell growth and, in the absence of growth factors, reduced cell apoptosis by 40%, downregulating Caspase 3 and BAX. This effect was associated with an increase in pAkt levels. The involvement of the PI3 K pathway was confirmed by the ability of the PI3 K specific inhibitor (LY-294,002) to abolish T3 pro-survival action. CONCLUSIONS THs influence cell survival of ovarian granulosa cells. This effect likely contributes to the TH-induced follicle volume increase.
Collapse
Affiliation(s)
- R Canipari
- DAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy
| | - C Mangialardo
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy
| | - V Di Paolo
- DAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy
- Department of Hematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - F Alfei
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy
| | - S Ucci
- Pasteur Institute of Rome, Rome, Italy
| | - V Russi
- Pasteur Institute of Rome, Rome, Italy
| | - M G Santaguida
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy
| | - C Virili
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy
| | - M Segni
- Department of Pediatrics and Pediatric Neuropsychiatry, Sapienza, University of Rome, Rome, Italy
| | - S Misiti
- Department of Experimental Medicine, Sapienza, University of Rome, Rome, Italy
| | - M Centanni
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy
| | - C Verga Falzacappa
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza, University of Rome, Rome, Italy.
| |
Collapse
|
5
|
Aguayo-Mazzucato C, Lee TB, Matzko M, DiIenno A, Rezanejad H, Ramadoss P, Scanlan T, Zavacki AM, Larsen PR, Hollenberg A, Colton C, Sharma A, Bonner-Weir S. T 3 Induces Both Markers of Maturation and Aging in Pancreatic β-Cells. Diabetes 2018; 67:1322-1331. [PMID: 29625991 PMCID: PMC6014556 DOI: 10.2337/db18-0030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/29/2018] [Indexed: 12/25/2022]
Abstract
Previously, we showed that thyroid hormone (TH) triiodothyronine (T3) enhanced β-cell functional maturation through induction of Mafa High levels of T3 have been linked to decreased life span in mammals and low levels to lengthened life span, suggesting a relationship between TH and aging. Here, we show that T3 increased p16Ink4a (a β-cell senescence marker and effector) mRNA in rodent and human β-cells. The kinetics of Mafa and p16Ink4a induction suggested both genes as targets of TH via TH receptors (THRs) binding to specific response elements. Using specific agonists CO23 and GC1, we showed that p16Ink4a expression was controlled by THRA and Mafa by THRB. Using chromatin immunoprecipitation and a transient transfection yielding biotinylated THRB1 or THRA isoforms to achieve specificity, we determined that THRA isoform bound to p16Ink4a , whereas THRB1 bound to Mafa but not to p16Ink4a On a cellular level, T3 treatment accelerated cell senescence as shown by increased number of β-cells with acidic β-galactosidase activity. Our data show that T3 can simultaneously induce both maturation (Mafa) and aging (p16Ink4a ) effectors and that these dichotomous effects are mediated through different THR isoforms. These findings may be important for further improving stem cell differentiation protocols to produce functional β-cells for replacement therapies in diabetes.
Collapse
Affiliation(s)
| | - Terence B Lee
- Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | | | - Amanda DiIenno
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | | | - Preeti Ramadoss
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Thomas Scanlan
- Department of Physiology & Pharmacology, Oregon Health & Science University, Portland, OR
| | - Ann Marie Zavacki
- Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - P Reed Larsen
- Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Anthony Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Clark Colton
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Arun Sharma
- Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | | |
Collapse
|
6
|
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: 11] [Impact Index Per Article: 1.4] [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
|
7
|
Gil-Ibáñez P, Belinchón MM, Morte B, Obregón MJ, Bernal J. Is the Intrinsic Genomic Activity of Thyroxine Relevant In Vivo? Effects on Gene Expression in Primary Cerebrocortical and Neuroblastoma Cells. Thyroid 2017; 27:1092-1098. [PMID: 28605984 DOI: 10.1089/thy.2017.0024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND The possibility that the intrinsic genomic activity of thyroxine (T4) is of physiological relevance has been frequently hypothesized. It might explain gene expression patterns in the brain found in type 2-deiodinase (Dio2)-deficient mice. These mice display normal expression of most thyroid hormone-dependent genes, despite decreased brain triiodothyronine (T3). METHODS The relative effects of T4 and T3 on gene expression were analyzed in mouse neuro-2a (N2a) cells stably expressing the thyroid hormone receptor α1, and in primary mouse cerebrocortical cells enriched in astrocytes or in neurons. Cortical cells were derived from Dio2-deficient mice to prevent conversion of T4 to T3. T4 and T3 were measured in the media at the beginning and end of incubation, and T4 and T3 antibodies were used to block T4 and T3 action. RESULTS In all cell types, T4 had intrinsic genomic activity. In N2a cells, T4 activity was higher on negative regulation (1/5th of T3 activity) than on positive regulation (1/40th of T3 activity). T4 activity on positive regulation was dependent on the cell context, and was higher in primary cells than in N2a cells. CONCLUSION T4 has intrinsic genomic activity. Positive regulation depends on the cell context, and primary cells appear much more sensitive than neuroblastoma cells. In all cells, negative regulation is more sensitive to T4 than positive regulation. These properties may explain the mostly normal gene expression in the brain of Dio2-deficient mice.
Collapse
Affiliation(s)
- Pilar Gil-Ibáñez
- 1 Instituto de Investigaciones Biomédicas , Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- 2 Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III , Madrid, Spain
| | - Mónica M Belinchón
- 1 Instituto de Investigaciones Biomédicas , Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- 2 Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III , Madrid, Spain
| | - Beatriz Morte
- 2 Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III , Madrid, Spain
| | - Maria Jesus Obregón
- 1 Instituto de Investigaciones Biomédicas , Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Bernal
- 1 Instituto de Investigaciones Biomédicas , Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- 2 Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III , Madrid, Spain
| |
Collapse
|
8
|
Aguayo-Mazzucato C, DiIenno A, Hollister-Lock J, Cahill C, Sharma A, Weir G, Colton C, Bonner-Weir S. MAFA and T3 Drive Maturation of Both Fetal Human Islets and Insulin-Producing Cells Differentiated From hESC. J Clin Endocrinol Metab 2015; 100. [PMID: 26207953 PMCID: PMC4596039 DOI: 10.1210/jc.2015-2632] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
CONTEXT Human embryonic stem cells (hESCs) differentiated toward β-cells and fetal human pancreatic islet cells resemble each other transcriptionally and are characterized by immaturity with a lack of glucose responsiveness, low levels of insulin content, and impaired proinsulin-to-insulin processing. However, their response to stimuli that promote functionality have not been compared. OBJECTIVE The objective of the study was to evaluate the effects of our previous strategies for functional maturation developed in rodents in these two human models of β-cell immaturity and compare their responses. Design, Settings, Participants, and Interventions: In proof-of-principle experiments using either adenoviral-mediated overexpression of V-Maf avian musculoaponeurotic fibrosarcoma oncogene homolog A (MAFA) or the physiologically driven path via thyroid hormone (T3) and human fetal islet-like cluster (ICC) functional maturity was evaluated. Then the effects of T3 were evaluated upon the functional maturation of hESCs differentiated toward β-cells. MAIN OUTCOME MEASURES Functional maturation was evaluated by the following parameters: glucose responsiveness, insulin content, expression of the mature β-cell transcription factor MAFA, and proinsulin-to-insulin processing. RESULTS ICCs responded positively to MAFA overexpression and T3 treatment as assessed by two different maturation parameters: increased insulin secretion at 16.8 mM glucose and increased proinsulin-to-insulin processing. In hESCs differentiated toward β-cells, T3 enhanced MAFA expression, increased insulin content (probably mediated by the increased MAFA), and increased insulin secretion at 16.8 mM glucose. CONCLUSION T3 is a useful in vitro stimulus to promote human β-cell maturation as shown in both human fetal ICCs and differentiated hESCs. The degree of maturation induced varied in the two models, possibly due to the different developmental status at the beginning of the study.
Collapse
Affiliation(s)
- Cristina Aguayo-Mazzucato
- Joslin Diabetes Center (C.A.-M., J.H.-L., C.Ca., A.S., G.W., S.B.-W.), Harvard Medical School, Boston, Massachusetts 02215; and Department of Chemical Engineering (A.D., C.Co.), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| | - Amanda DiIenno
- Joslin Diabetes Center (C.A.-M., J.H.-L., C.Ca., A.S., G.W., S.B.-W.), Harvard Medical School, Boston, Massachusetts 02215; and Department of Chemical Engineering (A.D., C.Co.), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| | - Jennifer Hollister-Lock
- Joslin Diabetes Center (C.A.-M., J.H.-L., C.Ca., A.S., G.W., S.B.-W.), Harvard Medical School, Boston, Massachusetts 02215; and Department of Chemical Engineering (A.D., C.Co.), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| | - Christopher Cahill
- Joslin Diabetes Center (C.A.-M., J.H.-L., C.Ca., A.S., G.W., S.B.-W.), Harvard Medical School, Boston, Massachusetts 02215; and Department of Chemical Engineering (A.D., C.Co.), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| | - Arun Sharma
- Joslin Diabetes Center (C.A.-M., J.H.-L., C.Ca., A.S., G.W., S.B.-W.), Harvard Medical School, Boston, Massachusetts 02215; and Department of Chemical Engineering (A.D., C.Co.), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| | - Gordon Weir
- Joslin Diabetes Center (C.A.-M., J.H.-L., C.Ca., A.S., G.W., S.B.-W.), Harvard Medical School, Boston, Massachusetts 02215; and Department of Chemical Engineering (A.D., C.Co.), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| | - Clark Colton
- Joslin Diabetes Center (C.A.-M., J.H.-L., C.Ca., A.S., G.W., S.B.-W.), Harvard Medical School, Boston, Massachusetts 02215; and Department of Chemical Engineering (A.D., C.Co.), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| | - Susan Bonner-Weir
- Joslin Diabetes Center (C.A.-M., J.H.-L., C.Ca., A.S., G.W., S.B.-W.), Harvard Medical School, Boston, Massachusetts 02215; and Department of Chemical Engineering (A.D., C.Co.), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| |
Collapse
|
9
|
Alshehri B, D'Souza DG, Lee JY, Petratos S, Richardson SJ. The diversity of mechanisms influenced by transthyretin in neurobiology: development, disease and endocrine disruption. J Neuroendocrinol 2015; 27:303-23. [PMID: 25737004 DOI: 10.1111/jne.12271] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 12/12/2022]
Abstract
Transthyretin (TTR) is a protein that binds and distributes thyroid hormones (THs). TTR synthesised in the liver is secreted into the bloodstream and distributes THs around the body, whereas TTR synthesised in the choroid plexus is involved in movement of thyroxine from the blood into the cerebrospinal fluid and the distribution of THs in the brain. This is important because an adequate amount of TH is required for normal development of the brain. Nevertheless, there has been heated debate on the role of TTR synthesised by the choroid plexus during the past 20 years. We present both sides of the debate and how they can be reconciled by the discovery of TH transporters. New roles for TTR have been suggested, including the promotion of neuroregeneration, protection against neurodegeneration, and involvement in schizophrenia, behaviour, memory and learning. Recently, TTR synthesis was revealed in neurones and peripheral Schwann cells. Thus, the synthesis of TTR in the central nervous system (CNS) is more extensive than previously considered and bolsters the hypothesis that TTR may play wide roles in neurobiological function. Given the high conservation of TTR structure, function and tissue specificity and timing of gene expression, this implies that TTR has a fundamental role, during development and in the adult, across vertebrates. An alarming number of 'unnatural' chemicals can bind to TTR, thus potentially interfering with its functions in the brain. One role of TTR is delivery of THs throughout the CNS. Reduced TH availability during brain development results in a reduced IQ. The combination of the newly discovered sites of TTR synthesis in the CNS, the increasing number of neurological diseases being associated with TTR, the newly discovered functions of TTR and the awareness of the chemicals that can interfere with TTR biology render this a timely review on TTR in neurobiology.
Collapse
Affiliation(s)
- B Alshehri
- School of Medical Sciences, RMIT University, Bundoora, VIC, Australia
| | | | | | | | | |
Collapse
|
10
|
Richardson SJ. Tweaking the structure to radically change the function: the evolution of transthyretin from 5-hydroxyisourate hydrolase to triiodothyronine distributor to thyroxine distributor. Front Endocrinol (Lausanne) 2014; 5:245. [PMID: 25717318 PMCID: PMC4324301 DOI: 10.3389/fendo.2014.00245] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/23/2014] [Indexed: 11/25/2022] Open
Abstract
Often, we elucidate evolutionary processes backwards, starting with eutherian mammals and gradually climbing down the evolutionary tree to those species who have survived since long before mammals evolved. This is also true for elucidating the evolution of specific proteins, in this case, the protein currently known as "transthyretin" (TTR). TTR was first described in eutherian mammals and was known as a thyroxine (T4) binding protein. However, mammals are the exception among vertebrates in respect to the function of TTR, as in teleost fish, amphibians, reptiles and birds TTR preferentially binds triiodothyronine (T3), which is the active form of thyroid hormone (TH). The TTR gene possibly arose as a duplication of the transthyretin-like protein (TLP) gene, around the stage of the agnathans. Some vertebrate species have both the TTR and TLP genes, while others have "lost" the TLP gene. TLP genes have been found in all kingdoms. The TLPs analyzed to date do not bind THs or their analogs, but are enzymes involved in uric acid metabolism; specifically, they are 5-hydroxyisourate hydrolases. A Salmonella TLP knock-out strain demonstrated that TLP was essential for the bacteria's survival in the high uric acid environment of the chicken alimentary tract. Many other TLPs are yet to be characterized for their function although several have been confirmed as 5-hydroxyisourate hydrolases. This review describes the evolution of TLP/TTR and how subtle changes in gene structure or amino acid substitution can drastically change the function of this protein, without altering its overall 3D conformation.
Collapse
Affiliation(s)
- Samantha J. Richardson
- School of Medical Sciences, RMIT University, Bundoora, VIC, Australia
- *Correspondence: Samantha J. Richardson, School of Medical Sciences, RMIT University, PO Box 71 Bundoora, VIC 3083, Australia e-mail:
| |
Collapse
|
11
|
Aguayo-Mazzucato C, Zavacki AM, Marinelarena A, Hollister-Lock J, El Khattabi I, Marsili A, Weir GC, Sharma A, Larsen PR, Bonner-Weir S. Thyroid hormone promotes postnatal rat pancreatic β-cell development and glucose-responsive insulin secretion through MAFA. Diabetes 2013; 62:1569-80. [PMID: 23305647 PMCID: PMC3636623 DOI: 10.2337/db12-0849] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neonatal β cells do not secrete glucose-responsive insulin and are considered immature. We previously showed the transcription factor MAFA is key for the functional maturation of β cells, but the physiological regulators of this process are unknown. Here we show that postnatal rat β cells express thyroid hormone (TH) receptor isoforms and deiodinases in an age-dependent pattern as glucose responsiveness develops. In vivo neonatal triiodothyronine supplementation and TH inhibition, respectively, accelerated and delayed metabolic development. In vitro exposure of immature islets to triiodothyronine enhanced the expression of Mafa, the secretion of glucose-responsive insulin, and the proportion of responsive cells, all of which are effects that were abolished in the presence of dominant-negative Mafa. Using chromatin immunoprecipitation and electrophoretic mobility shift assay, we show that TH has a direct receptor-ligand interaction with the Mafa promoter and, using a luciferase reporter, that this interaction was functional. Thus, TH can be considered a physiological regulator of functional maturation of β cells via its induction of Mafa.
Collapse
Affiliation(s)
- Cristina Aguayo-Mazzucato
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Ann Marie Zavacki
- Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alejandra Marinelarena
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Hollister-Lock
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Ilham El Khattabi
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Alessandro Marsili
- Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gordon C. Weir
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Arun Sharma
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - P. Reed Larsen
- Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Susan Bonner-Weir
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Susan Bonner-Weir,
| |
Collapse
|
12
|
Freitas J, Cano P, Craig-Veit C, Goodson ML, David Furlow J, Murk AJ. Detection of thyroid hormone receptor disruptors by a novel stable in vitro reporter gene assay. Toxicol In Vitro 2011; 25:257-66. [DOI: 10.1016/j.tiv.2010.08.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 07/29/2010] [Accepted: 08/16/2010] [Indexed: 10/19/2022]
|
13
|
Regulation of the cardiomyocyte population in the developing heart. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2010; 106:289-99. [PMID: 21147149 DOI: 10.1016/j.pbiomolbio.2010.11.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/16/2010] [Accepted: 11/26/2010] [Indexed: 11/21/2022]
Abstract
During fetal life the myocardium expands through replication of cardiomyocytes. In sheep, cardiomyocytes begin the process of becoming terminally differentiated at about 100 gestation days out of 145 days term. In this final step of development, cardiomyocytes become binucleated and stop dividing. The number of cells at birth is important in determining the number of cardiomyocytes for life. Therefore, the regulation of cardiomyocyte growth in the womb is critical to long term disease outcome. Growth factors that stimulate proliferation of fetal cardiomyocytes include angiotensin II, cortisol and insulin-like growth factor-1. Increased ventricular wall stress leads to short term increases in proliferation but longer-term loss of cardiomyocyte generative capacity. Two normally circulating hormones have been identified that suppress proliferation: atrial natriuretic peptide (ANP) and tri-iodo-L-thyronine (T₃). Atrial natriuretic peptide signals through the NPRA receptor that serves as a guanylate cyclase and signals through cGMP. ANP powerfully suppresses mitotic activity in cardiomyocytes in the presence of angiotensin II in culture. Addition of a cGMP analog has the same effect as ANP. ANP suppresses both the extracellular receptor kinases and the phosphoinositol-3 kinase pathways. T₃ also suppresses increased mitotic activity of stimulated cardiomyocytes but does so by increasing the cell cycle suppressant, p21, and decreasing the cell cycle activator, cyclin D1.
Collapse
|
14
|
Williams AJ, O'Shea PJ, Williams GR. Complex interactions between thyroid hormone and fibroblast growth factor signalling. Curr Opin Endocrinol Diabetes Obes 2007; 14:410-5. [PMID: 17940472 DOI: 10.1097/med.0b013e3282eee92c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Thyroid hormone and fibroblast growth factors are critically important for normal development. Recent evidence points to complex interactions between thyroid hormone and fibroblast growth factors that regulate cell proliferation and differentiation. We discuss mechanisms of thyroid hormone and fibroblast growth factor action, and identify downstream signalling responses that offer opportunities for regulatory crosstalk. RECENT FINDINGS Thyroid hormone action is mediated by nuclear receptors that regulate gene expression in response to thyroid hormone. Recent studies have shown thyroid hormone also acts at the cell membrane via the alpha(V)beta(3) integrin receptor and these actions also communicate with nuclear responses to thyroid hormone. Fibroblast growth factors act via receptor tyrosine kinases to stimulate second messenger pathways that also communicate with nuclear events. Several common pathways, including mitogen-activated protein kinase, phosphatidylinositol 3-kinase, and signal transducer and activator of transcription signalling, are activated by thyroid hormone and fibroblast growth factor, and may act as points of convergence for interaction in tissues, such as bone, central nervous system and heart, as well as in the extra-cellular matrix and during angiogenesis. SUMMARY Although there is convincing evidence that thyroid hormone and fibroblast growth factors interact widely, little is known about molecular mechanisms that determine this interplay. Future research in this expanding field may result in identification of new pharmacological targets for manipulation of cell proliferation and differentiation.
Collapse
Affiliation(s)
- Allan J Williams
- Molecular Endocrinology Group, Division of Medicine & MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, UK
| | | | | |
Collapse
|
15
|
Jung KK, Kim SY, Kim TG, Kang JH, Kang SY, Cho JY, Kim SH. Differential regulation of thyroid hormone receptor-mediated function by endocrine disruptors. Arch Pharm Res 2007; 30:616-23. [PMID: 17615682 DOI: 10.1007/bf02977657] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
It is well known that endocrine disruptors (EDs) act as anti-estrogenic agents and affect the function of reproductive organ. EDs are also thought to affect thyroid hormone (TH) system which is important for biological functions such as growth, development and metabolism. However, it is still not clear how EDs are able to regulate TH receptor (TR)-mediated functions. In this study, therefore, the modulatory effects of representative EDs such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), polychlorinated biphenyl (Aroclor 1254) and bisphenol A (BPA) were examined using TR-expressing GH3 cells (a rat pituitary gland epithelial tumor cell line) activated by triiodothyronine (T3). EDs tested significantly blocked T3 binding to TR in a dose-dependent manner. Biochemical characterization by Scatchard and Lineweaver-Burk plot analyses indicated that TCDD and aroclor 1254 bound to TH receptors in a competitive inhibitory manner, whereas BPA bound to TH receptors in a non-competitive pattern. The different inhibitory mode of action by EDs was also found in regulating TR-mediated production of prolactin (PRL). Aroclor 1254 exposure for 48 h enhanced T3-mediated PRL production, but BPA down-regulated. These results suggest that the EDs (TCDD, Aroclor 1254 and BPA) could differentially bind to TR and distinctly regulate the action of TR function, even though EDs are structurally similar.
Collapse
Affiliation(s)
- Ki Kyung Jung
- Department of Pharmacology, Biochemical Pharmacology Team, National Institute of Toxicological Research, Seoul 122-704, Korea
| | | | | | | | | | | | | |
Collapse
|
16
|
Sandler B, Webb P, Apriletti JW, Huber BR, Togashi M, Cunha Lima ST, Juric S, Nilsson S, Wagner R, Fletterick RJ, Baxter JD. Thyroxine-thyroid hormone receptor interactions. J Biol Chem 2004; 279:55801-8. [PMID: 15466465 DOI: 10.1074/jbc.m410124200] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Thyroid hormone (TH) actions are mediated by nuclear receptors (TRs alpha and beta) that bind triiodothyronine (T(3), 3,5,3'-triiodo-l-thyronine) with high affinity, and its precursor thyroxine (T(4), 3,5,3',5'-tetraiodo-l-thyronine) with lower affinity. T(4) contains a bulky 5' iodine group absent from T(3). Because T(3) is buried in the core of the ligand binding domain (LBD), we have predicted that TH analogues with 5' substituents should fit poorly into the ligand binding pocket and perhaps behave as antagonists. We therefore examined how T(4) affects TR activity and conformation. We obtained several lines of evidence (ligand dissociation kinetics, migration on hydrophobic interaction columns, and non-denaturing gels) that TR-T(4) complexes adopt a conformation that differs from TR-T(3) complexes in solution. Nonetheless, T(4) behaves as an agonist in vitro (in effects on coregulator and DNA binding) and in cells, when conversion to T(3) does not contribute to agonist activity. We determined x-ray crystal structures of the TRbeta LBD in complex with T(3) and T(4) at 2.5-A and 3.1-A resolution. Comparison of the structures reveals that TRbeta accommodates T(4) through subtle alterations in the loop connecting helices 11 and 12 and amino acid side chains in the pocket, which, together, enlarge a niche that permits helix 12 to pack over the 5' iodine and complete the coactivator binding surface. While T(3) is the major active TH, our results suggest that T(4) could activate nuclear TRs at appropriate concentrations. The ability of TR to adapt to the 5' extension should be considered in TR ligand design.
Collapse
Affiliation(s)
- Ben Sandler
- Metabolic Research Unit and Diabetes Center, School of Medicine, University of California-San Francisco, 513 Parnassus Avenue, San Francisco, CA 94122-0540, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Shepherdley CA, Daniels CB, Orgeig S, Richardson SJ, Evans BK, Darras VM. Glucocorticoids, thyroid hormones, and iodothyronine deiodinases in embryonic saltwater crocodiles. Am J Physiol Regul Integr Comp Physiol 2002; 283:R1155-63. [PMID: 12376409 DOI: 10.1152/ajpregu.00015.2002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated the relationship between glucocorticoids, thyroid hormones, and outer ring and inner ring deiodinases (ORD and IRD) during embryonic development in the saltwater crocodile (Crocodylus porosus). We treated the embryos with the synthetic glucocorticoid dexamethasone (Dex), 3,3',5-triiodothyronine (T(3)), and a combination of these two hormones (Dex + T(3)). The effects of these treatments were specific in different tissues and at different stages of development and also brought about changes in plasma concentrations of free thyroid hormones and corticosterone. Administration of Dex to crocodile eggs resulted in a decrease in 3,3',5,5'-tetraiodothyronine (T(4)) ORD activities in liver and kidney microsomes, and a decrease in the high-K(m) rT(3) ORD activity in kidney microsomes, on day 60 of incubation. Dex treatment increased the T(4) ORD activity in liver microsomes, but not kidney microsomes, on day 75 of incubation. Dex administration decreased T(3) IRD activity in liver microsomes. However, this decrease did not change plasma-free T(3) concentrations, which suggests that free thyroid hormone levels are likely to be tightly regulated during development.
Collapse
|
18
|
Shepherdley CA, Richardson SJ, Evans BK, Kühn ER, Darras VM. Thyroid hormone deiodinases during embryonic development of the saltwater crocodile (Crocodylus porosus). Gen Comp Endocrinol 2002; 126:153-64. [PMID: 12030771 DOI: 10.1006/gcen.2002.7786] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
All tissues of the embryonic saltwater crocodile (Crocodylus porosus) gradually increased in weight during development except for lung tissue, which had a peak weight of 1.09 g at day 67, thereafter decreasing in weight. The brain was a relatively large organ. Deiodinase activities in liver, kidney, lung, heart, gut, and brain from day 29 to day 77 of development of the saltwater crocodile were investigated. High-K(m) reverse triiodothyronine (rT(3)) outer ring deiodination (ORD) activity was present in all tissues except the brain. Activity ranged from 559 +/- 51.3 pmol rT(3) deiodinated/mg protein/min in the liver at day 77 to below 10 pmol rT(3) deiodinated/mg protein/min in gut, lung, and heart tissue. rT(3) ORD increased during development in the liver and kidney but decreased in the gut and lung. Activity in the heart was very low (less than 2 pmol rT(3) deiodinated/mg protein/min) and did not change during development. Low-K(m) thyroxine (T(4)) ORD in liver and kidney tissue had peaks of activity around day 49 of incubation (0.52 and 0.09 fmol T(4) deiodinated/mg protein/min, respectively). After day 49, T(4) ORD activity in these tissues decreased. T(4) ORD activity in gut, lung, and heart was very low (less than 0.04 fmol T(4) deiodinated/mg protein/min), with activity in lung increasing slightly during the rest of development. T(4) ORD activity in the brain increased toward day 77 (0.14 +/- 0.03 fmol T(4) deiodinated/mg protein/min), illustrating its importance in local triiodothyronine (T(3)) production during brain development. T(3) inner ring deiodination activity was present only in the embryonic liver and peaked at day 49 (10.1 fmol T(3) deiodinated/mg protein/min), after which activity decreased.
Collapse
|
19
|
Shepherdley CA, Richardson SJ, Evans BK, Kühn ER, Darras VM. Characterization of outer ring iodothyronine deiodinases in tissues of the saltwater crocodile (Crocodylus porosus). Gen Comp Endocrinol 2002; 125:387-98. [PMID: 11884083 DOI: 10.1006/gcen.2001.7764] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The distribution and characterization of outer ring deiodination (ORD) using reverse triiodothyronine (rT3) and thyroxine (T4) as substrates is reported in microsomes of liver, kidney, lung, heart, gut, and brain tissues from juvenile saltwater crocodiles (Crocodylus porosus). In lung and heart only small amounts of rT3 ORD and T4 ORD were detected, while in brain only a small amount of T4 ORD was detected. More detailed characterization studies could be performed on liver, kidney, and gut microsomes. Reverse T3 outer ring deiodination (rT3 ORD) was the predominant activity in liver and kidney microsomes. The properties of crocodile liver and kidney rT3 ORD, such as preference for rT3 as substrate, a dithiothreitol (DTT) requirement of 10 mM, inhibition by propylthiouracil (PTU), and Michaelis-Menten (Km) constant in the micromolar range, correspond to the properties previously reported for a type I deiodinase. The temperature optimum for rT3 ORD was between 30 and 35 degrees. There was also rT3 ORD activity in gut microsomes, along with what appeared to be a type II-like, low-Km deiodinase with a substrate preference for T4. There was also a small amount of T4 ORD activity in liver and kidney microsomes. Liver T4 ORD, like a type II deiodinase, had a preference for T4 as substrate at low substrate concentrations and a DTT requirement of 15 mM and was insensitive to PTU. However, at high substrate concentrations the predominant activity was of the type I deiodinase nature. T4 ORD in liver had an optimal incubation temperature of 30 to 35 degrees. Gut microsomal T4 ORD was also type II-like at low substrate concentrations and type I-like at high substrate concentrations. Gut T4 ORD had an optimal incubation temperature of 25 to 30 degrees and a DTT requirement of 20 mM DTT. Kidney microsomal T4 ORD had the same optimal temperature and DTT requirement as that in gut microsomes; however, there was no competition by low substrate concentrations. These results suggest that ORD in juvenile saltwater crocodile kidney is most likely exclusively catalyzed by a type I-like deiodinase. Liver and gut ORD, in contrast, is catalyzed by two enzymes, with a predominance of a type I-like deiodinase in liver and a type II-like deiodinase in gut. Low-Km T3 IRD activity could not be detected in any tissues of the juvenile saltwater crocodile.
Collapse
Affiliation(s)
- Caroline A Shepherdley
- Laboratory of Comparative Endocrinology, Zoological Institute, K.U. Leuven, B-3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|
20
|
Mercier G, Turque N, Schumacher M. Rapid effects of triiodothyronine on immediate-early gene expression in Schwann cells. Glia 2001; 35:81-9. [PMID: 11460264 DOI: 10.1002/glia.1073] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the peripheral nervous system, triiodothyronine (T3) plays an important role in the development and regeneration of nerve fibers and in myelin formation. However, the target genes of T3 in peripheral nerves remain to be identified. We investigated whether T3 activated genes of transcription factors in Schwann cells. Expression of egr-1 (krox-24), egr-2 (krox-20), egr-3, c-jun, junB, c-fos, fosB, fra-1, fra-2, and CREB genes was analyzed by reverse transcription-polymerase chain reaction (RT-PCR) in Schwann cells isolated from neonatal rat sciatic nerves and in the cell lines MSC-80 (mouse Schwann cells), NIH-3T3 (mouse fibroblasts), and CHO (Chinese hamster ovary cells). Some of these transcription factors have been shown to be involved in Schwann cell differentiation. T3 triggered a rapid (15-30 min), transient (1-2-h) and strong (6- to 15-fold) stimulation of Egr-1, Egr-2, Egr-3, Jun B, c-Fos, and Fos B mRNA expression in Schwann cells. In contrast, expression of c-Jun, Fra-1, Fra-2, and CREB mRNA was not affected by T3. The stimulatory effects of T3 could be abolished by adding actinomycin D. T3 triggered the same pattern of gene stimulation in the mouse Schwann cell line MSC80, but not in the NIH-3T3 and CHO cell lines. Serum activated all the genes that responded to T3 and in addition fra-1 and fra-2, but not c-jun and CREB. Immunoblotting showed that the increase in Egr-1 and c-Fos mRNA levels was accompanied by an increase in the corresponding proteins. In addition, shifts of the protein bands indicated a posttranslational modification of the two proteins. These effects of T3 are likely to be mediated by the intracellular T3 receptor, as the D-isomer RT3 and T0, which do not bind to T3 receptors, proved ineffective. The present data suggested that T3 may regulate Schwann cell functions and differentiation by transiently activating the expression of specific transcription factors.
Collapse
Affiliation(s)
- G Mercier
- U488 INSERM, Kremlin-Bicêtre, France.
| | | | | |
Collapse
|
21
|
Prapunpoj P, Yamauchi K, Nishiyama N, Richardson SJ, Schreiber G. Evolution of structure, ontogeny of gene expression, and function of Xenopus laevis transthyretin. Am J Physiol Regul Integr Comp Physiol 2000; 279:R2026-41. [PMID: 11080066 DOI: 10.1152/ajpregu.2000.279.6.r2026] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Xenopus laevis transthyretin (xTTR) cDNA was cloned and sequenced. The derived amino acid sequence was very similar to those of other vertebrate transthyretins (TTR). TTR gene expression was observed during metamorphosis in X. laevis tadpole liver but not in tadpole brain nor adult liver. Recombinant xTTR was synthesized in Pichia pastoris and identified by amino acid sequence, subunit molecular mass, tetramer formation, and binding to retinol-binding protein. Contrary to mammalian xTTRs, the affinity of xTTR was higher for L-triiodothyronine than for L-thyroxine. The regions of the TTR genes coding for the NH(2)-terminal sections of the polypeptide chains of TTR seem to have evolved by stepwise shifts of mRNA splicing sites between exons 1 and 2, resulting in shorter and more hydrophilic NH(2) termini. This may be one molecular mechanism of positive Darwinian evolution. Open reading frames with xTTR-like sequences in the genomes of C. elegans and several microorganisms suggested evolution of the TTR gene from ancestor TTR gene-like "DNA modules." Increasing preference for binding of L-thyroxine over L-triiodothyronine may be associated with evolving tissue-specific regulation of thyroid hormone action by deiodination.
Collapse
Affiliation(s)
- P Prapunpoj
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|
22
|
Zhu XG, Hanover JA, Hager GL, Cheng SY. Hormone-induced translocation of thyroid hormone receptors in living cells visualized using a receptor green fluorescent protein chimera. J Biol Chem 1998; 273:27058-63. [PMID: 9765220 DOI: 10.1074/jbc.273.42.27058] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Thyroid hormone nuclear receptors (TRs) are ligand-dependent transcription factors that regulate growth, differentiation, and development. To understand the role of the hormone, 3,3', 5-triiodo-L-thyronine (T3), in the nuclear translocation and targeting of TRs to the regulatory sites in chromatin, we appended green fluorescent protein (GFP) to the human TR subtype beta1 (TRbeta1). The fusion of GFP to the amino terminus of TRbeta1 protein did not alter T3 binding or transcriptional activities of the receptor. The subcellular localization of GFP-TRbeta1 in living cells was visualized by laser-scanning confocal microscopy. In the presence of T3, the expressed GFP-TRbeta1 was predominately localized in the nucleus, exhibiting a nuclear/cytoplasmic ratio of approximately 5.5. No GFP-TRbeta1 was detected in the nucleolus. In the absence of T3, more GFP-TRbeta1 was present in the cytoplasm, exhibiting a nuclear/cytoplasmic ratio of approximately 1.5. In these cells, cytoplasmic GFP-TRbeta1 could be induced to enter the nucleus by T3. The T3-induced translocation was blocked when Lys184-Arg185 in domain D of TRbeta1 was mutated to Ala184-Ala185. Furthermore, the inability of the mutant TR to translocate to the nucleus correlated with the loss of most of its transcriptional activity. These results suggest that TR functions may, in part, be regulated by T3-induced nuclear entry.
Collapse
Affiliation(s)
- X G Zhu
- Laboratory of Molecular Biology, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
23
|
Moreno M, Lombardi A, Lombardi P, Goglia F, Lanni A. Effect of 3,5-diiodo-L-thyronine on thyroid stimulating hormone and growth hormone serum levels in hypothyroid rats. Life Sci 1998; 62:2369-77. [PMID: 9651103 DOI: 10.1016/s0024-3205(98)00219-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have investigated the biological effects of physiological doses of 3,5-diiodo-L-thyronine (3,5-T2) and 3,3'-diiodo-L-thyronine (3,3'-T2) (at doses from 2.5 to 10 microg/100 g BW) on serum TSH and GH levels in rats made hypothyroid by propylthiouracil and iopanoic acid administration. In such animals deiodinase activities were inhibited and thyroid hormones serum levels strongly reduced. The effects of T2s were compared with those elicited by 3,5,3'-triiodo-L-thyronine (T3) (2.5 microg/100 g BW).The serum TSH level was much greater in hypothyroid rats than in euthyroid ones. T3 administration suppressed TSH by 88% compared to control (i.e, the level in hypothyroid rats); it thus reached a value not significantly different from that seen in the euthyroid rats. 3,5-T2 produced a similar effect, suppressing the TSH level by about 75% compared to control; it thus reached values not significantly different from those of the euthyroid and T3-treated rats. By contrast, 3,3'-T2 had no effect on TSH, whatever the dose. The serum GH level was much lower in hypothyroid rats than in euthyroid ones. T3 administration increased the GH level by about 5-fold, restoring it to the value seen in euthyroid rats. 3,5-T2-treated hypothyroid rats, at all the doses used (from 2.5 to 10 microg/100 g BW), showed increased serum GH levels: at a dose of 10 microg/100 g BW the level reached a value about 5-fold higher than that in hypothyroid rats. This value was not significantly different from those of euthyroid and T3-treated rats. 3,3'-T2 did not affect GH levels whatever the dose. Thus, 3,5-T2 (but not 3,3'-T2) seems to mimic the effects of T3 on serum TSH and GH levels in rats.
Collapse
Affiliation(s)
- M Moreno
- Dipartimento di Chimica, Facoltà di Scienze, Università degli Studi di Salerno, Italy
| | | | | | | | | |
Collapse
|
24
|
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
|
25
|
Romani A, Marfella C, Lakshmanan M. Mobilization of Mg2+ from rat heart and liver mitochondria following the interaction of thyroid hormone with the adenine nucleotide translocase. Thyroid 1996; 6:513-9. [PMID: 8936681 DOI: 10.1089/thy.1996.6.513] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The in vitro addition of thyroid hormone to isolated rat heart or liver mitochondria induces the extrusion of approximately 2-4 nmol Mg2+/mg protein from both mitochondria preparations. The mobilization of Mg2+ is not accompanied by extrusion of matrix ATP or K+, or by mitochondria swelling, thus excluding that the phenomenon occurs through the nonspecific opening of the mitochondrial permeability transition pore. Moreover, the Mg2+ extrusion is completely prevented by bongkrekic acid, a membrane-permeant inhibitor of the adenine nucleotide translocase (AdNT), and by cyclosporine, which has also been reported to inhibit AdNT in a bongkrekate-like manner, operating at the matrix site of the translocase. By contrast, atractyloside, another specific inhibitor of AdNT that operates at the cytosolic site of the AdNT, only partially affects the Mg2+ mobilization (< 30% inhibition). These findings and the binding of 125I-labeled thyroid hormone to both the dimeric and monomeric moiety of AdNT support the hypothesis that AdNT can operate as a specific receptor for thyroid hormone in the mitochondria, and suggest that thyroid hormone operates at the matrix site of the translocase. In addition, these observations may imply that some of the so called "nongenomic effects" exerted by thyroid hormone on mitochondrial metabolism could occur through changes in the matrix content of Mg2+.
Collapse
Affiliation(s)
- A Romani
- Department of Physiology and Biophysis, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106-4970, USA
| | | | | |
Collapse
|
26
|
Thyagaraja BS, Masler EP, Kelly TJ, Borkovec AB. Thyroxine-induced changes in ovarian protein and ecdysteroid levels in the silkworm, Bombyx mori L.: Effect on ovarian maturation and egg production. ACTA ACUST UNITED AC 1993. [DOI: 10.1016/0300-9629(93)90312-r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
27
|
Lakshmanan M, Goncalves E, Pontecorvi A, Robbins J. Differential effect of a new thyromimetic on triiodothyronine transport into myoblasts and hepatoma and neuroblastoma cells. BIOCHIMICA ET BIOPHYSICA ACTA 1992; 1133:213-7. [PMID: 1731960 DOI: 10.1016/0167-4889(92)90071-i] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
3,5-Dibromo-3'-pyridazinone-L-thyronine (L-94901), a member of a novel class of thyromimetics, reduces cholesterol plasma levels with little effect on cardiac function in rats. Because receptor binding of L-94901 in isolated heart and liver nuclei is similar but binding in liver nuclei in vivo was 50-fold higher than in cardiac nuclei, we studied its effect on triiodothyronine (T3) transport across the plasma membrane of myoblasts, hepatoma cells and neuroblasts. Previously, we had demonstrated saturable, stereospecific and energy dependent transport of T3 into the three cell lines. After equilibrium of intact cells with hormone, nuclear binding of T3 was decreased by L-94901 in all three cell lines. While whole cell uptake and whole cell binding of T3 was only slightly affected by L-94901, kinetic analysis of the initial rate of uptake showed uncompetitive or noncompetitive inhibition and a differential decrease in Vmax. Furthermore, the Ki for the liver and brain derived cells was 10-fold lower than for the muscle derived cells. This effect on the plasma membrane transport of T3 may explain the differential effect reported in the intact animal.
Collapse
Affiliation(s)
- M Lakshmanan
- Clinical Endocrinology Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892
| | | | | | | |
Collapse
|
28
|
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
|
29
|
Sandhu GS, Steele R, Gonnella NC. Effect of L-thyroxine (LT4) and D-thyroxine (DT4) on cardiac function and high-energy phosphate metabolism: a 31P NMR study. Magn Reson Med 1991; 18:237-43. [PMID: 2062236 DOI: 10.1002/mrm.1910180125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
31P NMR spectroscopy was used to monitor the cardiac energy metabolism in hypothyroid rat hearts. Differential alterations in phosphocreatine and inorganic phosphate levels were observed upon treatment of hypothyroid animals with DT4 and LT4, while both agents were equipotent in reducing cholesterol. These results show potential for NMR spectroscopy as a technique to determine therapeutic selectivity.
Collapse
Affiliation(s)
- G S Sandhu
- Research Department Pharma Division, CIBA-GEIGY Corporation, Summit, New Jersey 07901
| | | | | |
Collapse
|
30
|
Abstract
Cellular binding proteins of thyroid hormones are present in the cell nucleus, cytosol, cell membrane, and mitochondria. While nuclear binding is proven to mediate hormone action, the exact roles of the other binding sites remain to be established. Nuclear receptor associates with DNA, core histone, and nuclear matrix and preferentially distributes in transcriptionally active chromatin due to interaction with H1 histone. Of particular importance is the binding of nuclear receptor to specific DNA sequences of target genes, termed thyroid-responsive elements. The binding is stabilized by non-receptor nuclear protein. Upon binding thyroid hormone, nuclear receptor is activated through alterations in the steric configuration, leading to changes in the rate of transcription of the target genes. Multiple nuclear receptor forms exist with likely distinct functional roles. Cytosolic thyroid hormone binding proteins are also heterogeneous. One form is under the control of cell metabolism (NADP and NADPH) and it may have a role in transport of the hormone to mitochondria and nucleus. Membrane-linked thyroid hormone binding proteins may have dual functional roles: one is to mediate hormone action and the other is to support active uptake of hormones by cells. Mitochondrial function may be regulated by thyroid hormone through mitochondrial binding sites in cooperation with nuclear receptor-mediated pathway. Further studies are required to elucidate the exact functional roles of non nuclear thyroid hormone binding proteins.
Collapse
Affiliation(s)
- K Ichikawa
- Department of Geriatrics, Shinshu University School of Medicine, Nagano, Japan
| | | |
Collapse
|
31
|
GLASS CHRISTOPHERK, ROSENFELD MICHAELG. Regulation of gene transcription by thyroid hormones and retinoic acid. ACTA ACUST UNITED AC 1991. [DOI: 10.1016/b978-0-444-81382-4.50013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
32
|
Darby IA, Bouhnik J, Coezy ED, Corvol P. Thyroid hormone receptors and stimulation of angiotensinogen production in HepG2 cells. IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY : JOURNAL OF THE TISSUE CULTURE ASSOCIATION 1991; 27:21-4. [PMID: 1849506 DOI: 10.1007/bf02630890] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Binding characteristics and effects of 3,5,-3'-triiodo-L-thyronine (T3) on angiotensinogen production in HepG2 were studied in serum-free medium. Binding was performed on intact cells and on partially purified isolated nuclei using [125I]T3. Scatchard plots revealed one class of high affinity binding sites with a Kd of approximately 80 pmol/liter. Calculation of maximum binding showed that HepG2 possess approximately 1000 binding sites per cell. Unlabeled T3 and T4 competed for binding sites on intact HepG2 with 50% inhibition of [125I] T3 binding at approximately 3.0 and 38.0 pmol/liter, respectively. The HepG2 showed a dose-dependent increase in angiotensinogen production in serum-free medium which was maximal at 10(-5) mol/liter (two-fold increase/10(6) cells/24 h) and had an EC50 of approximately 5.0 x 10(-8) mol/liter. T3 also produced after 24 h a dose-dependent increase in DNA highly correlated with T3 applied (r = 0.88, P less than 0.01). In conclusion, this study shows that HepG2 possess specific high affinity binding sites for T3 and that T3 stimulates angiotensinogen production and DNA synthesis in these cells.
Collapse
|
33
|
Glass CK, Holloway JM. Regulation of gene expression by the thyroid hormone receptor. BIOCHIMICA ET BIOPHYSICA ACTA 1990; 1032:157-76. [PMID: 2261492 DOI: 10.1016/0304-419x(90)90002-i] [Citation(s) in RCA: 33] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- C K Glass
- Eukaryotic Regulatory Biology Program Center for Molecular Genetics, University of California, San Diego School of Medicine, La Jolla 92093
| | | |
Collapse
|
34
|
Ichikawa K, Hashizume K, Furuta S, Osumi T, Miyamoto T, Yamauchi K, Takeda T, Yamada T. Human c-erb A protein expressed in Escherichia coli: changes in hydrophobicity upon thyroid hormone binding. Mol Cell Endocrinol 1990; 70:175-84. [PMID: 2161365 DOI: 10.1016/0303-7207(90)90157-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The human c-erb A beta gene sequence was inserted in an Escherichia coli expression vector plasmid. The E. coli cells transformed with this plasmid produced proteins with molecular masses of 52 and 50 kDa. These products bound 3,5,3'-triiodo-L-thyronine (T3) with an affinity constant of 4.3 x 10(9) liter/mol. The order of affinity for iodothyronine analogs was triiodothyroacetic acid greater than T3 greater than 3,5,3'-triiodo-D-thyronine greater than L-thyroxine. Affinity labeling experiments showed that the 50 kDa protein was covalently labeled with [125I]T3, and this was competed by triiodothyroacetic acid, T3, and L-thyroxine (from potent to weaker competitor). The c-erb A protein bound to calf thymus DNA-cellulose and the binding was inhibited by 0.3 M KCl or 10 mM pyridoxal 5'-phosphate. Aqueous two-phase partitioning studies showed that the c-erb A product became less hydrophobic upon T3 or triiodothyroacetic acid binding. The same finding was obtained when T3 bound to partially purified rat liver nuclear thyroid hormone receptor. However, thyroxine binding globulin became more hydrophobic upon T3 binding. Since the T3 molecule partitioned preferentially into the upper polyethylene glycol-rich phase, the alteration of partitioning behavior of thyroxine binding globulin was explained by a simple additive effect of T3. In contrast, the alteration of partitioning behavior of the c-erb A product or receptor reflected a conformational transition upon T3 binding. The c-erb A protein expressed in E. coli showed various characteristics similar to classical thyroid hormone receptor and may be useful in studying the structure and function of the thyroid hormone receptor.
Collapse
Affiliation(s)
- K Ichikawa
- Department of Gerontology, Endocrinology, and Metabolism, Shinshu University School of Medicine, Matsumoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Samuels HH, Casanova J, Copp RP, Janocko L, Raaka BM, Sahnoun H, Yaffe BM. Thyroid hormone receptors and action: the 5'-flanking region of the rat growth hormone gene can mediate regulated gene expression. Endocr Res 1989; 15:495-545. [PMID: 2699452 DOI: 10.3109/07435808909036350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- H H Samuels
- Department of Medicine, New York University Medical Center, New York 10016
| | | | | | | | | | | | | |
Collapse
|
36
|
Franklyn JA. The molecular mechanisms of thyroid hormone action. BAILLIERE'S CLINICAL ENDOCRINOLOGY AND METABOLISM 1988; 2:891-909. [PMID: 2908318 DOI: 10.1016/s0950-351x(88)80023-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
37
|
Transcriptional and posttranscriptional regulation of rat hepatic 3-hydroxy-3-methylglutaryl-coenzyme A reductase by thyroid hormones. J Biol Chem 1988. [DOI: 10.1016/s0021-9258(18)37775-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
38
|
Yusta B, Ortiz-Caro J, Pascual A, Aranda A. Mechanism of L-triiodothyronine (T3) uptake by glial C6 cells: regulation by butyrate. Mol Cell Endocrinol 1988; 58:191-8. [PMID: 3208994 DOI: 10.1016/0303-7207(88)90154-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The mode of entry of triiodothyronine (T3) and its regulation by butyrate was studied in cultured glial C6 cells. Uptake of [125I]T3 increases for at least 60 min in C6 cells. The amount of cell-associated radioactivity is 2- to 4-fold higher during the entire time-course in cells previously exposed to 2 mM butyrate for 48 h. Uptake was non-saturable since uptake velocity was linearly related to the extracellular hormone concentration between 0.2 and 800 nM T3 in control and butyrate-treated cells. Uptake velocity increased by more than 3-fold in the cells incubated with the fatty acid. T3 uptake was temperature dependent and the effect of butyrate was observed at the different temperatures examined. Preincubation with metabolic inhibitors did not block [125I]T3 uptake in either group, and monodansylcadaverine was also ineffective. Present results suggest that in C6 cells T3 uptake proceeds by a passive, energy-independent, non-saturable process, that is markedly affected by short-chain fatty acids. Additionally, this is the first study documenting that a natural compound directly influences the entry of thyroid hormones into cells.
Collapse
Affiliation(s)
- B Yusta
- Unidad Endocrinologia Experimental, Facultad de Medicina, Universidad Autonóma, Madrid, Spain
| | | | | | | |
Collapse
|
39
|
The thyroid hormone receptor binds to multiple domains of the rat growth hormone 5′-flanking sequence. J Biol Chem 1988. [DOI: 10.1016/s0021-9258(19)76557-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
40
|
Horowitz ZD, Sahnoun H, Pascual A, Casanova J, Samuels HH. Analysis of photoaffinity label derivatives to probe thyroid hormone receptor in human fibroblasts, GH1 cells, and soluble receptor preparations. J Biol Chem 1988. [DOI: 10.1016/s0021-9258(18)68689-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
41
|
Samuels HH, Forman BM, Horowitz ZD, Ye ZS. Regulation of gene expression by thyroid hormone. J Clin Invest 1988; 81:957-67. [PMID: 3280604 PMCID: PMC329618 DOI: 10.1172/jci113449] [Citation(s) in RCA: 207] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- H H Samuels
- Department of Medicine, New York University Medical Center, New York 10016
| | | | | | | |
Collapse
|
42
|
Chapter 5 Mechanism of action of thyroid hormone. ACTA ACUST UNITED AC 1988. [DOI: 10.1016/s0167-7306(08)60640-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
43
|
Samuels HH, Aranda A, Casanova J, Copp RP, Flug F, Forman BM, Horowitz ZD, Janocko L, Park HY, Pascual A. Identification of the cis-acting elements and trans-acting factors that mediate cell-specific and thyroid hormone stimulation of growth hormone gene expression. RECENT PROGRESS IN HORMONE RESEARCH 1988; 44:53-114. [PMID: 3064211 DOI: 10.1016/b978-0-12-571144-9.50007-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
44
|
Franklyn JA, Gammage MD, Sheppard MC. Amiodarone and thyroid hormone effects on anterior pituitary hormone gene expression. Clin Endocrinol (Oxf) 1987; 27:373-82. [PMID: 3427795 DOI: 10.1111/j.1365-2265.1987.tb01164.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Amiodarone therapy results in marked changes in circulating thyroid hormone and TSH concentrations in man. In the present study we have demonstrated that amiodarone treatment of the rat increases serum TSH and pituitary cytoplasmic concentrations of TSH beta and alpha subunit messenger RNAs (mRNAs) and reduces PRL mRNA as measured by cytoplasmic dot hybridization with specific complementary (c) DNA probes. The fall in circulating TSH and TSH mRNA resulting from thyroid hormone treatment was less marked in animals receiving amiodarone in addition to T3 and T4. In contrast, in the hypothyroid state, increases in serum TSH, TSH beta and alpha mRNA, and reductions in PRL and GH mRNA were less marked in rats treated with amiodarone. In studies of rat anterior pituitary cells in primary monolayer culture we demonstrated a direct effect of amiodarone on PRL gene expression which was antagonized by T3. Changes in circulating thyroid hormone concentrations and deiodination of T4 and T3 induced by amiodarone in vivo may be important in the regulation of pituitary hormone gene expression but we have, in addition, shown a direct interaction between amiodarone and T3 effects on the anterior pituitary cell.
Collapse
Affiliation(s)
- J A Franklyn
- Department of Endocrinology, University of Birmingham, Queen Elizabeth Hospital, Edgbaston, UK
| | | | | |
Collapse
|
45
|
Valcavi R, Dieguez C, Preece M, Taylor A, Portioli I, Scanlon MF. Effect of thyroxine replacement therapy on plasma insulin-like growth factor 1 levels and growth hormone responses to growth hormone releasing factor in hypothyroid patients. Clin Endocrinol (Oxf) 1987; 27:85-90. [PMID: 3115637 DOI: 10.1111/j.1365-2265.1987.tb00842.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The aim of this study was to evaluate the effect of T4 replacement therapy on plasma insulin-like growth factor 1 (IGF-1) levels in patients with primary hypothyroidism to see whether recovery of pituitary GH responsiveness to GRF was associated with increased plasma IGF-1 levels. IGF-1 levels and GH responses to GRF (1 microgram/kg) were measured in 21 patients with primary hypothyroidism before and after T4 replacement therapy. T4 increased plasma IGF-1 levels (57.2 +/- 4.4 vs 75.9 +/- 8.8 ng/ml, mean +/- SEM, P less than 0.05) and GH responses to GRF as assessed both by peak GH levels (9 +/- 1.5 ng/ml before treatment vs 16.7 +/- 3 ng/ml after treatment, mean +/- SEM, P less than 0.05) and area under curve (496 +/- 92 before treatment vs 896 +/- 161 after treatment, mean- +/- SEM, P less than 0.05). Linear regression analysis showed a positive correlation between free T3 and IGF-1 levels after treatment (r = 0.37, P less than 0.05) and a negative relationship between plasma IGF-1 levels before treatment and delta IGF following T4 replacement therapy (r = 0.45, P less than 0.025). However, no correlation was found between plasma IGF-1 levels and GH responses to GRF, suggesting that GH responses to GRF are of no predictive value in relation to the recovery of plasma IGF-1 levels following T4 replacement therapy in hypothyroid patients.
Collapse
Affiliation(s)
- R Valcavi
- Sezione Endocrino Metabolica, Unita Sanitaria Locale, Reggio Emilia, Italy
| | | | | | | | | | | |
Collapse
|
46
|
Ichikawa K, DeGroot LJ. Purification and characterization of rat liver nuclear thyroid hormone receptors. Proc Natl Acad Sci U S A 1987; 84:3420-4. [PMID: 3472213 PMCID: PMC304882 DOI: 10.1073/pnas.84.10.3420] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Nuclear thyroid hormone receptor was purified to 904 pmol of L-3,5,3'-triiodothyronine (T3) binding capacity per mg of protein with 2.5-5.2% recovery by sequentially using hydroxylapatite column chromatography, ammonium sulfate precipitation, Sephadex G-150 gel filtration, DNA-cellulose column chromatography, DEAE-Sephadex column chromatography, and heparin-Sepharose column chromatography. Assuming that one T3 molecule binds to the 49,000-Da unit of the receptor, we reproducibly obtained 6.4-14.7 micrograms of receptor protein with 4.2-4.9% purity from 4-5 kg of rat liver. Elution of receptor from the heparin-Sepharose column was performed using 10 mM pyridoxal 5'-phosphate, which was observed to diminish binding of receptor to heparin-Sepharose or DNA-cellulose. This effect was specific for pyridoxal 5'-phosphate, since related compounds were not effective. Purified receptor bound T3 with high affinity (6.0 X 10(9) liter/mol), and the order of affinity of iodothyronine analogues to purified receptor was identical to that observed with crude receptor preparations [3,5,3'-triiodothyroacetic acid greater than L-T3 greater than D-3,5,3'-triiodothyronine (D-T3) greater than L-thyroxine greater than D-thyroxine]. Purified receptor had a sedimentation coefficient of 3.4 S, Stokes radius of 34 A, and calculated molecular mass of 49,000. Among several bands identified by silver staining after electrophoresis in NaDodSO4/polyacrylamide gels, one 49,000-Da protein showed photoaffinity labeling with [125I]thyroxine that was displaceable with excess unlabeled T3. The tryptic fragment and endogenous proteinase-digested fragment of the affinity-labeled receptor showed saturable binding in 27,000-Da and 36,000-Da peptides, respectively. These molecular masses are in agreement with estimates from gel filtration and gradient sedimentation, indicating that affinity labeling occurred at the hormone binding domain of nuclear thyroid hormone receptor. This procedure reproducibly provides classical native rat liver T3 nuclear receptor in useful quantity and purity and of the highest specific activity so far reported.
Collapse
|
47
|
Abstract
Rat liver nuclear thyroid hormone receptor was purified to 700-1600 pmol T3 binding capacity/mg protein by sequentially using hydroxylapatite column, ammonium sulfate precipitation, Sephadex G-150 gel filtration, DNA-cellulose column, DEAE-Sephadex A-50 column, and heparin-Sepharose column. Serum from a mouse immunized using this purified receptor preparation caused a shift of [125I]T3-receptor peak on glycerol density gradient sedimentation from 3.4 S to approximately 7 S. [125I]T3-receptor complex was immunoprecipitated using this serum and goat anti-mouse IgG. The serum showed reduced ability to immunoprecipitate the globular T3 binding fragment with Stokes radius of 22 A produced by trypsin digestion, a receptor fragment which has core histone and hormone binding but not DNA binding activity. These data indicate the production of anti-nuclear thyroid hormone receptor antibody which mainly recognized epitopes unrelated to hormone and core histone binding domain.
Collapse
|
48
|
Discrete positive and negative thyroid hormone-responsive transcription regulatory elements of the rat growth hormone gene. J Biol Chem 1987. [DOI: 10.1016/s0021-9258(18)45625-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
49
|
Ortiz-Caro J, Montiel F, Yusta B, Pascual A, Aranda A. Down-regulation of thyroid hormone nuclear receptor levels by L-triiodothyronine in cultured glial C6 cells. Mol Cell Endocrinol 1987; 49:255-63. [PMID: 3556756 DOI: 10.1016/0303-7207(87)90220-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
L-Triiodothyronine (T3) produced a time- and dose-dependent depletion of nuclear thyroid hormone receptor levels in C6 cells, a rat glioma cell line. Receptor number diminished by 30-40% after a 48 h incubation with concentrations of T3 that saturate the nuclear receptor. The nuclear binding curve obtained in cells incubated for 48 h with T3 was shifted leftward of the curve obtained after a 3 h incubation, which indicates an apparent increase in receptor affinity after long-term incubation with T3. However, this change probably represents a further equilibration of the hormone, since the dissociation rate from the nuclei was similar in C6 cells after long- and short-term incubation with T3. The effect of T3 was further demonstrated in C6 cells incubated with short-chain fatty acids. Butyrate and isobutyrate increased receptor levels, and T3 partially decreased the response to these compounds. These findings suggest the existence of a desensitization process by which C6 glial cells would be protected against an excess of thyroid hormone.
Collapse
|
50
|
Horowitz ZD, Samuels HH. Photoaffinity labeling of thyroid hormone receptors. Pharmacol Ther 1987; 33:317-32. [PMID: 3310036 DOI: 10.1016/0163-7258(87)90069-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Photoaffinity label probes of iodothyronines can interact with nuclear receptors in intact cells and in solubilized receptor preparations. These probes have certain advantages over a chemical affinity label in analyzing receptor structure. First, a photoaffinity label probe covalently cross-links only after photoactivation. Therefore, it is possible to demonstrate with appropriate competitive inhibition studies that the photoaffinity label probe associates with the receptor in question. Secondly, since cross-linking only occurs after photolysis, it is possible to adjust the concentration of the photoaffinity label to maximize association with "specific" binding sites relative to "non-specific" associations prior to covalent linkage by photoactivation. The different [125I]iodothyronine-PAL analogues may be useful as probes of the thyroid hormone receptor binding domain since PAL compounds with different affinities for receptor may photocouple to different receptor residues within or proximate to the hormone binding region. These probes may also be useful as an adjunct to receptor purification and in probing the organization of the receptor in chromatin. Lastly, they may provide insights into possible alterations of receptor structure in patients with partial end organ resistance to thyroid hormone (Refetoff et al., 1967; Eil et al., 1982).
Collapse
Affiliation(s)
- Z D Horowitz
- Rose F. Tishman Laboratories for Geriatric Endocrinology, Department of Medicine, New York University Medical Center, New York 10016
| | | |
Collapse
|