1
|
Zhu X, Cheng SY. Thyroid Hormone Receptors as Tumor Suppressors in Cancer. Endocrinology 2024; 165:bqae115. [PMID: 39226152 PMCID: PMC11406550 DOI: 10.1210/endocr/bqae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/05/2024]
Abstract
Accumulated research has revealed the multifaceted roles of thyroid hormone receptors (TRs) as potent tumor suppressors across various cancer types. This review explores the intricate mechanisms underlying TR-mediated tumor suppression, drawing insights from preclinical mouse models and cancer biology. This review examines the tumor-suppressive functions of TRs, particularly TRβ, in various cancers using preclinical models, revealing their ability to inhibit tumor initiation, progression, and metastasis. Molecular mechanisms underlying TR-mediated tumor suppression are discussed, including interactions with oncogenic signaling pathways like PI3K-AKT, JAK-STAT, and transforming growth factor β. Additionally, this paper examines TRs' effect on cancer stem cell activity and differentiation, showcasing their modulation of key cellular processes associated with tumor progression and therapeutic resistance. Insights from preclinical studies underscore the therapeutic potential of targeting TRs to impede cancer stemness and promote cancer cell differentiation, paving the way for precision medicine in cancer treatment and emphasizing the potential of TR-targeted therapies as promising approaches for treating cancers and improving patient outcomes.
Collapse
Affiliation(s)
- Xuguang Zhu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sheue-yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
2
|
Kuliczkowska-Płaksej J, Zdrojowy-Wełna A, Jawiarczyk-Przybyłowska A, Gojny Ł, Bolanowski M. Diagnosis and therapeutic approach to bone health in patients with hypopituitarism. Rev Endocr Metab Disord 2024; 25:513-539. [PMID: 38565758 DOI: 10.1007/s11154-024-09878-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
The results of many studies in recent years indicate a significant impact of pituitary function on bone health. The proper function of the pituitary gland has a significant impact on the growth of the skeleton and the appearance of sexual dimorphism. It is also responsible for achieving peak bone mass, which protects against the development of osteoporosis and fractures later in life. It is also liable for the proper remodeling of the skeleton, which is a physiological mechanism managing the proper mechanical resistance of bones and the possibility of its regeneration after injuries. Pituitary diseases causing hypofunction and deficiency of tropic hormones, and thus deficiency of key hormones of effector organs, have a negative impact on the skeleton, resulting in reduced bone mass and susceptibility to pathological fractures. The early appearance of pituitary dysfunction, i.e. in the pre-pubertal period, is responsible for failure to achieve peak bone mass, and thus the risk of developing osteoporosis in later years. This argues for the need for a thorough assessment of patients with hypopituitarism, not only in terms of metabolic disorders, but also in terms of bone disorders. Early and properly performed treatment may prevent patients from developing the bone complications that are so common in this pathology. The aim of this review is to discuss the physiological, pathophysiological, and clinical insights of bone involvement in pituitary disease.
Collapse
Affiliation(s)
- Justyna Kuliczkowska-Płaksej
- Department and Clinic of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, Wybrzeże Pasteura 4, Wrocław, 50-367, Poland
| | - Aleksandra Zdrojowy-Wełna
- Department and Clinic of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, Wybrzeże Pasteura 4, Wrocław, 50-367, Poland
| | - Aleksandra Jawiarczyk-Przybyłowska
- Department and Clinic of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, Wybrzeże Pasteura 4, Wrocław, 50-367, Poland.
| | - Łukasz Gojny
- Department and Clinic of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, Wybrzeże Pasteura 4, Wrocław, 50-367, Poland
| | - Marek Bolanowski
- Department and Clinic of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, Wybrzeże Pasteura 4, Wrocław, 50-367, Poland
| |
Collapse
|
3
|
Lademann F, Tsourdi E, Hofbauer LC, Rauner M. Thyroid hormone receptor Thra and Thrb knockout differentially affects osteoblast biology and thyroid hormone responsiveness in vitro. J Cell Biochem 2023; 124:1948-1960. [PMID: 37992217 DOI: 10.1002/jcb.30500] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023]
Abstract
Thyroid hormones (TH) are important modulators of bone remodeling and thus, thyroid diseases, in particular hyperthyroidism, are able to compromise bone quality and fracture resistance. TH actions on bone are mediated by the thyroid hormone receptors (TR) TRα1 and TRβ1, encoded by Thra and Thrb, respectively. Skeletal phenotypes of mice lacking Thra (Thra0/0 ) and Thrb (Thrb-/- ) are well-described and suggest that TRα1 is the predominant mediator of TH actions in bone. Considering that bone cells might be affected by systemic TH changes seen in these mutant mice, here we investigated the effects of TR knockout on osteoblasts exclusively at the cellular level. Primary osteoblasts obtained from Thra0/0 , Thrb-/- , and respective wildtype (WT) mice were analyzed regarding their differentiation potential, activity and TH responsiveness in vitro. Thra, but not Thrb knockout promoted differentiation and activity of early, mature and late osteoblasts as compared to respective WT cells. Interestingly, while mineralization capacity and expression of osteoblast marker genes and TH target gene Klf9 was increased by TH in WT and Thra-deficient osteoblasts, Thrb knockout mitigated the responsiveness of osteoblasts to short (48 h) and long term (10 d) TH treatment. Further, we found a low ratio of Rankl, a potent osteoclast stimulator, over osteoprotegerin, an osteoclast inhibitor, in Thrb-deficient osteoblasts and in line, supernatants obtained from Thrb-/- osteoblasts reduced numbers of primary osteoclasts in vitro. In accordance to the increased Rankl/Opg ratio in TH-treated WT osteoblasts only, supernatants from these cells, but not from TH-treated Thrb-/- osteoblasts increased the expression of Trap and Ctsk in osteoclasts, suggesting that osteoclasts are indirectly stimulated by TH via TRβ1 in osteoblasts. In conclusion, our study shows that both Thra and Thrb differentially affect activity, differentiation and TH response of osteoblasts in vitro and emphasizes the importance of TRβ1 to mediate TH actions in bone.
Collapse
Affiliation(s)
- Franziska Lademann
- Department of Medicine III and University Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| | - Elena Tsourdi
- Department of Medicine III and University Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III and University Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and University Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| |
Collapse
|
4
|
Wu X, Zhai F, Chang A, Wei J, Guo Y, Zhang J. Association between sensitivity to thyroid hormone indices and osteoporosis in euthyroid patients with type 2 diabetes mellitus. Ther Adv Chronic Dis 2023; 14:20406223231189230. [PMID: 37538345 PMCID: PMC10395177 DOI: 10.1177/20406223231189230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 06/08/2023] [Indexed: 08/05/2023] Open
Abstract
Background Thyroid hormones are known to regulate bone metabolism and may influence bone mineral density (BMD), as well as the risk of osteoporosis (OP) and fractures in patients with type 2 diabetes mellitus (T2DM). Recently, sensitivity to thyroid hormone indices has been linked with T2DM and OP independently. However, the relationship between thyroid hormone sensitivity and OP in euthyroid T2DM patients has yet to be investigated. Objectives The aim of this study was to determine the association between sensitivity to thyroid hormone indices and the risk of OP in euthyroid patients with T2DM. Design This study employed a retrospective, cross-sectional design and utilized data acquired from the Cangzhou Central Hospital in China between 2019 and 2020. Methods We retrospectively analyzed the data of 433 patients with T2DM for anthropometric measurements, clinical laboratory test results, and BMD. The thyroid-stimulating hormone index, thyrotroph thyroxine resistance index, and thyroid feedback quantile-based index (TFQI) were calculated to determine thyroid hormone sensitivity. Finally, multivariable logistic regression, generalized additive models, and subgroup analysis were performed to detect the association between sensitivity to thyroid hormone indices and the risk of OP in these patients. Results We did not observe a statistically significant linear relationship between sensitivity to thyroid hormones indices and OP after covariate adjustment. However, a nonlinear relationship existed between TFQI and the prevalence of OP. The inflection point of the TFQI was at -0.29. The effect sizes (odds ratio) on the left and right of the inflection point were 0.07 [95% confidence interval (CI): 0.01-0.71; p = 0.024] and 2.78 (95% CI: 1.02-7.58; p = 0.046), respectively. This trend was consistent in older female patients with higher body mass index (BMI; 25-30 kg/m2). Conclusion An approximate U-shaped relationship was observed between sensitivity to thyroid hormone indices and OP risk in euthyroid patients with T2DM with variations in sex, age, and BMI. These findings provide a new perspective to elucidate the role of thyroid hormones in OP, specifically in patients with T2DM.
Collapse
Affiliation(s)
- Xuelun Wu
- Department of Endocrinology, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou City 061000, Hebei Province, PR China
| | - Furui Zhai
- Gynecological clinic, Cangzhou Central Hospital, Cangzhou City, Hebei Province, PR China
| | - Ailing Chang
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, PR China
| | - Jing Wei
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, PR China
| | - Yanan Guo
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, PR China
| | - Jincheng Zhang
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, PR China
| |
Collapse
|
5
|
Zhu S, Pang Y, Xu J, Chen X, Zhang C, Wu B, Gao J. Endocrine Regulation on Bone by Thyroid. Front Endocrinol (Lausanne) 2022; 13:873820. [PMID: 35464058 PMCID: PMC9020229 DOI: 10.3389/fendo.2022.873820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND As an endocrine organ, the thyroid acts on the entire body by secreting a series of hormones, and bone is one of the main target organs of the thyroid. SUMMARY This review highlights the roles of thyroid hormones and thyroid diseases in bone homeostasis. CONCLUSION Thyroid hormones play significant roles in the growth and development of bone, and imbalance of thyroid hormones can impair bone homeostasis.
Collapse
Affiliation(s)
- Siyuan Zhu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yidan Pang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jun Xu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiaoyi Chen
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Changqing Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Junjie Gao, ; Bo Wu, ; Changqing Zhang,
| | - Bo Wu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Junjie Gao, ; Bo Wu, ; Changqing Zhang,
| | - Junjie Gao
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
- *Correspondence: Junjie Gao, ; Bo Wu, ; Changqing Zhang,
| |
Collapse
|
6
|
Lakatos P, Szili B, Bakos B, Takacs I, Putz Z, Istenes I. Thyroid Hormones, Glucocorticoids, Insulin, and Bone. Handb Exp Pharmacol 2020; 262:93-120. [PMID: 32036458 DOI: 10.1007/164_2019_314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Several endocrine systems have important effects on bone tissue. Thyroid hormones are essential for normal growth and development. Excess of these hormones will result in clinically significant changes that may require intervention. Glucocorticoids also have a marked effect on bone metabolism by several pathways. Their endogenous or exogenous excess will induce pathological processes that might elevate the risk of fractures. Insulin and the carbohydrate metabolism elicit a physiological effect on bone; however, the lack of insulin (type 1 diabetes) or insulin resistance (type 2 diabetes) have deleterious influence on bone tissue.
Collapse
Affiliation(s)
- Peter Lakatos
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary.
| | - Balazs Szili
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Bence Bakos
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Istvan Takacs
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Putz
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Ildiko Istenes
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
7
|
Leitch VD, Bassett JHD, Williams GR. Role of thyroid hormones in craniofacial development. Nat Rev Endocrinol 2020; 16:147-164. [PMID: 31974498 DOI: 10.1038/s41574-019-0304-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2019] [Indexed: 02/07/2023]
Abstract
The development of the craniofacial skeleton relies on complex temporospatial organization of diverse cell types by key signalling molecules. Even minor disruptions to these processes can result in deleterious consequences for the structure and function of the skull. Thyroid hormone deficiency causes delayed craniofacial and tooth development, dysplastic facial features and delayed development of the ossicles in the middle ear. Thyroid hormone excess, by contrast, accelerates development of the skull and, in severe cases, might lead to craniosynostosis with neurological sequelae and facial hypoplasia. The pathogenesis of these important abnormalities remains poorly understood and underinvestigated. The orchestration of craniofacial development and regulation of suture and synchondrosis growth is dependent on several critical signalling pathways. The underlying mechanisms by which these key pathways regulate craniofacial growth and maturation are largely unclear, but studies of single-gene disorders resulting in craniofacial malformations have identified a number of critical signalling molecules and receptors. The craniofacial consequences resulting from gain-of-function and loss-of-function mutations affecting insulin-like growth factor 1, fibroblast growth factor receptor and WNT signalling are similar to the effects of altered thyroid status and mutations affecting thyroid hormone action, suggesting that these critical pathways interact in the regulation of craniofacial development.
Collapse
Affiliation(s)
- Victoria D Leitch
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Royal Melbourne Institute of Technology (RMIT) Centre for Additive Manufacturing, RMIT University, Melbourne, VIC, Australia
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
8
|
Shibata Y, Wen L, Okada M, Shi YB. Organ-Specific Requirements for Thyroid Hormone Receptor Ensure Temporal Coordination of Tissue-Specific Transformations and Completion of Xenopus Metamorphosis. Thyroid 2020; 30:300-313. [PMID: 31854240 PMCID: PMC7047119 DOI: 10.1089/thy.2019.0366] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Thyroid hormone (triiodothyronine [T3]) is essential for the development throughout vertebrates. Anuran metamorphosis mimics mammalian postembryonic development, a period around birth when plasma T3 level peaks and many organs/tissues mature into their adult forms. Compared with the uterus-enclosed mammalian embryos, tadpoles can be easily manipulated to study the roles of T3 and T3 receptors (TRs) in tissue remodeling and adult organ development. We and others have previously knocked out TRα or TRβ in the diploid anuran Xenopus tropicalis and reported distinct effects of the two receptor knockouts on metamorphosis. However, animals lacking either TRα or TRβ can complete metamorphosis and develop into reproductive adults. Methods: We have generated TRα and TRβ double knockout animals and carried out molecular and morphological analyses to determine if TR is required for Xenopus development. Results: We found that the TR double knockout tadpoles do not respond to T3, supporting the view that there are no other TR genes in X. tropicalis and that TR is essential for mediating the effects of T3 in vivo. Surprisingly, the double knockout tadpoles are able to initiate metamorphosis and accomplish many metamorphic changes, such as limb development. However, all double knockout tadpoles stall and eventually die at stage 61, the climax of metamorphosis, before tail resorption takes place. Analyses of the knockout tadpoles at stage 61 revealed various developmental abnormalities, including precocious ossification and extra vertebrae. Conclusions: Our data indicate that TRs are not required for the initiation of metamorphosis but is essential for the completion of metamorphosis. Furthermore, the differential effects of TR knockout on different organs/tissues suggest tissue-specific roles for TR to control temporal coordination and progression of metamorphosis in various organs.
Collapse
Affiliation(s)
- Yuki Shibata
- Section on Molecular Morphogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Luan Wen
- Section on Molecular Morphogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Morihiro Okada
- Section on Molecular Morphogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
- Address correspondence to: Yun-Bo Shi, PhD, Section on Molecular Morphogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
9
|
Han CR, Holmsen E, Carrington B, Bishop K, Zhu YJ, Starost M, Meltzer P, Sood R, Liu P, Cheng SY. Generation of Novel Genetic Models to Dissect Resistance to Thyroid Hormone Receptor α in Zebrafish. Thyroid 2020; 30:314-328. [PMID: 31952464 PMCID: PMC7047097 DOI: 10.1089/thy.2019.0598] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Patients with mutations of the thyroid hormone receptor alpha (THRA) gene show resistance to thyroid hormone alpha (RTHα). No amendable mouse models are currently available to elucidate deleterious effects of TRα1 mutants during early development. Zebrafish with transient suppressed expression by morpholino knockdown and ectopic expression of TRα1 mutants in the embryos have been reported. However, zebrafish with germline transmittable mutations have not been reported. The stable expression of thra mutants from embryos to adulthood facilitated the study of molecular actions of TRα1 mutants during development. Methods: In contrast to human and mice, the thra gene is duplicated in zebrafish, thraa, and thrab. Using CRISPR/Cas9-mediated targeted mutagenesis, we created dominant negative mutations in the two duplicated thra genes. We comprehensively analyzed the molecular and phenotypic characteristics of mutant fish during development. Results: Adult and juvenile homozygous thrab 1-bp ins (m/m) mutants exhibited severe growth retardation, but adult homozygous thraa 8-bp ins (m/m) mutants had very mild growth impairment. Expression of the growth hormone (gh1) and insulin-like growth factor 1 was markedly suppressed in homozygous thrab 1-bp ins (m/m) mutants. Decreased messenger RNA and protein levels of triiodothyronine-regulated keratin genes and inhibited keratinocyte proliferation resulted in hypoplasia of the epidermis in adult and juvenile homozygous thrab 1-bp ins (m/m) mutants, but not homozygous thraa 8-bp ins (m/m) mutants. RNA-seq analysis showed that homozygous thrab 1-bp ins (m/m) mutation had global impact on the functions of the adult pituitary. However, no morphological defects nor any changes in the expression of gh1 and keratin genes were observed in the embryos and early larvae. Thus, mutations of either the thraa or thrab gene did not affect initiation of embryogenesis. But the mutation of the thrab gene, but not the thraa gene, is detrimental in postlarval growth and skin development. Conclusions: The thra duplicated genes are essential to control temporal coordination in postlarval growth and development in a tissue-specific manner. We uncovered novel functions of the duplicated thra genes in zebrafish in development. These mutant zebrafish could be used as a model for further analysis of TRα1 mutant actions and for rapid screening of therapeutics for RTHα.
Collapse
Affiliation(s)
- Cho Rong Han
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Erik Holmsen
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Blake Carrington
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Kevin Bishop
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Yuelin Jack Zhu
- Laboratory of Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Matthew Starost
- Division of Veterinary Resources, Diagnostic and Research Services Branch, National Institutes of Health, Bethesda, Maryland
| | - Paul Meltzer
- Laboratory of Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Raman Sood
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Paul Liu
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Sheue-yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Address correspondence to: Sheue-yann Cheng, PhD, Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5128, Bethesda, MD 20892-4264
| |
Collapse
|
10
|
Abstract
Thyroid hormone has profound effects on skeletal development and adult bone maintenance. Here, we review the current literature concerning thyroid hormone action in bone and cartilage in relation to human disease and animal models. We describe state-of-the-art imaging and biomechanical methods used to determine structural and functional parameters in the skeletal phenotyping of mouse models.
Collapse
|
11
|
Lee H, Lee M, Kim HK, Kim YO, Kwon JT, Kim HJ. Influence of clozapine on neurodevelopmental protein expression and behavioral patterns in animal model of psychiatric disorder induced by low-level of lead. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2019; 23:467-474. [PMID: 31680768 PMCID: PMC6819901 DOI: 10.4196/kjpp.2019.23.6.467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 08/26/2019] [Accepted: 10/08/2019] [Indexed: 11/15/2022]
Abstract
Exposure to lead during pregnancy is a risk factor for the development of psychiatric disorders in the offspring. In this study, we investigated whether exposure to low levels of lead acetate (0.2%) in drinking water during pregnancy and lactation causes behavioral impairment and affects the expression of proteins associated with neurodevelopment. Lead exposure altered several parameters in rat offspring compared with those unexposed in open-field, social interaction, and pre-pulse inhibition tests. These parameters were restored to normal levels after clozapine treatment. Western blot and immunohistochemical analyses of the hippocampus revealed that several neurodevelopmental proteins were downregulated in lead-exposed rats. The expression was normalized after clozapine treatment (5 mg/kg/day, postnatal day 35–56). These findings demonstrate that downregulation of several proteins in lead-exposed rats affected subsequent behavioral changes. Our results suggest that lead exposure in early life may induce psychiatric disorders and treatment with antipsychotics such as clozapine may reduce their incidence.
Collapse
Affiliation(s)
- Hwayoung Lee
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Minyoung Lee
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Hyung-Ki Kim
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Young Ock Kim
- Department of Bio-Environmental Chemistry, College of Agriculture and Life Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Jun-Tack Kwon
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Hak-Jae Kim
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| |
Collapse
|
12
|
Zaitune CR, Fonseca TL, Capelo LP, Freitas FR, Beber EH, Dora JM, Wang CC, Miranda-Rodrigues M, Nonaka KO, Maia AL, Gouveia CHA. Abnormal Thyroid Hormone Status Differentially Affects Bone Mass Accrual and Bone Strength in C3H/HeJ Mice: A Mouse Model of Type I Deiodinase Deficiency. Front Endocrinol (Lausanne) 2019; 10:300. [PMID: 31156551 PMCID: PMC6530334 DOI: 10.3389/fendo.2019.00300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/26/2019] [Indexed: 12/26/2022] Open
Abstract
C3H/HeJ (C3H) mice are deficient of type I deiodinase (D1), an enzyme that activates thyroid hormone (TH), converting thyroxine (T4) to triiodothyronine (T3). Nevertheless, C3H mice present normal serum T3 and a gross euthyroid phenotype. To investigate if a global D1 deficiency interferes in the TH effects on bone, we compared bone growth, bone mass accrual and bone strength of C3H and C57BL/6J (B6) mice under abnormal TH status. Four-week-old female mice of both strains were grouped as Euthyroid, Hypothyroid (pharmacologically-induced), 1xT4 and 10xT4 (hypothyroid animals receiving 1- or 10-fold the physiological dose of T4 /day/16 weeks). Hypothyroidism and TH excess similarly impaired body weight (BW) gain and body growth in both mice strains. In contrast, whereas hypothyroidism only slightly impaired bone mineral density (BMD) accrual in B6 mice, it severely impaired BMD accrual in C3H mice. No differences were observed in serum and bone concentrations of T3 between hypothyroid animals of both strains. Interestingly, treatment with 10xT4 was less deleterious to BMD accrual in C3H than in B6 mice and resulted in less elevated T3 serum levels in B6 than in C3H mice, which is probably explained by the lower D1 activity in C3H mice. In addition, hypothyroidism decreased bone strength only in C3H but not in B6 mice, while TH excess decreased this parameter in both strains. These findings indicate that D1 deficiency contributes to the TH excess-induced differences in bone mass accrual in C3H vs. B6 mice and suggest that deiodinase-unrelated genetic factors might account for the different skeleton responses to hypothyroidism between strains.
Collapse
Affiliation(s)
- Clarissa R. Zaitune
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Institute of Healthy Sciences, Paulista University, São Paulo, Brazil
| | - Tatiana L. Fonseca
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Chicago, Chigago, IL, United States
| | - Luciane P. Capelo
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Institute of Science and Technology, Federal University of São Paulo, São Paulo, Brazil
| | - Fatima R. Freitas
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Heart Institute (InCor) of Medical School Hospital, University of São Paulo, São Paulo, Brazil
| | - Eduardo H. Beber
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - José M. Dora
- Endocrine Division, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Charles C. Wang
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Manuela Miranda-Rodrigues
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Keico O. Nonaka
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Ana L. Maia
- Endocrine Division, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Cecilia H. A. Gouveia
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- *Correspondence: Cecilia H. A. Gouveia
| |
Collapse
|
13
|
Mazziotti G, Frara S, Giustina A. Pituitary Diseases and Bone. Endocr Rev 2018; 39:440-488. [PMID: 29684108 DOI: 10.1210/er.2018-00005] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022]
Abstract
Neuroendocrinology of bone is a new area of research based on the evidence that pituitary hormones may directly modulate bone remodeling and metabolism. Skeletal fragility associated with high risk of fractures is a common complication of several pituitary diseases such as hypopituitarism, Cushing disease, acromegaly, and hyperprolactinemia. As in other forms of secondary osteoporosis, pituitary diseases generally affect bone quality more than bone quantity, and fractures may occur even in the presence of normal or low-normal bone mineral density as measured by dual-energy X-ray absorptiometry, making difficult the prediction of fractures in these clinical settings. Treatment of pituitary hormone excess and deficiency generally improves skeletal health, although some patients remain at high risk of fractures, and treatment with bone-active drugs may become mandatory. The aim of this review is to discuss the physiological, pathophysiological, and clinical insights of bone involvement in pituitary diseases.
Collapse
Affiliation(s)
| | - Stefano Frara
- Institute of Endocrinology, Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Giustina
- Institute of Endocrinology, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
14
|
Daniš R, Hill M, Sedlak P. Differences in the auxological characters of children with short stature - Differential diagnostic possibilities of hypothyreosis. HOMO-JOURNAL OF COMPARATIVE HUMAN BIOLOGY 2018; 69:139-145. [PMID: 30017377 DOI: 10.1016/j.jchb.2018.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 05/01/2018] [Indexed: 11/25/2022]
Abstract
This study aimed to define the differences in growth characteristics in the three most frequent causes of growth retardation - growth hormone deficiency, hypothyreosis and constitutional delay of growth and development - in order to provide diagnostic means for distinguishing these disorders. The study included 166 children with growth disorders aged 4-18 years. The height for age, the bone age using the TW3 method, the predicted height as the target height and the current prediction using the TW3 method were studied. For bone age, the radius, ulna and short bones compartment (RUS) and carpal bones (CARP) were evaluated separately and the difference in their delay in relation to chronological age (ΔBA_RUS_CARP) was determined. The relationship of the studied variables with sex and the underlying diagnosis was tested and the relationship of hypothyreosis and growth data was estimated. The model was tested on the growth data of 104 randomly selected patients with a growth disorder. The largest significant distinction was demonstrated by the difference ΔBA_RUS_CARP in hypothyreosis. The created linear regression model was highly statistically significant (χ2 = 19.4, p < 0.0001) and showed high selectivity (0.609, 95% CI 0.409; 0.808) as well as high specificity (0.864, 95% CI 0.781; 0.946). The clinical validity of the model demonstrated a 61% predictive value for the detection and an 81% successful specification of hypothyreosis. The study demonstrated the possibility of distinguishing suspected hypothyreosis from other causes of growth retardation based on differences in severity of the ossification delay in skeletal compartments of the hand.
Collapse
Affiliation(s)
- R Daniš
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, Prague, Viničná 1594/7, 128 44 Prague 2, Czech Republic
| | - M Hill
- Institute of Endocrinology, Národní 139/8, 110 00 Prague 1, Czech Republic
| | - P Sedlak
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, Prague, Viničná 1594/7, 128 44 Prague 2, Czech Republic.
| |
Collapse
|
15
|
|
16
|
Gouveia CHA, Miranda-Rodrigues M, Martins GM, Neofiti-Papi B. Thyroid Hormone and Skeletal Development. VITAMINS AND HORMONES 2018; 106:383-472. [PMID: 29407443 DOI: 10.1016/bs.vh.2017.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thyroid hormone (TH) is essential for skeletal development from the late fetal life to the onset of puberty. During this large window of actions, TH has key roles in endochondral and intramembranous ossifications and in the longitudinal bone growth. There is evidence that TH acts directly in skeletal cells but also indirectly, specially via the growth hormone/insulin-like growth factor-1 axis, to control the linear skeletal growth and maturation. The presence of receptors, plasma membrane transporters, and activating and inactivating enzymes of TH in skeletal cells suggests that direct actions of TH in these cells are crucial for skeletal development, which has been confirmed by several in vitro and in vivo studies, including mouse genetic studies, and clinical studies in patients with resistance to thyroid hormone due to dominant-negative mutations in TH receptors. This review examines progress made on understanding the mechanisms by which TH regulates the skeletal development.
Collapse
Affiliation(s)
- Cecilia H A Gouveia
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil.
| | | | - Gisele M Martins
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil; Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Bianca Neofiti-Papi
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
17
|
Abstract
Thyroid hormones are essential for skeletal development and are important regulators of bone maintenance in adults. Childhood hypothyroidism causes delayed skeletal development, retarded linear growth and impaired bone mineral accrual. Epiphyseal dysgenesis is evidenced by classic features of stippled epiphyses on X-ray. In severe cases, post-natal growth arrest results in a complex skeletal dysplasia. Thyroid hormone replacement stimulates catch-up growth and bone maturation, but recovery may be incomplete dependent on the duration and severity of hypothyroidism prior to treatment. A severe phenotype characteristic of hypothyroidism occurs in children with resistance to thyroid hormone due to mutations affecting THRA encoding thyroid hormone receptor α (TRα). Discovery of this rare condition recapitulated animal studies demonstrating that TRα mediates thyroid hormone action in the skeleton. In adults, thyrotoxicosis is well known to cause severe osteoporosis and fracture, but cases are rare because of prompt diagnosis and treatment. Recent data, however, indicate that subclinical hyperthyroidism is associated with low bone mineral density (BMD) and an increased risk of fracture. Population studies have also shown that variation in thyroid status within the reference range in post-menopausal women is associated with altered BMD and fracture risk. Thus, thyroid status at the upper end of the euthyroid reference range is associated with low BMD and increased risk of osteoporotic fragility fracture. Overall, extensive data demonstrate that euthyroid status is required for normal post-natal growth and bone mineral accrual, and is fundamental for maintenance of adult bone structure and strength.
Collapse
Affiliation(s)
- G. R. Williams
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, 10N5 Commonwealth Building, London, W12 0NN UK
| | - J. H. D. Bassett
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, 10N6 Commonwealth Building, London, W12 0NN UK
| |
Collapse
|
18
|
Thyroid Hormone Signaling in the Development of the Endochondral Skeleton. VITAMINS AND HORMONES 2018; 106:351-381. [PMID: 29407442 PMCID: PMC9830754 DOI: 10.1016/bs.vh.2017.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Thyroid hormone (TH) is an established regulator of skeletal growth and maintenance both in clinical studies and in laboratory models. The clinical consequences of altered thyroid status on the skeleton during development and in adulthood are well known, and genetic mouse models in which elements of the TH signaling axis have been manipulated illuminate the mechanisms which underlie TH regulation of the skeleton. TH is involved in the regulation of the balance between proliferation and differentiation in several skeletal cell types including chondrocytes, osteoblasts, and osteoclasts. The effects of TH are mediated primarily via the thyroid hormone receptors (TRs) α and β, ligand-inducible nuclear receptors which act as transcription factors to regulate target gene expression. Both TRα and TRβ signaling are important for different stages of skeletal development. The molecular mechanisms of TH action in bone are complex and include interaction with a number of growth factor signaling pathways. This review provides an overview of the regulation and mechanisms of TH action in bone, focusing particularly on the role of TH in endochondral bone formation during postnatal growth.
Collapse
|
19
|
Leitch VD, Di Cosmo C, Liao XH, O’Boy S, Galliford TM, Evans H, Croucher PI, Boyde A, Dumitrescu A, Weiss RE, Refetoff S, Williams GR, Bassett JHD. An Essential Physiological Role for MCT8 in Bone in Male Mice. Endocrinology 2017; 158:3055-3066. [PMID: 28637283 PMCID: PMC5659673 DOI: 10.1210/en.2017-00399] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/12/2017] [Indexed: 11/19/2022]
Abstract
T3 is an important regulator of skeletal development and adult bone maintenance. Thyroid hormone action requires efficient transport of T4 and T3 into target cells. We hypothesized that monocarboxylate transporter (MCT) 8, encoded by Mct8 on the X-chromosome, is an essential thyroid hormone transporter in bone. To test this hypothesis, we determined the juvenile and adult skeletal phenotypes of male Mct8 knockout mice (Mct8KO) and Mct8D1D2KO compound mutants, which additionally lack the ability to convert the prohormone T4 to the active hormone T3. Prenatal skeletal development was normal in both Mct8KO and Mct8D1D2KO mice, whereas postnatal endochondral ossification and linear growth were delayed in both Mct8KO and Mct8D1D2KO mice. Furthermore, bone mass and mineralization were decreased in adult Mct8KO and Mct8D1D2KO mice, and compound mutants also had reduced bone strength. Delayed bone development and maturation in Mct8KO and Mct8D1D2KO mice is consistent with decreased thyroid hormone action in growth plate chondrocytes despite elevated serum T3 concentrations, whereas low bone mass and osteoporosis reflects increased thyroid hormone action in adult bone due to elevated systemic T3 levels. These studies identify an essential physiological requirement for MCT8 in chondrocytes, and demonstrate a role for additional transporters in other skeletal cells during adult bone maintenance.
Collapse
Affiliation(s)
- Victoria D. Leitch
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| | - Caterina Di Cosmo
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637
| | - Sam O’Boy
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| | - Thomas M. Galliford
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| | - Holly Evans
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Peter I. Croucher
- The Garvan Institute of Medical Research and St. Vincent’s Clinical School, University of New South Wales Medicine, Sydney, New South Wales 2010, Australia
| | - Alan Boyde
- Queen Mary University of London, Oral Growth and Development, Bart’s and The London School of Medicine and Dentistry, London E1 4NS, United Kingdom
| | | | - Roy E. Weiss
- Department of Medicine, University of Miami, Miami, Florida 33136
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637
- Department of Pediatrics, The University of Chicago, Chicago, Illinois 60637
- Committee on Genetics, The University of Chicago, Chicago, Illinois 60637
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| | - J. H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| |
Collapse
|
20
|
Durham E, Howie RN, Parsons T, Bennfors G, Black L, Weinberg SM, Elsalanty M, Yu JC, Cray JJ. Thyroxine Exposure Effects on the Cranial Base. Calcif Tissue Int 2017; 101:300-311. [PMID: 28391432 PMCID: PMC5545063 DOI: 10.1007/s00223-017-0278-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/03/2017] [Indexed: 01/19/2023]
Abstract
Thyroid hormone is important for skull bone growth, which primarily occurs at the cranial sutures and synchondroses. Thyroid hormones regulate metabolism and act in all stages of cartilage and bone development and maintenance by interacting with growth hormone and regulating insulin-like growth factor. Aberrant thyroid hormone levels and exposure during development are exogenous factors that may exacerbate susceptibility to craniofacial abnormalities potentially through changes in growth at the synchondroses of the cranial base. To elucidate the direct effect of in utero therapeutic thyroxine exposure on the synchondroses in developing mice, we provided scaled doses of the thyroid replacement drug, levothyroxine, in drinking water to pregnant C57BL6 wild-type dams. The skulls of resulting pups were subjected to micro-computed tomography analysis revealing less bone volume relative to tissue volume in the synchondroses of mouse pups exposed in utero to levothyroxine. Histological assessment of the cranial base area indicated more active synchondroses as measured by metabolic factors including Igf1. The cranial base of the pups exposed to high levels of levothyroxine also contained more collagen fiber matrix and an increase in markers of bone formation. Such changes due to exposure to exogenous thyroid hormone may drive overall morphological changes. Thus, excess thyroid hormone exposure to the fetus during pregnancy may lead to altered craniofacial growth and increased risk of anomalies in offspring.
Collapse
Affiliation(s)
- Emily Durham
- Department of Oral Health Sciences, Medical University of South Carolina, 173 Ashley Avenue, BS 230B, Charleston, SC, 29425, USA
| | - R Nicole Howie
- Department of Oral Health Sciences, Medical University of South Carolina, 173 Ashley Avenue, BS 230B, Charleston, SC, 29425, USA
| | - Trish Parsons
- Department of Oral Biology, School of Dental Medicine, Center for Craniofacial and Dental Genetics, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA, 15213, USA
| | - Grace Bennfors
- Department of Oral Health Sciences, Medical University of South Carolina, 173 Ashley Avenue, BS 230B, Charleston, SC, 29425, USA
| | - Laurel Black
- Department of Oral Health Sciences, Medical University of South Carolina, 173 Ashley Avenue, BS 230B, Charleston, SC, 29425, USA
| | - Seth M Weinberg
- Department of Oral Biology, School of Dental Medicine, Center for Craniofacial and Dental Genetics, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA, 15213, USA
| | - Mohammed Elsalanty
- Departments of Oral Biology, Cellular Biology and Anatomy, Orthopaedic Surgery and Oral and Maxillofacial Surgery, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
- Institute for Regenerative and Reparative Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Jack C Yu
- Institute for Regenerative and Reparative Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
- Division of Plastic Surgery, Department of Surgery, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - James J Cray
- Department of Oral Health Sciences, Medical University of South Carolina, 173 Ashley Avenue, BS 230B, Charleston, SC, 29425, USA.
| |
Collapse
|
21
|
Ornitz DM, Legeai-Mallet L. Achondroplasia: Development, pathogenesis, and therapy. Dev Dyn 2017; 246:291-309. [PMID: 27987249 DOI: 10.1002/dvdy.24479] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/04/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022] Open
Abstract
Autosomal dominant mutations in fibroblast growth factor receptor 3 (FGFR3) cause achondroplasia (Ach), the most common form of dwarfism in humans, and related chondrodysplasia syndromes that include hypochondroplasia (Hch), severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN), and thanatophoric dysplasia (TD). FGFR3 is expressed in chondrocytes and mature osteoblasts where it functions to regulate bone growth. Analysis of the mutations in FGFR3 revealed increased signaling through a combination of mechanisms that include stabilization of the receptor, enhanced dimerization, and enhanced tyrosine kinase activity. Paradoxically, increased FGFR3 signaling profoundly suppresses proliferation and maturation of growth plate chondrocytes resulting in decreased growth plate size, reduced trabecular bone volume, and resulting decreased bone elongation. In this review, we discuss the molecular mechanisms that regulate growth plate chondrocytes, the pathogenesis of Ach, and therapeutic approaches that are being evaluated to improve endochondral bone growth in people with Ach and related conditions. Developmental Dynamics 246:291-309, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laurence Legeai-Mallet
- Imagine Institute, Inserm U1163, Université Paris Descartes, Service de Génétique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| |
Collapse
|
22
|
Effects of In Utero Thyroxine Exposure on Murine Cranial Suture Growth. PLoS One 2016; 11:e0167805. [PMID: 27959899 PMCID: PMC5154521 DOI: 10.1371/journal.pone.0167805] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/21/2016] [Indexed: 01/05/2023] Open
Abstract
Large scale surveillance studies, case studies, as well as cohort studies have identified the influence of thyroid hormones on calvarial growth and development. Surveillance data suggests maternal thyroid disorders (hyperthyroidism, hypothyroidism with pharmacological replacement, and Maternal Graves Disease) are linked to as much as a 2.5 fold increased risk for craniosynostosis. Craniosynostosis is the premature fusion of one or more calvarial growth sites (sutures) prior to the completion of brain expansion. Thyroid hormones maintain proper bone mineral densities by interacting with growth hormone and aiding in the regulation of insulin like growth factors (IGFs). Disruption of this hormonal control of bone physiology may lead to altered bone dynamics thereby increasing the risk for craniosynostosis. In order to elucidate the effect of exogenous thyroxine exposure on cranial suture growth and morphology, wild type C57BL6 mouse litters were exposed to thyroxine in utero (control = no treatment; low ~167 ng per day; high ~667 ng per day). Thyroxine exposed mice demonstrated craniofacial dysmorphology (brachycranic). High dose exposed mice showed diminished area of the coronal and widening of the sagittal sutures indicative of premature fusion and compensatory growth. Presence of thyroid receptors was confirmed for the murine cranial suture and markers of proliferation and osteogenesis were increased in sutures from exposed mice. Increased Htra1 and Igf1 gene expression were found in sutures from high dose exposed individuals. Pathways related to the HTRA1/IGF axis, specifically Akt and Wnt, demonstrated evidence of increased activity. Overall our data suggest that maternal exogenous thyroxine exposure can drive calvarial growth alterations and altered suture morphology.
Collapse
|
23
|
Abstract
The skeleton is an exquisitely sensitive and archetypal T3-target tissue that demonstrates the critical role for thyroid hormones during development, linear growth, and adult bone turnover and maintenance. Thyrotoxicosis is an established cause of secondary osteoporosis, and abnormal thyroid hormone signaling has recently been identified as a novel risk factor for osteoarthritis. Skeletal phenotypes in genetically modified mice have faithfully reproduced genetic disorders in humans, revealing the complex physiological relationship between centrally regulated thyroid status and the peripheral actions of thyroid hormones. Studies in mutant mice also established the paradigm that T3 exerts anabolic actions during growth and catabolic effects on adult bone. Thus, the skeleton represents an ideal physiological system in which to characterize thyroid hormone transport, metabolism, and action during development and adulthood and in response to injury. Future analysis of T3 action in individual skeletal cell lineages will provide new insights into cell-specific molecular mechanisms and may ultimately identify novel therapeutic targets for chronic degenerative diseases such as osteoporosis and osteoarthritis. This review provides a comprehensive analysis of the current state of the art.
Collapse
Affiliation(s)
- J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom
| |
Collapse
|
24
|
Klein Hesselink E, Links T. Radioiodine Treatment and Thyroid Hormone Suppression Therapy for Differentiated Thyroid Carcinoma: Adverse Effects Support the Trend toward Less Aggressive Treatment for Low-Risk Patients. Eur Thyroid J 2015; 4:82-92. [PMID: 26279993 PMCID: PMC4521066 DOI: 10.1159/000432397] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/18/2015] [Indexed: 12/18/2022] Open
Abstract
Over the past decades, the incidence of differentiated thyroid carcinoma (DTC) has steadily increased, with especially a growing number of low-risk patients. Whereas DTC used to be treated rather aggressively, it is now acknowledged that aggressive treatment does not affect outcome for low-risk patients and that it can induce adverse effects. In this review an overview of the most clinically relevant adverse effects of radioiodine treatment and thyroid hormone suppression therapy (THST) is presented, and the trend toward less aggressive treatment for low-risk patients is outlined. Salivary gland dysfunction occurs in roughly 30% of patients, and is probably due to the concentration of radioiodine in the salivary glands by the sodium/iodide symporter. Beta radiation from radioiodine can result in sialoadenitis and eventually fibrosis and loss of salivary function. Furthermore, patients can experience bone marrow dysfunction following radioiodine treatment. Although this is in general subclinical and transient, patients that receive very high cumulative radioiodine doses may be at risk for more severe bone marrow dysfunction. THST can induce adverse cardiovascular effects in patients with DTC, such as diastolic and systolic dysfunction, and also adverse vascular and prothrombotic effects have been described. Finally, the effects of THST on bone formation and resorption are outlined; especially postmenopausal women with DTC on THST seem to be at risk of bone loss. In the past years, advances have been made in preventing low-risk patients from being overtreated. Improved biomarkers are still needed to further optimize risk stratification and personalize medicine.
Collapse
Affiliation(s)
| | - T.P. Links
- *Prof. Dr. T.P. Links, University of Groningen, University Medical Center Groningen, Department of Endocrinology, HPC AA31, PO Box 30.001, NL-9700 RB Groningen (The Netherlands), E-Mail
| |
Collapse
|
25
|
|
26
|
Zhang J, Chang JR, Duan XH, Yu YR, Zhang BH. Thyroid hormone attenuates vascular calcification induced by vitamin D3 plus nicotine in rats. Calcif Tissue Int 2015; 96:80-7. [PMID: 25416842 DOI: 10.1007/s00223-014-9934-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 11/17/2014] [Indexed: 01/08/2023]
Abstract
Thyroid hormones (THs) including thyroxine (T4) and triiodothyronine (T3) play critical roles in bone remodeling. However, the role and mechanism of THs in vascular calcification (VC) have been unclear. To explore the pathophysiological roles of T3 on VC, we investigated the changes in plasma and aortas of THs concentrations and the effect of T3 on rat VC induced by vitamin D3 plus nicotine (VDN). VDN-treated rat showed decreased plasma T3 content, increased vascular calcium deposition, and alkaline phosphatase (ALP) activity. Administration of T3 (0.2 mg/kg body weight IP) for 10 days greatly reduced vascular calcium deposition and ALP activity in calcified rat aortas when compared with controls. Concurrently, the loss of smooth muscle lineage markers α-actin and SM22a was restored, and the increased bone-associated molecules, such as runt-related transcription factor2 (Runx2), Osterix, and osteopontin (OPN) levels in calcified aorta, were reduced by administration of T3. The suppression of klotho in calcified rat aorta was restored by T3. Methimazole (400 mg/L) blocked the beneficial effect of T3 on VC. These results suggested that T3 can inhibit VC development.
Collapse
Affiliation(s)
- Jing Zhang
- School of P.E. and Sports Science, Beijing Normal University, Beijing, 100875, China,
| | | | | | | | | |
Collapse
|
27
|
Ortiga-Carvalho TM, Sidhaye AR, Wondisford FE. Thyroid hormone receptors and resistance to thyroid hormone disorders. Nat Rev Endocrinol 2014; 10:582-91. [PMID: 25135573 PMCID: PMC4578869 DOI: 10.1038/nrendo.2014.143] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Thyroid hormone action is predominantly mediated by thyroid hormone receptors (THRs), which are encoded by the thyroid hormone receptor α (THRA) and thyroid hormone receptor β (THRB) genes. Patients with mutations in THRB present with resistance to thyroid hormone β (RTHβ), which is a disorder characterized by elevated levels of thyroid hormone, normal or elevated levels of TSH and goitre. Mechanistic insights about the contributions of THRβ to various processes, including colour vision, development of the cochlea and the cerebellum, and normal functioning of the adult liver and heart, have been obtained by either introducing human THRB mutations into mice or by deletion of the mouse Thrb gene. The introduction of the same mutations that mimic human THRβ alterations into the mouse Thra and Thrb genes resulted in distinct phenotypes, which suggests that THRA and THRB might have non-overlapping functions in human physiology. These studies also suggested that THRA mutations might not be lethal. Seven patients with mutations in THRα have since been described. These patients have RTHα and presented with major abnormalities in growth and gastrointestinal function. The hypothalamic-pituitary-thyroid axis in these individuals is minimally affected, which suggests that the central T3 feedback loop is not impaired in patients with RTHα, in stark contrast to patients with RTHβ.
Collapse
Affiliation(s)
- Tânia M Ortiga-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, S/N, Cidade Universitária, 21941-902, Rio de Janeiro, Brazil
| | - Aniket R Sidhaye
- Departments of Paediatrics and Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, CMSC 10-113, Baltimore, MD 21287, USA
| | - Fredric E Wondisford
- Departments of Paediatrics and Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, CMSC 10-113, Baltimore, MD 21287, USA
| |
Collapse
|
28
|
Fonseca TL, Teixeira MBCG, Miranda-Rodrigues M, Silva MV, Martins GM, Costa CC, Arita DY, Perez JD, Casarini DE, Brum PC, Gouveia CHA. Thyroid hormone interacts with the sympathetic nervous system to modulate bone mass and structure in young adult mice. Am J Physiol Endocrinol Metab 2014; 307:E408-18. [PMID: 25005498 DOI: 10.1152/ajpendo.00643.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To investigate whether thyroid hormone (TH) interacts with the sympathetic nervous system (SNS) to modulate bone mass and structure, we studied the effects of daily T3 treatment in a supraphysiological dose for 12 wk on the bone of young adult mice with chronic sympathetic hyperactivity owing to double-gene disruption of adrenoceptors that negatively regulate norepinephrine release, α(2A)-AR, and α(2C)-AR (α(2A/2C)-AR(-/-) mice). As expected, T3 treatment caused a generalized decrease in the areal bone mineral density (aBMD) of WT mice (determined by DEXA), followed by deleterious effects on the trabecular and cortical bone microstructural parameters (determined by μCT) of the femur and vertebra and on the biomechanical properties (maximum load, ultimate load, and stiffness) of the femur. Surprisingly, α(2A/2C)-AR(-/-) mice were resistant to most of these T3-induced negative effects. Interestingly, the mRNA expression of osteoprotegerin, a protein that limits osteoclast activity, was upregulated and downregulated by T3 in the bone of α(2A/2C)-AR(-/-) and WT mice, respectively. β1-AR mRNA expression and IGF-I serum levels, which exert bone anabolic effects, were increased by T3 treatment only in α(2A/2C)-AR(-/-) mice. As expected, T3 inhibited the cell growth of calvaria-derived osteoblasts isolated from WT mice, but this effect was abolished or reverted in cells isolated from KO mice. Collectively, these findings support the hypothesis of a TH-SNS interaction to control bone mass and structure of young adult mice and suggests that this interaction may involve α2-AR signaling. Finally, the present findings offer new insights into the mechanisms through which TH regulates bone mass, structure, and physiology.
Collapse
Affiliation(s)
- Tatiana L Fonseca
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marilia B C G Teixeira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Marcos V Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gisele M Martins
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cristiane C Costa
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Danielle Y Arita
- Department of Internal Medicine, Renal Division, Federal University of São Paulo School of Medicine, São Paulo, Brazil; and
| | - Juliana D Perez
- Department of Internal Medicine, Renal Division, Federal University of São Paulo School of Medicine, São Paulo, Brazil; and
| | - Dulce E Casarini
- Department of Internal Medicine, Renal Division, Federal University of São Paulo School of Medicine, São Paulo, Brazil; and
| | - Patricia C Brum
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Cecilia H A Gouveia
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil;
| |
Collapse
|
29
|
Desjardin C, Charles C, Benoist-Lasselin C, Riviere J, Gilles M, Chassande O, Morgenthaler C, Laloé D, Lecardonnel J, Flamant F, Legeai-Mallet L, Schibler L. Chondrocytes play a major role in the stimulation of bone growth by thyroid hormone. Endocrinology 2014; 155:3123-35. [PMID: 24914940 DOI: 10.1210/en.2014-1109] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormone (T3) is required for postnatal skeletal growth. It exerts its effect by binding to nuclear receptors, TRs including TRα1 and TRβ1, which are present in most cell types. These cell types include chondrocytes and osteoblasts, the interactions of which are known to regulate endochondral bone formation. In order to analyze the respective functions of T3 stimulation in chondrocytes and osteoblasts during postnatal growth, we use Cre/loxP recombination to express a dominant-negative TRα1(L400R) mutant receptor in a cell-specific manner. Phenotype analysis revealed that inhibiting T3 response in chondrocytes is sufficient to reproduce the defects observed in hypothyroid mice, not only for cartilage maturation, but also for ossification and mineralization. TRα1(L400R) in chondrocytes also results in skull deformation. In the meantime, TRα1(L400R) expression in mature osteoblasts has no visible effect. Transcriptome analysis identifies a number of changes in gene expression induced by TRα1(L400R) in cartilage. These changes suggest that T3 normally cross talks with several other signaling pathways to promote chondrocytes proliferation, differentiation, and skeletal growth.
Collapse
Affiliation(s)
- Clémence Desjardin
- Institut National de la Recherche Agronomique (INRA) (C.D., J.R., M.G., C.M., D.L., J.L., L.S.), UMR1313, Biologie Intégrative et Génétique Animale, Jouy-en-Josas, France; Centre National de la Recherche Scientifique (CNRS) UMR 5242 (C.C.), ENS Lyon, Institut de Génomique Fonctionnelle, Université de Lyon, Lyon, France; Institut Imagine (C.B.-L., L.L.-G.) Institut National de la Santé et de la Recherche Medicale, U1163, Université Paris Descartes, 75015 Paris, France; University of Bordeaux (O.C.), U1026, Bioingénierie Tissulaire, Bordeaux, France; and Institut de Génomique Fonctionnelle de Lyon (F.F.), Université de Lyon, CNRS, INRA, École Normale Supérieure de Lyon, 69364 Lyon Cedex 07, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bianco AC, Anderson G, Forrest D, Galton VA, Gereben B, Kim BW, Kopp PA, Liao XH, Obregon MJ, Peeters RP, Refetoff S, Sharlin DS, Simonides WS, Weiss RE, Williams GR. American Thyroid Association Guide to investigating thyroid hormone economy and action in rodent and cell models. Thyroid 2014; 24:88-168. [PMID: 24001133 PMCID: PMC3887458 DOI: 10.1089/thy.2013.0109] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND An in-depth understanding of the fundamental principles that regulate thyroid hormone homeostasis is critical for the development of new diagnostic and treatment approaches for patients with thyroid disease. SUMMARY Important clinical practices in use today for the treatment of patients with hypothyroidism, hyperthyroidism, or thyroid cancer are the result of laboratory discoveries made by scientists investigating the most basic aspects of thyroid structure and molecular biology. In this document, a panel of experts commissioned by the American Thyroid Association makes a series of recommendations related to the study of thyroid hormone economy and action. These recommendations are intended to promote standardization of study design, which should in turn increase the comparability and reproducibility of experimental findings. CONCLUSIONS It is expected that adherence to these recommendations by investigators in the field will facilitate progress towards a better understanding of the thyroid gland and thyroid hormone dependent processes.
Collapse
Affiliation(s)
- Antonio C. Bianco
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, Florida
| | - Grant Anderson
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota Duluth, Duluth, Minnesota
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Valerie Anne Galton
- Department of Physiology and Neurobiology, Dartmouth Medical School, Lebanon, New Hampshire
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Brian W. Kim
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, Florida
| | - Peter A. Kopp
- Division of Endocrinology, Metabolism, and Molecular Medicine, and Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao Hui Liao
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - Maria Jesus Obregon
- Institute of Biomedical Investigation (IIB), Spanish National Research Council (CSIC) and Autonomous University of Madrid, Madrid, Spain
| | - Robin P. Peeters
- Division of Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Samuel Refetoff
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - David S. Sharlin
- Department of Biological Sciences, Minnesota State University, Mankato, Minnesota
| | - Warner S. Simonides
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Roy E. Weiss
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - Graham R. Williams
- Department of Medicine, Imperial College London, Hammersmith Campus, London, United Kingdom
| |
Collapse
|
31
|
Abstract
Thyroid hormone receptors (TR) are prototypes of nuclear transcription factors that regulate the expression of target genes. These receptors play an important role in many physiological processes. Moreover, a dysfunction of these proteins is often implicated in several human diseases and malignancies. Here we report genetic variations and alterations of the TRs that have been described in the literature as well as their potential role in the development of some human diseases including cancers. The functional effects of some mutations and polymorphisms in TRs on disease susceptibility, especially on cancer risk, are now established. Therefore, further investigations are needed in order to use these receptors as therapeutic targets or as biological markers to decide on appropriate forms of treatment.
Collapse
Affiliation(s)
- Maha Rebaï
- Molecular and Cellular Diagnosis Processes, Centre of Biotechnology of Sfax, University of Sfax, Route Sidi Mansour, PO Box 1177, 3018 Sfax, Tunisia
| | | | | |
Collapse
|
32
|
Kim HY, Mohan S. Role and Mechanisms of Actions of Thyroid Hormone on the Skeletal Development. Bone Res 2013; 1:146-61. [PMID: 26273499 DOI: 10.4248/br201302004] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 04/28/2013] [Indexed: 01/03/2023] Open
Abstract
The importance of the thyroid hormone axis in the regulation of skeletal growth and maintenance has been well established from clinical studies involving patients with mutations in proteins that regulate synthesis and/or actions of thyroid hormone. Data from genetic mouse models involving disruption and overexpression of components of the thyroid hormone axis also provide direct support for a key role for thyroid hormone in the regulation of bone metabolism. Thyroid hormone regulates proliferation and/or differentiated actions of multiple cell types in bone including chondrocytes, osteoblasts and osteoclasts. Thyroid hormone effects on the target cells are mediated via ligand-inducible nuclear receptors/transcription factors, thyroid hormone receptor (TR) α and β, of which TRα seems to be critically important in regulating bone cell functions. In terms of mechanisms for thyroid hormone action, studies suggest that thyroid hormone regulates a number of key growth factor signaling pathways including insulin-like growth factor-I, parathyroid hormone related protein, fibroblast growth factor, Indian hedgehog and Wnt to influence skeletal growth. In this review we describe findings from various genetic mouse models and clinical mutations of thyroid hormone signaling related mutations in humans that pertain to the role and mechanism of action of thyroid hormone in the regulation of skeletal growth and maintenance.
Collapse
Affiliation(s)
- Ha-Young Kim
- Musculoskeletal Disease Center, Loma Linda VA HealthCare System , Loma Linda, CA 92357, USA ; Departments of Medicine, Loma Linda University , Loma Linda, CA 92354, USA ; Division of Endocrinology, Department of Internal Medicine, Wonkwang University Sanbon Hospital , Gunpo, Gyeonggi, Korea
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA HealthCare System , Loma Linda, CA 92357, USA ; Departments of Medicine, Loma Linda University , Loma Linda, CA 92354, USA
| |
Collapse
|
33
|
Shoemaker TJ, Kono T, Mariash CN, Evans-Molina C. Thyroid hormone analogues for the treatment of metabolic disorders: new potential for unmet clinical needs? Endocr Pract 2013; 18:954-64. [PMID: 22784847 DOI: 10.4158/ep12086.ra] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To provide a comprehensive review of the discovery and development of selective thyroid hormone receptor agonists and provide a discussion of their use in hyperlipidemia, obesity, and type 2 diabetes mellitus. METHODS Preclinical and clinical English language literature from 1930 to present was reviewed and thematically summarized. RESULTS Human trials have shown that thyroid hormone receptor β (TRβ) agonists effectively lower low-density lipoprotein, triglycerides, apolipoprotein B, and lipoprotein(a) levels. In preclinical studies, TRβ agonists enhance reverse cholesterol transport and decrease atherosclerosis in selected models. While animal data suggest these drugs may have additional utility to modulate weight and improve glucose homeostasis, human studies have not shown similar results. CONCLUSION TRβ agonists are a novel therapeutic class for lipid management. Their mechanism of action for lipid lowering is distinct from statin drugs, suggesting a strong possibility for synergistic effects with combined therapy. The long-term effects of these drugs on cardiovascular outcomes, however, are unknown. Recently, the development of the most promising agent in this class, eprotirome, was halted over toxicology concerns following long-term canine studies. Consequently, the future of contemporary TRβ agonists is unclear. The creation of a next generation of TRβ agonists that provide additional tissue specific effects or bind TRβ with even higher selectivity may lead to improved safety and efficacy and allow for their application to other metabolic disorders like obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Timothy J Shoemaker
- Department of Medicine, Indiana University School of Medicine Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
34
|
Wu SM, Cheng WL, Lin CD, Lin KH. Thyroid hormone actions in liver cancer. Cell Mol Life Sci 2013; 70:1915-36. [PMID: 22955376 PMCID: PMC11113324 DOI: 10.1007/s00018-012-1146-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 08/06/2012] [Accepted: 08/20/2012] [Indexed: 12/13/2022]
Abstract
The thyroid hormone 3,3',5-triiodo-L-thyronine (T3) mediates several physiological processes, including embryonic development, cellular differentiation, metabolism, and the regulation of cell proliferation. Thyroid hormone receptors (TRs) generally act as heterodimers with the retinoid X receptor (RXR) to regulate target genes. In addition to their developmental and metabolic functions, TRs have been shown to play a tumor suppressor role, suggesting that their aberrant expression can lead to tumor transformation. Conversely, recent reports have shown an association between overexpression of wild-type TRs and tumor metastasis. Signaling crosstalk between T3/TR and other pathways or specific TR coregulators appear to affect tumor development. Since TR actions are complex as well as cell context-, tissue- and time-specific, aberrant expression of the various TR isoforms has different effects during diverse tumorigenesis. Therefore, elucidation of the T3/TR signaling mechanisms in cancers should facilitate the identification of novel therapeutic targets. This review provides a summary of recent studies focusing on the role of TRs in hepatocellular carcinomas (HCCs).
Collapse
Affiliation(s)
- Sheng-Ming Wu
- Department of Biochemistry, College of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, 333 Taiwan
| | - Wan-Li Cheng
- Department of Biochemistry, College of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, 333 Taiwan
| | - Crystal D. Lin
- Pre-med Program, Pacific Union College, Angwin, CA 94508 USA
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, 333 Taiwan
| |
Collapse
|
35
|
Williams GR. Thyroid hormone actions in cartilage and bone. Eur Thyroid J 2013; 2:3-13. [PMID: 24783033 PMCID: PMC3821494 DOI: 10.1159/000345548] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 11/01/2012] [Indexed: 11/19/2022] Open
Abstract
Thyroid hormones exert widespread and complex actions in almost all tissues during development, throughout childhood and in adults. The skeleton is an important T3-target tissue that exemplifies these processes, and yet understanding of the specific cellular and molecular mechanisms of T3 action in bone and cartilage remains incomplete. Here, the skeleton is considered as a T3-target tissue. The actions of thyroid hormones during skeletal development and in chondrocytes and growth plate cartilage during post-natal linear growth are outlined. The physiological importance of these actions are discussed in relation to patients with autosomal dominant mutations in genes encoding the thyroid hormone receptors TRα1 and TRβ, and in mice harbouring deletions or mutations of the orthologous genes. The role of thyroid hormones and the control of T3 action in bone turnover and maintenance are also outlined, and T3 action in bone-forming osteoblasts and bone-resorbing osteoclasts discussed. The physiological and functional consequences of T3 action in bone are considered in relation to mutant mouse models and to effects on bone mineral density and fracture susceptibility in humans. Finally, new studies identifying a putative role for thyroid hormone metabolism in articular cartilage maintenance and the pathogenesis of osteoarthritis are considered. The pharmacological context of these new findings is discussed, emphasising the importance of this emerging field of study in thyroid hormone pathophysiology.
Collapse
Affiliation(s)
- Graham R. Williams
- Molecular Endocrinology Group, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
36
|
Gorka J, Taylor-Gjevre RM, Arnason T. Metabolic and clinical consequences of hyperthyroidism on bone density. Int J Endocrinol 2013; 2013:638727. [PMID: 23970897 PMCID: PMC3736466 DOI: 10.1155/2013/638727] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 06/25/2013] [Indexed: 11/23/2022] Open
Abstract
In 1891, Von Recklinghausen first established the association between the development of osteoporosis in the presence of overt hyperthyroidism. Subsequent reports have demonstrated that BMD loss is common in frank hyperthyroidism, and, to a lesser extent, in subclinical presentations. With the introduction of antithyroid medication in the 1940s to control biochemical hyperthyroidism, the accompanying bone disease became less clinically apparent as hyperthyroidism was more successfully treated medically. Consequently, the impact of the above normal thyroid hormones in the pathogenesis of osteoporosis may be presently underrecognized due to the widespread effective treatments. This review aims to present the current knowledge of the consequences of hyperthyroidism on bone metabolism. The vast number of recent papers touching on this topic highlights the recognized impact of this common medical condition on bone health. Our focus in this review was to search for answers to the following questions. What is the mechanisms of action of thyroid hormones on bone metabolism? What are the clinical consequences of hyperthyroidism on BMD and fracture risk? What differences are there between men and women with thyroid disease and how does menopause change the clinical outcomes? Lastly, we report how different treatments for hyperthyroidism benefit thyroid hormone-induced osteoporosis.
Collapse
Affiliation(s)
- Jagoda Gorka
- Department of Medicine, University of Saskatchewan, Saskatoon, Canada S7N 0W8
| | - Regina M. Taylor-Gjevre
- Division of Rheumatology, Department of Medicine, University of Saskatchewan, Saskatoon, Canada S7N 0W8
| | - Terra Arnason
- Division of Endocrinology and Metabolism, Department of Medicine, University of Saskatchewan, Saskatoon, Canada S7N 0W8
- *Terra Arnason:
| |
Collapse
|
37
|
Bassett JHD, Logan JG, Boyde A, Cheung MS, Evans H, Croucher P, Sun XY, Xu S, Murata Y, Williams GR. Mice lacking the calcineurin inhibitor Rcan2 have an isolated defect of osteoblast function. Endocrinology 2012; 153:3537-48. [PMID: 22593270 DOI: 10.1210/en.2011-1814] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Calcineurin-nuclear factor of activated T cells signaling controls the differentiation and function of osteoclasts and osteoblasts, and regulator of calcineurin-2 (Rcan2) is a physiological inhibitor of this pathway. Rcan2 expression is regulated by T(3), which also has a central role in skeletal development and bone turnover. To investigate the role of Rcan2 in bone development and maintenance, we characterized Rcan2(-/-) mice and determined its skeletal expression in T(3) receptor (TR) knockout and thyroid-manipulated mice. Rcan2(-/-) mice had normal linear growth but displayed delayed intramembranous ossification, impaired cortical bone formation, and reduced bone mineral accrual during development as well as increased mineralization of adult bone. These abnormalities resulted from an isolated defect in osteoblast function and are similar to skeletal phenotypes of mice lacking the type 2 deiodinase thyroid hormone activating enzyme or with dominant-negative mutations of TRα, the predominant TR isoform in bone. Rcan2 mRNA was expressed in primary osteoclasts and osteoblasts, and its expression in bone was differentially regulated in TRα and TRβ knockout and thyroid-manipulated mice. However, in primary osteoblast cultures, T(3) treatment did not affect Rcan2 mRNA expression or nuclear factor of activated T cells c1 expression and phosphorylation. Overall, these studies establish that Rcan2 regulates osteoblast function and its expression in bone is regulated by thyroid status in vivo.
Collapse
Affiliation(s)
- J H Duncan Bassett
- Molecular Endocrinology Group, Department of Medicine, Imperial College London, Hammersmith Campus, London W12 0NN, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Euthyroid status is essential for normal skeletal development and the maintenance of adult bone structure and strength. Established thyrotoxicosis has long been recognised as a cause of high bone turnover osteoporosis and fracture but more recent studies have suggested that subclinical hyperthyroidism and long-term suppressive doses of thyroxine (T4) may also result in decreased bone mineral density (BMD) and an increased risk of fragility fracture, particularly in postmenopausal women. Furthermore, large population studies of euthyroid individuals have demonstrated that a hypothalamic-pituitary-thyroid axis set point at the upper end of the normal reference range is associated with reduced BMD and increased fracture susceptibility. Despite these findings, the cellular and molecular mechanisms of thyroid hormone action in bone remain controversial and incompletely understood. In this review, we discuss the role of thyroid hormones in bone and the skeletal consequences of hyperthyroidism.
Collapse
Affiliation(s)
- Jonathan J Nicholls
- Molecular Endocrinology Group, Department of Medicine, Imperial College London, Hammersmith Campus, Room 7N2b, Commonwealth Building, Du Cane Road, London W12 0NN, UK
| | | | | | | |
Collapse
|
39
|
Wojcicka A, Bassett JHD, Williams GR. Mechanisms of action of thyroid hormones in the skeleton. Biochim Biophys Acta Gen Subj 2012; 1830:3979-86. [PMID: 22634735 DOI: 10.1016/j.bbagen.2012.05.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 04/19/2012] [Accepted: 05/18/2012] [Indexed: 11/25/2022]
Abstract
BACKGROUND Thyroid hormones regulate skeletal development, acquisition of peak bone mass and adult bone maintenance. Abnormal thyroid status during childhood disrupts bone maturation and linear growth, while in adulthood it results in altered bone remodeling and an increased risk of fracture SCOPE OF REVIEW This review considers the cellular effects and molecular mechanisms of thyroid hormone action in the skeleton. Human clinical and population data are discussed in relation to the skeletal phenotypes of a series of genetically modified mouse models of disrupted thyroid hormone signaling. MAJOR CONCLUSIONS Euthyroid status is essential for normal bone development and maintenance. Major thyroid hormone actions in skeletal cells are mediated by thyroid hormone receptor α (TRα) and result in anabolic responses during growth and development but catabolic effects in adulthood. These homeostatic responses to thyroid hormone are locally regulated in individual skeletal cell types by the relative activities of the type 2 and 3 iodothyronine deiodinases, which control the supply of the active thyroid hormone 3,5,3'-L-triiodothyronine (T3) to its receptor. GENERAL SIGNIFICANCE Population studies indicate that both thyroid hormone deficiency and excess are associated with an increased risk of fracture. Understanding the cellular and molecular basis of T3 action in skeletal cells will lead to the identification of new targets to regulate bone turnover and mineralization in the prevention and treatment of osteoporosis. This article is part of a Special Issue entitled Thyroid hormone signaling.
Collapse
Affiliation(s)
- Anna Wojcicka
- The Medical Centre of Postgraduate Education, Department of Biochemistry and Molecular Biology, ul.Marymoncka 99/103, 01-813 Warsaw, Poland
| | | | | |
Collapse
|
40
|
Waung JA, Bassett JHD, Williams GR. Thyroid hormone metabolism in skeletal development and adult bone maintenance. Trends Endocrinol Metab 2012; 23:155-62. [PMID: 22169753 DOI: 10.1016/j.tem.2011.11.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 10/26/2011] [Accepted: 11/03/2011] [Indexed: 02/01/2023]
Abstract
Metabolism of thyroid hormones by the type 2 and type 3 iodothyronine deiodinases (D2, D3) in T3-responsive target cells is a sophisticated mechanism that helps to maintain local T3 concentrations and facilitates T3 action in a cell-specific manner that is independent of circulating thyroid hormone concentrations. Recent findings have demonstrated an essential physiological role for the thyroid hormone-activating enzyme D2 in the optimization of bone mineralization and strength. Emerging population studies have also identified the genes encoding D2 and the thyroid hormone-inactivating enzyme D3 as susceptibility loci for osteoarthritis. These new data reveal an essential role for the local control of T3 availability in osteoblasts and chondrocytes during maintenance and repair of bone and cartilage.
Collapse
Affiliation(s)
- Julian A Waung
- Molecular Endocrinology Group, Department of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | | | | |
Collapse
|
41
|
Thyroid hormone receptors, cell growth and differentiation. Biochim Biophys Acta Gen Subj 2012; 1830:3908-16. [PMID: 22484490 DOI: 10.1016/j.bbagen.2012.03.012] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 03/01/2012] [Accepted: 03/20/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Tissue homeostasis depends on the balance between cell proliferation and differentiation. Thyroid hormones (THs), through binding to their nuclear receptors, can regulate the expression of many genes involved in cell cycle control and cellular differentiation. This can occur by direct transcriptional regulation or by modulation of the activity of different signaling pathways. SCOPE OF REVIEW In this review we will summarize the role of the different receptor isoforms in growth and maturation of selected tissues and organs. We will focus on mammalian tissues, and therefore we will not address the fundamental role of the THs during amphibian metamorphosis. MAJOR CONCLUSIONS The actions of THs are highly pleiotropic, affecting many tissues at different developmental stages. As a consequence, their effects on proliferation and differentiation are highly heterogeneous depending on the cell type, the cellular context, and the developmental or transformation status. Both during development and in the adult, stem cells are essential for proper organ formation, maintenance and regeneration. Recent evidence suggests that some of the actions of the thyroid hormone receptors could be secondary to regulation of stem/progenitor cell function. Here we will also include the latest knowledge on the role of these receptors in proliferation and differentiation of embryonic and adult stem cells. GENERAL SIGNIFICANCE The thyroid hormone receptors are potent regulators of proliferation and differentiation of many cell types. This can explain the important role of the thyroid hormones and their receptors in key processes such as growth, development, tissue homeostasis or cancer. This article is part of a Special Issue entitled Thyroid hormone signalling.
Collapse
|
42
|
O'Shea PJ, Kim DW, Logan JG, Davis S, Walker RL, Meltzer PS, Cheng SY, Williams GR. Advanced bone formation in mice with a dominant-negative mutation in the thyroid hormone receptor β gene due to activation of Wnt/β-catenin protein signaling. J Biol Chem 2012; 287:17812-17822. [PMID: 22442145 DOI: 10.1074/jbc.m111.311464] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Thyroid hormone (T(3)) acts in chondrocytes and bone-forming osteoblasts to control bone development and maintenance, but the signaling pathways mediating these effects are poorly understood. Thrb(PV/PV) mice have a severely impaired pituitary-thyroid axis and elevated thyroid hormone levels due to a dominant-negative mutant T(3) receptor (TRβ(PV)) that cannot bind T(3) and interferes with the actions of wild-type TR. Thrb(PV/PV) mice have accelerated skeletal development due to unknown mechanisms. We performed microarray studies in primary osteoblasts from wild-type mice and Thrb(PV/PV) mice. Activation of the canonical Wnt signaling in Thrb(PV/PV) mice was confirmed by in situ hybridization analysis of Wnt target gene expression in bone during postnatal growth. By contrast, T(3) treatment inhibited Wnt signaling in osteoblastic cells, suggesting that T(3) inhibits the Wnt pathway by facilitating proteasomal degradation of β-catenin and preventing its accumulation in the nucleus. Activation of the Wnt pathway in Thrb(PV/PV) mice, however, results from a gain of function for TRβ(PV) that stabilizes β-catenin despite the presence of increased thyroid hormone levels. These studies demonstrate novel interactions between T(3) and Wnt signaling pathways in the regulation of skeletal development and bone formation.
Collapse
Affiliation(s)
- Patrick J O'Shea
- Gene Regulation Section, Laboratory of Molecular Biology, NCI, National Institutes of Health, Bethesda, Maryland 20892; Molecular Endocrinology Group, Department of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Dong Wook Kim
- Gene Regulation Section, Laboratory of Molecular Biology, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - John G Logan
- Molecular Endocrinology Group, Department of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Sean Davis
- Molecular Genetics Section, Genetics Branch, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Robert L Walker
- Molecular Genetics Section, Genetics Branch, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Paul S Meltzer
- Molecular Genetics Section, Genetics Branch, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Sheue-Yann Cheng
- Gene Regulation Section, Laboratory of Molecular Biology, NCI, National Institutes of Health, Bethesda, Maryland 20892.
| | - Graham R Williams
- Molecular Endocrinology Group, Department of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom.
| |
Collapse
|
43
|
Resistance to thyroid hormone is modulated in vivo by the nuclear receptor corepressor (NCOR1). Proc Natl Acad Sci U S A 2011; 108:17462-7. [PMID: 21987803 DOI: 10.1073/pnas.1107474108] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mutations in the ligand-binding domain of the thyroid hormone receptor β (TRβ) lead to resistance to thyroid hormone (RTH). These TRβ mutants function in a dominant-negative fashion to interfere with the transcription activity of wild-type thyroid hormone receptors (TRs), leading to dysregulation of the pituitary-thyroid axis and resistance in peripheral tissues. The molecular mechanism by which TRβ mutants cause RTH has been postulated to be an inability of the mutants to properly release the nuclear corepressors (NCORs), thereby inhibiting thyroid hormone (TH)-mediated transcription activity. To test this hypothesis in vivo, we crossed Thrb(PV) mice (a model of RTH) expressing a human TRβ mutant (PV) with mice expressing a mutant Ncor1 allele (Ncor1(ΔID) mice) that cannot recruit a TR or a PV mutant. Remarkably, in the presence of NCOR1ΔID, the abnormally elevated thyroid-stimulating hormone and TH levels found in Thrb(PV) mice were modestly but significantly corrected. Furthermore, thyroid hyperplasia, weight loss, and other hallmarks of RTH were also partially reverted in mice expressing NCOR1ΔID. Taken together, these data suggest that the aberrant recruitment of NCOR1 by RTH TRβ mutants leads to clinical RTH in humans. The present study suggests that therapies aimed at the TR-NCOR1 interaction or its downstream actions could be tested as potential targets in treating RTH.
Collapse
|
44
|
Santiago LA, Santiago DA, Faustino LC, Cordeiro A, Lisboa PC, Wondisford FE, Pazos-Moura CC, Ortiga-Carvalho TM. The Δ337T mutation on the TRβ causes alterations in growth, adiposity, and hepatic glucose homeostasis in mice. J Endocrinol 2011; 211:39-46. [PMID: 21746794 DOI: 10.1530/joe-11-0194] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mice bearing the genomic mutation Δ337T on the thyroid hormone receptor β (TRβ) gene present the classical signs of resistance to thyroid hormone (TH), with high serum TH and TSH. This mutant TR is unable to bind TH, remains constitutively bound to co-repressors, and has a dominant negative effect on normal TRs. In this study, we show that homozygous (TRβΔ337T) mice for this mutation have reduced body weight, length, and body fat content, despite augmented relative food intake and relative increase in serum leptin. TRβΔ337T mice exhibited normal glycemia and were more tolerant to an i.p. glucose load accompanied by reduced insulin secretion. Higher insulin sensitivity was observed after single insulin injection, when the TRβΔ337T mice developed a profound hypoglycemia. Impaired hepatic glucose production was confirmed by the reduction in glucose generation after pyruvate administration. In addition, hepatic glycogen content was lower in homozygous TRβΔ337T mice than in wild type. Collectively, the data suggest that TRβΔ337T mice have deficient hepatic glucose production, by reduced gluconeogenesis and lower glycogen deposits. Analysis of liver gluconeogenic gene expression showed a reduction in the mRNA of phosphoenolpyruvate carboxykinase, a rate-limiting enzyme, and of peroxisome proliferator-activated receptor-γ coactivator 1α, a key transcriptional factor essential to gluconeogenesis. Reduction in both gene expressions is consistent with resistance to TH action via TRβ, reproducing a hypothyroid phenotype. In conclusion, mice carrying the Δ337T-dominant negative mutation on the TRβ are leaner, exhibit impaired hepatic glucose production, and are more sensitive to hypoglycemic effects of insulin.
Collapse
Affiliation(s)
- L A Santiago
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Monfoulet LE, Rabier B, Dacquin R, Anginot A, Photsavang J, Jurdic P, Vico L, Malaval L, Chassande O. Thyroid hormone receptor β mediates thyroid hormone effects on bone remodeling and bone mass. J Bone Miner Res 2011; 26:2036-44. [PMID: 21594896 DOI: 10.1002/jbmr.432] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Excess thyroid hormone (TH) in adults causes osteoporosis and increases fracture risk. However, the mechanisms by which TH affects bone turnover are not elucidated. In particular, the roles of thyroid hormone receptor (TR) isotypes in the mediation of TH effects on osteoblast-mediated bone formation and osteoclast-mediated bone resorption are not established. In this study we have induced experimental hypothyroidism or hyperthyroidism in adult wild-type, TRα- or TRβ-deficient mice and analyzed the effects of TH status on the structure and remodeling parameters of trabecular bone. In wild-type mice, excess TH decreased bone volume and mineralization. High TH concentrations were associated with a high bone-resorption activity, assessed by increased osteoclast surfaces and elevated concentrations of serum bone-resorption markers. Serum markers of bone formation also were higher in TH-treated mice. TRα deficiency did not prevent TH action on bone volume, bone mineralization, bone formation, or bone resorption. In contrast, TRβ deficiency blocked all the early effects of excess TH observed in wild-type mice. However, prolonged exposure to low or high TH concentrations of TRβ-deficient mice induced mild modifications of bone structure and remodeling parameters. Together our data suggest that TRβ receptors mediate the acute effects produced by transient changes of TH concentrations on bone remodeling, whereas TRα receptors mediate long-term effects of chronic alterations of TH metabolism. These data shed new light on the respective roles of TRs in the control of bone metabolism by TH.
Collapse
|
46
|
Thyroid and bone. Arch Biochem Biophys 2010; 503:129-36. [DOI: 10.1016/j.abb.2010.06.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 06/15/2010] [Accepted: 06/18/2010] [Indexed: 11/20/2022]
|
47
|
Bassett JHD, Boyde A, Howell PGT, Bassett RH, Galliford TM, Archanco M, Evans H, Lawson MA, Croucher P, St. Germain DL, Galton VA, Williams GR. Optimal bone strength and mineralization requires the type 2 iodothyronine deiodinase in osteoblasts. Proc Natl Acad Sci U S A 2010; 107:7604-9. [PMID: 20368437 PMCID: PMC2867713 DOI: 10.1073/pnas.0911346107] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hypothyroidism and thyrotoxicosis are each associated with an increased risk of fracture. Although thyroxine (T4) is the predominant circulating thyroid hormone, target cell responses are determined by local intracellular availability of the active hormone 3,5,3'-L-triiodothyronine (T3), which is generated from T4 by the type 2 deiodinase enzyme (D2). To investigate the role of locally produced T3 in bone, we characterized mice deficient in D2 (D2KO) in which the serum T3 level is normal. Bones from adult D2KO mice have reduced toughness and are brittle, displaying an increased susceptibility to fracture. This phenotype is characterized by a 50% reduction in bone formation and a generalized increase in skeletal mineralization resulting from a local deficiency of T3 in osteoblasts. These data reveal an essential role for D2 in osteoblasts in the optimization of bone strength and mineralization.
Collapse
Affiliation(s)
- J. H. Duncan Bassett
- Molecular Endocrinology Group, Division of Medicine and Medical Research Council Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London W12 0NN, United Kingdom
| | - Alan Boyde
- Oral Growth and Development, Institute of Dentistry, Bart's and London School of Medicine, Queen Mary University of London, London E1 1BB, United Kingdom
| | - Peter G. T. Howell
- Oral Growth and Development, Institute of Dentistry, Bart's and London School of Medicine, Queen Mary University of London, London E1 1BB, United Kingdom
- Division of Restorative Dental Sciences, Eastman Dental Institute, and
| | - Richard H. Bassett
- Department of Civil and Environmental Engineering, University College London, London WC1E 6BT, United Kingdom
| | - Thomas M. Galliford
- Molecular Endocrinology Group, Division of Medicine and Medical Research Council Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London W12 0NN, United Kingdom
| | - Marta Archanco
- Molecular Endocrinology Group, Division of Medicine and Medical Research Council Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London W12 0NN, United Kingdom
| | - Holly Evans
- The Mellanby Centre for Bone Research, Department of Human Metabolism, University of Sheffield, Sheffield S10 2RX, United Kingdom; and
| | - Michelle A. Lawson
- The Mellanby Centre for Bone Research, Department of Human Metabolism, University of Sheffield, Sheffield S10 2RX, United Kingdom; and
| | - Peter Croucher
- The Mellanby Centre for Bone Research, Department of Human Metabolism, University of Sheffield, Sheffield S10 2RX, United Kingdom; and
| | | | - Valerie Anne Galton
- Departments of Physiology and Medicine, Dartmouth Medical School, Lebanon, NH 03756
| | - Graham R. Williams
- Molecular Endocrinology Group, Division of Medicine and Medical Research Council Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London W12 0NN, United Kingdom
| |
Collapse
|
48
|
El‐bakry A, El‐Gareib A, Ahmed R. Comparative study of the effects of experimentally induced hypothyroidism and hyperthyroidism in some brain regions in albino rats. Int J Dev Neurosci 2010; 28:371-89. [DOI: 10.1016/j.ijdevneu.2010.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Revised: 04/07/2010] [Accepted: 04/09/2010] [Indexed: 11/17/2022] Open
Affiliation(s)
- A.M. El‐bakry
- Zoology Department, Faculty of ScienceBeni Suef UniversityEgypt
| | - A.W. El‐Gareib
- Zoology Department, Faculty of ScienceCairo UniversityEgypt
| | - R.G. Ahmed
- Zoology Department, Faculty of ScienceBeni Suef UniversityEgypt
| |
Collapse
|
49
|
Abstract
Cellular actions of thyroid hormone may be initiated within the cell nucleus, at the plasma membrane, in cytoplasm, and at the mitochondrion. Thyroid hormone nuclear receptors (TRs) mediate the biological activities of T(3) via transcriptional regulation. Two TR genes, alpha and beta, encode four T(3)-binding receptor isoforms (alpha1, beta1, beta2, and beta3). The transcriptional activity of TRs is regulated at multiple levels. Besides being regulated by T(3), transcriptional activity is regulated by the type of thyroid hormone response elements located on the promoters of T(3) target genes, by the developmental- and tissue-dependent expression of TR isoforms, and by a host of nuclear coregulatory proteins. These nuclear coregulatory proteins modulate the transcription activity of TRs in a T(3)-dependent manner. In the absence of T(3), corepressors act to repress the basal transcriptional activity, whereas in the presence of T(3), coactivators function to activate transcription. The critical role of TRs is evident in that mutations of the TRbeta gene cause resistance to thyroid hormones to exhibit an array of symptoms due to decreasing the sensitivity of target tissues to T(3). Genetically engineered knockin mouse models also reveal that mutations of the TRs could lead to other abnormalities beyond resistance to thyroid hormones, including thyroid cancer, pituitary tumors, dwarfism, and metabolic abnormalities. Thus, the deleterious effects of mutations of TRs are more severe than previously envisioned. These genetic-engineered mouse models provide valuable tools to ascertain further the molecular actions of unliganded TRs in vivo that could underlie the pathogenesis of hypothyroidism. Actions of thyroid hormone that are not initiated by liganding of the hormone to intranuclear TR are termed nongenomic. They may begin at the plasma membrane or in cytoplasm. Plasma membrane-initiated actions begin at a receptor on integrin alphavbeta3 that activates ERK1/2 and culminate in local membrane actions on ion transport systems, such as the Na(+)/H(+) exchanger, or complex cellular events such as cell proliferation. Concentration of the integrin on cells of the vasculature and on tumor cells explains recently described proangiogenic effects of iodothyronines and proliferative actions of thyroid hormone on certain cancer cells, including gliomas. Thus, hormonal events that begin nongenomically result in effects in DNA-dependent effects. l-T(4) is an agonist at the plasma membrane without conversion to T(3). Tetraiodothyroacetic acid is a T(4) analog that inhibits the actions of T(4) and T(3) at the integrin, including angiogenesis and tumor cell proliferation. T(3) can activate phosphatidylinositol 3-kinase by a mechanism that may be cytoplasmic in origin or may begin at integrin alphavbeta3. Downstream consequences of phosphatidylinositol 3-kinase activation by T(3) include specific gene transcription and insertion of Na, K-ATPase in the plasma membrane and modulation of the activity of the ATPase. Thyroid hormone, chiefly T(3) and diiodothyronine, has important effects on mitochondrial energetics and on the cytoskeleton. Modulation by the hormone of the basal proton leak in mitochondria accounts for heat production caused by iodothyronines and a substantial component of cellular oxygen consumption. Thyroid hormone also acts on the mitochondrial genome via imported isoforms of nuclear TRs to affect several mitochondrial transcription factors. Regulation of actin polymerization by T(4) and rT(3), but not T(3), is critical to cell migration. This effect has been prominently demonstrated in neurons and glial cells and is important to brain development. The actin-related effects in neurons include fostering neurite outgrowth. A truncated TRalpha1 isoform that resides in the extranuclear compartment mediates the action of thyroid hormone on the cytoskeleton.
Collapse
Affiliation(s)
- Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
50
|
Guigon CJ, Cheng SY. Novel non-genomic signaling of thyroid hormone receptors in thyroid carcinogenesis. Mol Cell Endocrinol 2009; 308:63-9. [PMID: 19549593 PMCID: PMC2744088 DOI: 10.1016/j.mce.2009.01.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 12/16/2008] [Accepted: 01/06/2009] [Indexed: 11/28/2022]
Abstract
The thyroid hormone receptors (TRs) are transcription factors that mediate the pleiotropic activities of the thyroid hormone, T3. Four T3-binding isoforms, TRalpha1, TRbeta1, TRbeta2, and TRbeta3, are encoded by two genes, THRA and THRB. Mutations and altered expression of TRs have been reported in human cancers. A targeted germ-line mutation of the Thrbeta gene in the mouse leads to spontaneous development of follicular thyroid carcinoma (TRbeta(PV/PV) mouse). The TRbetaPV mutant has lost T3-binding activity and displays potent dominant negative activity. The striking phenotype of thyroid cancer exhibited by TRbeta(PV/PV) mice has recently led to the discovery of novel non-genomic actions of TRbetaPV that contribute to thyroid carcinogenesis. These actions involve direct physical interaction of TRbetaPV with cellular proteins, namely the regulatory subunit of the phosphatidylinositol 3-kinase (p85alpha), the pituitary tumor transforming gene (PTTG) and beta-catenin, that are critically involved in cell proliferation, motility, migration, and metastasis. Thus, a TRbeta mutant (TRbetaPV), via a novel mode of non-genomic action, acts as an oncogene in thyroid carcinogenesis.
Collapse
Affiliation(s)
| | - Sheue-yann Cheng
- To whom correspondence should be addressed at: Laboratory of Molecular Biology, National Cancer Institute, 37 Convent Dr, Room 5128, Bethesda, MD 20892-4264, Tel: (301) 496-4280; Fax: (301) 402-1344; E-mail:
| |
Collapse
|