Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 610
Filter
1.
Front Endocrinol (Lausanne) ; 13: 938596, 2022.
Article in English | MEDLINE | ID: mdl-36072926

ABSTRACT

Activation and/or modulation of the membrane-associated receptors plays a critical role in brain development. Thyroid hormone (TH) acts on both nuclear receptors (thyroid hormone receptor, TR) and membrane-associated receptors, particularly integrin αvß3 in neurons and glia. Integrin αvß3-mediated signal transduction mediates various cellular events during development including morphogenesis, migration, synaptogenesis, and intracellular metabolism. However, the involvement of integrin αvß3-mediated TH action during brain development remains poorly understood. Thus, we examined the integrin αvß3-mediated effects of TH (T3, T4, and rT3) in the neurons and astrocytes using primary cerebellar culture, astrocyte-enriched culture, Neuro-2A clonal cells, and co-culture of neurons and astrocytes. We found that TH augments dendrite arborization of cerebellar Purkinje cells. This augmentation was suppressed by knockdown of integrin αvß3, as well as TRα and TRß. A selective integrin αvß3 antagonist, LM609, was also found to suppress TH-induced arborization. However, whether this effect was a direct action of TH on Purkinje cells or due to indirect actions of other cells subset such as astrocytes was not clarified. To further study neuron-specific molecular mechanisms, we used Neuro-2A clonal cells and found TH also induces neurite growth. TH-induced neurite growth was reduced by co-exposure with LM609 or knockdown of TRα, but not TRß. Moreover, co-culture of Neuro-2A and astrocytes also increased TH-induced neurite growth, indicating astrocytes may be involved in neuritogenesis. TH increased the localization of synapsin-1 and F-actin in filopodia tips. TH exposure also increased phosphorylation of FAK, Akt, and ERK1/2. Phosphorylation was suppressed by co-exposure with LM609 and TRα knockdown. These results indicate that TRs and integrin αvß3 play essential roles in TH-induced dendritogenesis and neuritogenesis. Furthermore, astrocytes-neuron communication via TR-dependent and TR-independent signaling through membrane receptors and F-actin are required for TH-induced neuritogenesis.


Subject(s)
Actins , Integrin alphaVbeta3 , Actins/metabolism , Actins/pharmacology , Integrin alphaVbeta3/metabolism , Receptors, Thyroid Hormone/physiology , Signal Transduction/physiology , Thyroid Hormone Receptors beta , Thyroid Hormones/pharmacology , Thyroid Hormones/physiology
2.
Inflamm Res ; 71(2): 243-253, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35059772

ABSTRACT

OBJECTIVE: Inflammation and proliferation of vascular smooth muscle cells (VSMCs), induced by angiotensin II (AngII) and other growth factors, play important roles in the pathogenesis of hypertension, restenosis, and atherosclerosis. Dihydroartemisinin (DHA) exhibits broad protective effects. However, the effects of DHA on AngII-induced inflammation and proliferation of VSMCs remain unknown. MATERIALS AND METHODS: AngII was used to construct VSMCs and vascular inflammation model in vitro and in vivo. The protective roles of DHA in inflammatory response and proliferation were evaluated through CCK-8, BrdU assay and immunofluorescence staining. The level of mRNA N6-methyladenosine was measured by m6A-RNA immunoprecipitation (MeRIP) assay. Western blot and quantitative real-time PCR were used to investigate the relationship between FTO and its potential downstream signaling molecules. RESULTS: In the present study, we found that DHA significantly suppressed AngII-induced proliferation of VSMCs and the expression of IL-6 and Ccl2 in a dose-dependent manner. Additionally, we confirmed that fat mass and obesity-associated (FTO) plays a critical role in AngII-induced VSMC proliferation and inflammation. FTO knockdown increased the methylation level of NR4A3 mRNA, whereas FTO, but not mutated FTO overexpression, reduced the methylation level of NR4A3 mRNA. These results suggest that DHA plays a protective role in AngII-induced VSMC proliferation and the associated inflammation by inhibiting the FTO/NR4A3 axis. CONCLUSION: Our findings provide new insight into the mechanisms of DHA and its critical role in the pathogenesis of hypertension-related vascular complications.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/antagonists & inhibitors , Angiotensin II/pharmacology , Artemisinins/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Inflammation/prevention & control , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Receptors, Steroid/antagonists & inhibitors , Receptors, Thyroid Hormone/antagonists & inhibitors , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/physiology , Animals , Cell Proliferation/drug effects , Cells, Cultured , DNA-Binding Proteins/physiology , Mice , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Nerve Tissue Proteins/physiology , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/physiology , Signal Transduction/drug effects
3.
Life Sci ; 295: 120086, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-34710445

ABSTRACT

AIMS: The objective of the present study was to investigate the effect of melatonin and L-thyroxine (T4) on the expression of various receptors, and some metabolic, reproductive, and gonadotropic hormones in letrozole-induced polycystic ovary syndrome (PCOS) in rats. MATERIAL AND METHODS: Assessment of gravimetric, hormonal profile and thyroid histology and relative expression of melatonin receptors (MT1, MT2) and estrogen receptor α (Erα) in thyroid and ovary, and type II iodothyronine deiodinase (Dio2) and thyroid hormone receptor α (TRα) in the ovary were performed using standard protocols. KEY FINDINGS: A significant increase in thyroid follicles numbers was noted in the hyperthyroid rat. T4 treatment to PCOS showed the expected increment in the circulating level of triiodothyronine (T3) and T4. Melatonin and T4 treatment of PCOS rats resulted in a significant decrease in the circulating level of T3 and T4. Hyperthyroid rats showed a decrement in plasma melatonin levels. However, T4 treatment to PCOS rats showed increased circulating melatonin levels, and a decrease in the circulating level of gonadotropins (LH and FSH), and testosterone. Melatonin treatment to PCOS-hyperthyroid rats resulted in the normal expression of ovarian and thyroid MT1 and ERα, receptors, which had been altered in PCOS and hyperthyroid rats, without any significant change in the MT2 receptor. SIGNIFICANCE: The present findings suggest a fine interplay and cross-talk via melatonin and its two receptors with ERα, TRα, and Dio2in thyroid and ovarian tissue during PCOS and hyperthyroidism pathogenicity.


Subject(s)
Estrogen Receptor alpha/metabolism , Polycystic Ovary Syndrome/pathology , Receptors, Thyroid Hormone/metabolism , Animals , Disease Models, Animal , Estrogen Receptor alpha/physiology , Female , Gene Expression/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gonadotropins/metabolism , Hyperthyroidism/metabolism , Letrozole/pharmacology , Melatonin/metabolism , Melatonin/pharmacology , Ovary/metabolism , Ovary/physiology , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Rats , Rats, Wistar , Receptors, Thyroid Hormone/physiology , Testosterone/metabolism , Thyroid Gland/drug effects , Thyroid Hormones/metabolism , Thyroxine/metabolism
4.
Int J Mol Sci ; 22(21)2021 Oct 21.
Article in English | MEDLINE | ID: mdl-34768801

ABSTRACT

The mechanisms committed in the activation and response of vascular and inflammatory immune cells play a major role in tissue remodeling in cardiovascular diseases (CVDs) such as atherosclerosis, pulmonary arterial hypertension, and abdominal aortic aneurysm. Cardiovascular remodeling entails interrelated cellular processes (proliferation, survival/apoptosis, inflammation, extracellular matrix (ECM) synthesis/degradation, redox homeostasis, etc.) coordinately regulated by a reduced number of transcription factors. Nuclear receptors of the subfamily 4 group A (NR4A) have recently emerged as key master genes in multiple cellular processes and vital functions of different organs, and have been involved in a variety of high-incidence human pathologies including atherosclerosis and other CVDs. This paper reviews the major findings involving NR4A3 (Neuron-derived Orphan Receptor 1, NOR-1) in the cardiovascular remodeling operating in these diseases.


Subject(s)
Cardiovascular Diseases/pathology , Cardiovascular System/pathology , DNA-Binding Proteins/metabolism , Inflammation , Nerve Tissue Proteins/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Atherosclerosis , Atrial Remodeling , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , DNA-Binding Proteins/physiology , Humans , Nerve Tissue Proteins/physiology , Pulmonary Arterial Hypertension , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/physiology
5.
Exp Hematol ; 95: 13-22, 2021 03.
Article in English | MEDLINE | ID: mdl-33440185

ABSTRACT

Erythropoiesis is an intricate process starting in hematopoietic stem cells and leading to the daily production of 200 billion red blood cells (RBCs). Enucleation is a greatly complex and rate-limiting step during terminal maturation of mammalian RBC production involving expulsion of the nucleus from the orthochromatic erythroblasts, resulting in the formation of reticulocytes. The dynamic enucleation process involves many factors ranging from cytoskeletal proteins to transcription factors to microRNAs. Lack of optimum terminal erythroid maturation and enucleation has been an impediment to optimum RBC production ex vivo. Major efforts in the past two decades have exposed some of the mechanisms that govern the enucleation process. This review focuses in detail on mechanisms implicated in enucleation and discusses the future perspectives of this fascinating process.


Subject(s)
Cell Nucleus , Erythroblasts/ultrastructure , Erythrocytes/ultrastructure , Erythropoiesis , Reticulocytes/ultrastructure , Animals , Birds/blood , Calcium/physiology , Chromatin/ultrastructure , Colony-Forming Units Assay , Computational Biology , Cytokines/physiology , Cytoskeletal Proteins/physiology , DNA-Binding Proteins/physiology , Erythroblasts/cytology , Erythrocytes/cytology , Intercellular Signaling Peptides and Proteins/physiology , Mammals/blood , Mice , MicroRNAs/physiology , Proto-Oncogene Proteins/physiology , Receptors, Thyroid Hormone/physiology , Repressor Proteins/physiology , Reticulocytes/cytology , Transcription Factors/physiology , Transport Vesicles/physiology , Yolk Sac/cytology , rho GTP-Binding Proteins/physiology
6.
Physiol Rev ; 101(1): 319-352, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32584192

ABSTRACT

The extracellular domain of plasma membrane integrin αvß3 contains a cell surface receptor for thyroid hormone analogues. The receptor is largely expressed and activated in tumor cells and rapidly dividing endothelial cells. The principal ligand for this receptor is l-thyroxine (T4), usually regarded only as a prohormone for 3,5,3'-triiodo-l-thyronine (T3), the hormone analogue that expresses thyroid hormone in the cell nucleus via nuclear receptors that are unrelated structurally to integrin αvß3. At the integrin receptor for thyroid hormone, T4 regulates cancer and endothelial cell division, tumor cell defense pathways (such as anti-apoptosis), and angiogenesis and supports metastasis, radioresistance, and chemoresistance. The molecular mechanisms involve signal transduction via mitogen-activated protein kinase and phosphatidylinositol 3-kinase, differential expression of multiple genes related to the listed cell processes, and regulation of activities of other cell surface proteins, such as vascular growth factor receptors. Tetraiodothyroacetic acid (tetrac) is derived from T4 and competes with binding of T4 to the integrin. In the absence of T4, tetrac and chemically modified tetrac also have anticancer effects that culminate in altered gene transcription. Tumor xenografts are arrested by unmodified and chemically modified tetrac. The receptor requires further characterization in terms of contributions to nonmalignant cells, such as platelets and phagocytes. The integrin αvß3 receptor for thyroid hormone offers a large panel of cellular actions that are relevant to cancer biology and that may be regulated by tetrac derivatives.


Subject(s)
Integrins/physiology , Thyroid Hormones/physiology , Animals , Humans , Mitogen-Activated Protein Kinases/physiology , Receptors, Thyroid Hormone/physiology , Signal Transduction , Thyroxine/physiology , Triiodothyronine
7.
Proc Natl Acad Sci U S A ; 117(26): 15262-15269, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32541022

ABSTRACT

Thyroid hormone (TH) signaling plays an important role in the regulation of long-wavelength vision in vertebrates. In the retina, thyroid hormone receptor ß (thrb) is required for expression of long-wavelength-sensitive opsin (lws) in red cone photoreceptors, while in retinal pigment epithelium (RPE), TH regulates expression of a cytochrome P450 enzyme, cyp27c1, that converts vitamin A1 into vitamin A2 to produce a red-shifted chromophore. To better understand how TH controls these processes, we analyzed the phenotype of zebrafish with mutations in the three known TH nuclear receptor transcription factors (thraa, thrab, and thrb). We found that no single TH nuclear receptor is required for TH-mediated induction of cyp27c1 but that deletion of all three (thraa-/-;thrab-/-;thrb-/- ) completely abrogates its induction and the resulting conversion of A1- to A2-based retinoids. In the retina, loss of thrb resulted in an absence of red cones at both larval and adult stages without disruption of the underlying cone mosaic. RNA-sequencing analysis revealed significant down-regulation of only five genes in adult thrb-/- retina, of which three (lws1, lws2, and miR-726) occur in a single syntenic cluster. In the thrb-/- retina, retinal progenitors destined to become red cones were transfated into ultraviolet (UV) cones and horizontal cells. Taken together, our findings demonstrate cooperative regulation of cyp27c1 by TH receptors and a requirement for thrb in red cone fate determination. Thus, TH signaling coordinately regulates both spectral sensitivity and sensory plasticity.


Subject(s)
Color Vision/physiology , Cytochrome P-450 Enzyme System/metabolism , Opsins/metabolism , Receptors, Thyroid Hormone/physiology , Visual Perception/physiology , Zebrafish Proteins/metabolism , Animals , Color Vision/genetics , Cytochrome P-450 Enzyme System/genetics , Gene Deletion , Gene Expression Regulation , Opsins/genetics , Retinal Cone Photoreceptor Cells , Ultraviolet Rays , Zebrafish , Zebrafish Proteins/genetics
8.
Front Endocrinol (Lausanne) ; 11: 624122, 2020.
Article in English | MEDLINE | ID: mdl-33597928

ABSTRACT

The nuclear orphan receptors NR4A1, NR4A2, and NR4A3 are immediate early genes that are induced by various signals. They act as transcription factors and their activity is not regulated by ligand binding and are thus regulated via their expression levels. Their expression is transiently induced in T cells by triggering of the T cell receptor following antigen recognition during both thymic differentiation and peripheral T cell responses. In this review, we will discuss how NR4A family members impact different aspects of the life of a T cell from thymic differentiation to peripheral response against infections and cancer.


Subject(s)
DNA-Binding Proteins/physiology , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/physiology , T-Lymphocytes/physiology , Thymus Gland/physiology , Animals , Humans , Receptors, Antigen, T-Cell/physiology , Signal Transduction/physiology , Thymus Gland/cytology
9.
Thyroid ; 30(1): 8-12, 2020 01.
Article in English | MEDLINE | ID: mdl-31822204

ABSTRACT

Basic research in 2019 yielded exciting discoveries and advancements in thyroidology. Specifically, there have been breakthroughs in our understanding of the molecular actions of thyroid hormone and thyroid hormone receptors, thyroid hormone metabolism and transport, autoimmunity, and thyroid cancer. Next, I summarize important studies published over the past year and whose major data I presented during the 89th American Thyroid Association annual meeting at the opening plenary session The Year in Thyroidology.


Subject(s)
Endocrinology/methods , Endocrinology/trends , Thyroid Gland/physiology , Thyroid Hormones/metabolism , Thyroid Neoplasms/metabolism , Autoimmunity , Gene Expression Regulation , Heart/physiology , Humans , Myelin Sheath/physiology , Receptors, Thyroid Hormone/physiology , Regeneration , Signal Transduction , Societies, Medical , Symporters/physiology , Thyroid Epithelial Cells/cytology
10.
Life Sci ; 239: 116975, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31654748

ABSTRACT

AIMS: Previous study indicated that the increase of local bio-availability of 3'3'5-triiodothyronine (T3) influenced osteoarthritis (OA) initiation. We aimed to investigate the role of thyroid hormone receptors (THRs) signaling in OA osteoblasts. MATERIALS AND METHODS: THRs expression in OA was detected by immunohistochemistry, immunofluorescence, RT-qPCR and western blotting. These effects on the expression of angiogenesis-related factors were examined after THRα or THRß knockdown in OA osteoblasts. Fluorescence in situ hybridization was used to confirm the leading receptor for regulating angiogenesis-related factors. Co-culture model was utilized to observe the MMPs expression in chondrocytes after THRα knockdown in osteoblasts. The in vivo effects were also studied after intra-articular injection with THRα siRNA in OA model mice. Micro-CT and immunohistochemistry were employed to evaluate the changes of subchondral bone. KEY FINDINGS: THRs expression and nuclear translocation were upregulated in human OA osteoblasts. Immunohistochemistry showed that angiogenic activities were increased in OA subchondral bone of human and mice. VEGF, HIF-1α and IGF-1, these THR downstream genes were downregulated after THRα knockdown in OA osteoblasts. Fluorescence in situ hybridization further indicated that THRα signaling mainly regulated VEGF expression. Intra-articular injection with THRα siRNA reduced angiogenic activities in OA model mice subchondral bone and ameliorated cartilage degradation. Micro-CT analysis displayed that the aberrant subchondral bone formation in OA was promoted. SIGNIFICANCE: The microangiogenesis in subchondral bone may be partly attributed to abnormal THRα signaling in osteoblasts, and local inhibition of the THRα could be a potential target to treat OA.


Subject(s)
Neovascularization, Physiologic/physiology , Osteoblasts/metabolism , Receptors, Thyroid Hormone/metabolism , Aged , Aged, 80 and over , Animals , Bone and Bones/metabolism , China , Chondrocytes/metabolism , Female , Humans , Injections, Intra-Articular , Male , Mice , Mice, Inbred C57BL , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis, Knee/drug therapy , Osteoblasts/drug effects , Receptors, Thyroid Hormone/physiology , Signal Transduction/physiology , Thyroxine/analysis , Thyroxine/blood , Triiodothyronine/analysis , Triiodothyronine/blood
11.
Thyroid ; 29(9): 1173-1191, 2019 09.
Article in English | MEDLINE | ID: mdl-31389309

ABSTRACT

Background: Thyroid hormones (THs) exert a strong influence on mammalian lipid metabolism at the systemic and hepatic levels by virtue of their roles in regulating circulating lipoprotein, triglyceride (TAG), and cholesterol levels, as well as hepatic TAG storage and metabolism. These effects are mediated by intricate sensing and feedback systems that function at the physiological, metabolic, molecular, and transcriptional levels in the liver. Dysfunction in the pathways involved in lipid metabolism disrupts hepatic lipid homeostasis and contributes to the pathogenesis of metabolic diseases, such as nonalcoholic fatty liver disease (NAFLD) and hypercholesterolemia. There has been strong interest in understanding and employing THs, TH metabolites, and TH mimetics as lipid-modifying drugs. Summary: THs regulate many processes involved in hepatic TAG and cholesterol metabolism to decrease serum cholesterol and intrahepatic lipid content. TH receptor ß analogs designed to have less side effects than the natural hormone are currently being tested in phase II clinical studies for NAFLD and hypercholesterolemia. The TH metabolites, 3,5-diiodo-l-thyronine (T2) and T1AM (3-iodothyronamine), have different beneficial effects on lipid metabolism compared with triiodothyronine (T3), although their clinical application is still under investigation. Also, prodrugs and glucagon/T3 conjugates have been developed that direct TH to the liver. Conclusions: TH-based therapies show clinical promise for the treatment of NAFLD and hypercholesterolemia. Strategies for limiting side effects of TH are being developed and may enable TH metabolites and analogs to have specific effects in the liver for treatments of these conditions. These liver-specific effects and potential suppression of the hypothalamic/pituitary/thyroid axis raise the issue of monitoring liver-specific markers of TH action to assess clinical efficacy and dosing of these compounds.


Subject(s)
Hypercholesterolemia/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Thyroid Hormones/physiology , Animals , Humans , Hypercholesterolemia/metabolism , Lipid Metabolism , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Thyroid Hormone/physiology , Thyroid Hormones/agonists , Thyroid Hormones/therapeutic use
12.
Science ; 364(6436): 184-188, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30846611

ABSTRACT

Tissue regenerative potential displays striking divergence across phylogeny and ontogeny, but the underlying mechanisms remain enigmatic. Loss of mammalian cardiac regenerative potential correlates with cardiomyocyte cell-cycle arrest and polyploidization as well as the development of postnatal endothermy. We reveal that diploid cardiomyocyte abundance across 41 species conforms to Kleiber's law-the ¾-power law scaling of metabolism with bodyweight-and inversely correlates with standard metabolic rate, body temperature, and serum thyroxine level. Inactivation of thyroid hormone signaling reduces mouse cardiomyocyte polyploidization, delays cell-cycle exit, and retains cardiac regenerative potential in adults. Conversely, exogenous thyroid hormones inhibit zebrafish heart regeneration. Thus, our findings suggest that loss of heart regenerative capacity in adult mammals is triggered by increasing thyroid hormones and may be a trade-off for the acquisition of endothermy.


Subject(s)
Heart/physiology , Myocytes, Cardiac/physiology , Polyploidy , Regeneration/physiology , Thyroid Hormones/physiology , Animals , Body Temperature Regulation , Cell Cycle Checkpoints , Cell Proliferation , Diploidy , Mice , Myocytes, Cardiac/classification , Phylogeny , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/physiology , Regeneration/drug effects , Regeneration/genetics , Signal Transduction , Thyroid Hormones/pharmacology , Zebrafish
13.
Vitam Horm ; 106: 67-96, 2018.
Article in English | MEDLINE | ID: mdl-29407448

ABSTRACT

Nongenomic actions of thyroid hormone are initiated by the hormone at receptors in the plasma membrane, in cytoplasm, or in mitochondria and do not require the interaction of nuclear thyroid hormone receptors (TRs) with their primary ligand, 3,5,3'-triiodo-l-thyronine (T3). Receptors involved in nongenomic actions may or may not have structural homologies with TRs. Certain nongenomic actions that originate at the plasma membrane may modify the state and function of intranuclear TRs. Reviewed here are nongenomic effects of the hormone-T3 or, in some cases, l-thyroxine (T4)-that are initiated at (a) truncated TRα isoforms, e.g., p30 TRα1, (b) cytoplasmic proteins, or (c) plasma membrane integrin αvß3. p30 TRα1 is not transcriptionally competent, binds T3 at the cell surface, and consequently expresses a number of important functions in bone cells. Nongenomic hormonal control of mitochondrial respiration involves a TRα isoform, and another truncated TRα isoform nongenomically regulates the state of cellular actin. Cytoplasmic hormone-binding proteins involved in nongenomic actions of thyroid hormone include ketimine reductase, pyruvate kinase, and TRß that shuttle among intracellular compartments. Functions of the receptor for T4 on integrin αvß3 include stimulation of proliferation of cancer and endothelial cells (angiogenesis) and regulation of transcription of cancer cell survival pathway genes. T4 serves as a prohormone for T3 in genomic actions of thyroid hormone, but T4 is a hormone at αvß3 and more important to cancer cell function than is T3. Thus, characterization of nongenomic actions of the hormone has served to broaden our understanding of the cellular roles of T3 and T4.


Subject(s)
Receptors, Thyroid Hormone/physiology , Thyroid Hormones/physiology , Actin Cytoskeleton , Animals , Cytoplasm , Humans , Molecular Structure , Thyroid Hormones/chemistry
14.
Sci Rep ; 6: 38756, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27934948

ABSTRACT

Stathmin (STMN1), a recognized oncoprotein upregulated in various solid tumors, promotes microtubule disassembly and modulates tumor growth and migration activity. However, the mechanisms underlying the genetic regulation of STMN1 have yet to be elucidated. In the current study, we report that thyroid hormone receptor (THR) expression is negatively correlated with STMN1 expression in a subset of clinical hepatocellular carcinoma (HCC) specimens. We further identified the STMN1 gene as a target of thyroid hormone (T3) in the HepG2 hepatoma cell line. An analysis of STMN1 expression profile and mechanism of transcriptional regulation revealed that T3 significantly suppressed STMN1 mRNA and protein expression, and further showed that THR directly targeted the STMN1 upstream element to regulate STMN1 transcriptional activity. Specific knockdown of STMN1 suppressed cell proliferation and xenograft tumor growth in mice. In addition, T3 regulation of cell growth arrest and cell cycle distribution were attenuated by overexpression of STMN1. Our results suggest that the oncogene STMN1 is transcriptionally downregulated by T3 in the liver. This T3-mediated suppression of STMN1 supports the theory that T3 plays an inhibitory role in HCC tumor growth, and suggests that the lack of normal THR function leads to elevated STMN1 expression and malignant growth.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Division/physiology , Liver Neoplasms/genetics , Stathmin/genetics , Triiodothyronine/physiology , Carcinoma, Hepatocellular/pathology , Gene Knockdown Techniques , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Receptors, Thyroid Hormone/physiology
15.
Sci Rep ; 6: 30990, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27484112

ABSTRACT

Decreased thyroidal hormone production is found during lipopolysaccharide (LPS)-induced endotoxic shock in animals as well as in critically ill patients. Here we studied the role of the thyroid hormone receptors (TRs) in activation of STAT3, NF-κB and ERK, which play a key role in the response to inflammatory cytokines during sepsis. TR knockout mice showed down-regulation of hepatic inflammatory mediators, including interleukin 6 (IL-6) in response to LPS. Paradoxically, STAT3 and ERK activity were higher, suggesting that TRs could act as endogenous repressors of these pathways. Furthermore, hyperthyroidism increased cytokine production and mortality in response to LPS, despite decreasing hepatic STAT3 and ERK activity. This suggested that TRs could directly repress the response of the cells to inflammatory mediators. Indeed, we found that the thyroid hormone T3 suppresses IL-6 signalling in macrophages and hepatocarcinoma cells, inhibiting STAT3 activation. Consequently, the hormone strongly antagonizes IL-6-stimulated gene transcription, reducing STAT3 recruitment and histone acetylation at IL-6 target promoters. In conclusion, TRs are potent regulators of inflammatory responses and immune homeostasis during sepsis. Reduced responses to IL-6 should serve as a negative feedback mechanism for preventing deleterious effects of excessive hormone signaling during infections.


Subject(s)
Endotoxemia/etiology , Interleukin-6/antagonists & inhibitors , Lipopolysaccharides/toxicity , Liver/immunology , Receptors, Thyroid Hormone/physiology , Thyroid Hormones/administration & dosage , Animals , Endotoxemia/drug therapy , Endotoxemia/metabolism , Endotoxemia/pathology , Interleukin-6/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , NF-kappa B p50 Subunit/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
16.
Compr Physiol ; 6(3): 1221-37, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27347891

ABSTRACT

Thyroid hormones play important roles in regulating normal metabolism, development, and growth. They also stimulate cancer cell proliferation. Their metabolic and developmental effects and growth effects in normal tissues are mediated primarily by nuclear hormone receptors. A cell surface receptor for the hormone on integrin [alpha]vß3 is the initiation site for effects on tumor cells. Clinical hypothyroidism may retard cancer growth, and hyperthyroidism was recently linked to the prevalence of certain cancers. Local levels of thyroid hormones are controlled through activation and deactivation of iodothyronine deiodinases in different organs. The relative activities of different deiodinases that exist in tissues or organs also affect the progression and development of specific types of cancers. In this review, the effects of thyroid hormone on signaling pathways in breast, brain, liver, thyroid, and colon cancers are discussed. The importance of nuclear thyroid hormone receptor isoforms and of the hormone receptor on the extracellular domain of integrin [alpha]vß3 as potential cancer risk factors and therapeutic targets are addressed. We analyze the intracellular signaling pathways activated by thyroid hormones in cancer progression in hyperthyroidism or at physiological concentrations in the euthyroid state. Determining how to utilize the deaminated thyroid hormone analog (tetrac), and its nanoparticulate derivative to reduce risks of cancer progression, enhance therapeutic outcomes, and prevent cancer recurrence is also deliberated. © 2016 American Physiological Society. Compr Physiol 6:1221-1237, 2016.


Subject(s)
Apoptosis/physiology , Neoplasms/metabolism , Thyroid Hormones/physiology , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Digestive System Neoplasms/metabolism , Digestive System Neoplasms/pathology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasms/pathology , Receptors, Thyroid Hormone/physiology , Signal Transduction/physiology
17.
Clin Sci (Lond) ; 129(7): 589-99, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26186742

ABSTRACT

Skeletal muscle metabolism is highly dependent on mitochondrial function, with impaired mitochondrial biogenesis associated with the development of metabolic diseases such as insulin resistance and type 2 diabetes. Mitochondria display substantial plasticity in skeletal muscle, and are highly sensitive to levels of physical activity. It is thought that physical activity promotes mitochondrial biogenesis in skeletal muscle through increased expression of genes encoded in both the nuclear and the mitochondrial genome; however, how this process is co-ordinated at the cellular level is poorly understood. Nuclear receptors (NRs) are key signalling proteins capable of integrating environmental factors and mitochondrial function, thereby providing a potential link between exercise and mitochondrial biogenesis. The aim of this review is to highlight the function of NRs in skeletal muscle mitochondrial biogenesis and discuss the therapeutic potential of NRs for the management and treatment of chronic metabolic disease.


Subject(s)
Gene Expression Regulation , Mitochondria/physiology , Muscle, Skeletal/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Chickens , Diabetes Mellitus, Type 2/metabolism , Exercise , Genome, Mitochondrial , Humans , Mice , Mitochondria, Muscle/physiology , Muscular Diseases/metabolism , Ovalbumin/metabolism , Peroxisome Proliferator-Activated Receptors/physiology , Physical Conditioning, Animal , Rats , Receptors, Thyroid Hormone/physiology , Transcription Factors/physiology
18.
Blood ; 125(9): 1477-87, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25561507

ABSTRACT

The orphan nuclear receptors TR2 and TR4 have been shown to play key roles in repressing the embryonic and fetal globin genes in erythroid cells. However, combined germline inactivation of Tr2 and Tr4 leads to periimplantation lethal demise in inbred mice. Hence, we have previously been unable to examine the consequences of their dual loss of function in adult definitive erythroid cells. To circumvent this issue, we generated conditional null mutants in both genes and performed gene inactivation in vitro in adult bone marrow cells. Compound Tr2/Tr4 loss of function led to induced expression of the embryonic εy and ßh1 globins (murine counterparts of the human ε- and γ-globin genes). Additionally, TR2/TR4 function is required for terminal erythroid cell maturation. Loss of TR2/TR4 abolished their occupancy on the εy and ßh1 gene promoters, and concurrently impaired co-occupancy by interacting corepressors. These data strongly support the hypothesis that the TR2/TR4 core complex is an adult stage-specific, gene-selective repressor of the embryonic globin genes. Detailed mechanistic understanding of the roles of TR2/TR4 and their cofactors in embryonic and fetal globin gene repression may ultimately enhance the discovery of novel therapeutic agents that can effectively inhibit their transcriptional activity and be safely applied to the treatment of ß-globinopathies.


Subject(s)
Embryo, Mammalian/metabolism , Erythroid Cells/cytology , Fetus/metabolism , Gene Expression Regulation, Developmental , Nuclear Receptor Subfamily 2, Group C, Member 1/physiology , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/physiology , beta-Globins/metabolism , Animals , Blotting, Western , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Erythroid Cells/metabolism , Flow Cytometry , Gene Silencing , Humans , Integrases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , beta-Globins/genetics
19.
Biochim Biophys Acta ; 1849(2): 130-41, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24844179

ABSTRACT

BACKGROUND: Thyroid hormones (THs) play an essential role in vertebrate development, acting predominantly via nuclear TH receptors (TRs) which are ligand-dependent transcription factors. Binding of the ligand (predominantly T3) induces a switch from gene activation to gene repression or vice versa. Iodothyronine deiodinases (Ds) and TH transporters are important regulators of intracellular T3 availability and therefore contribute to the control of TR-dependent development. FOCUS: The present review discusses the possible roles of Ds and TH transporters in regulating embryonic and larval (pre-juvenile) TR-dependent development in vertebrates. It focuses mainly on well-known model species for direct and indirect vertebrate development, including zebrafish, Xenopus, chicken and mouse. Data are provided on stage- and tissue/cell-specific changes in expression of Ds and TH transporters. This information is combined with functional data obtained from gain-and-loss of function studies. CONCLUSION: Knockout/knockdown of each type of D has provided strong evidence for their implication in the control of important developmental processes and several D expression patterns and functions have been conserved throughout vertebrate evolution. Knockout/knockdown of the inactivating D3 enzyme indicates that a premature switch from unliganded to liganded TR action is often more detrimental than a delayed one. The majority of ontogenetic studies on TH transporter distribution and function have focused on brain development, showing variable impact of knockout/knockdown depending on the species. Future research in different models using conditional silencing will hopefully further improve our understanding on how TH transporters, Ds and TRs cooperate to regulate TR-mediated impact on vertebrate development. This article is part of a Special Issue entitled: Nuclear receptors in animal development.


Subject(s)
Receptors, Thyroid Hormone/physiology , Thyroid Hormones/metabolism , Amphibians/embryology , Amphibians/genetics , Animals , Chick Embryo , Gene Expression Regulation, Developmental , Humans , Iodide Peroxidase/physiology , Mice , Mice, Knockout , Receptors, Thyroid Hormone/metabolism , Zebrafish/embryology , Zebrafish/genetics
20.
Int J Cancer ; 136(4): 955-64, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-24975468

ABSTRACT

Testicular nuclear receptor 4 (TR4) plays protective roles against oxidative stress and DNA damage and might contribute to aging. Our recent clinical tumor tissue staining results showed higher expression of TR4 in prostate cancer (PCa) patients with high Gleason scores compared to the tissues with the low Gleason scores. In vitro migration/invasion assays after manipulation of the TR4 expression in PCa cells showed that TR4 promoted PCa cells migration/invasion. Mechanism dissection found that the CCL2/CCR2 signal plays the key role in the mediation of TR4-promoted PCa cells migration/invasion. Chromatin immunoprecipitation and Luciferase assays further confirmed TR4 modulation of CCL2 at the transcriptional level and addition of the CCR2 antagonist led to interruption of the TR4-enhanced PCa cells migration/invasion. Finally, the orthotopic xenografted mice studies using the luciferase expressing CWR22Rv1 cells found that TR4 enhanced PCa metastasis and this increased metastasis was reversed when the CCR2 antagonist was injected into the mice. Together, these in vitro and in vivo results revealed a positive role of TR4 in PCa metastasis and demonstrated CCL2/CCR2 signaling as an important mediator in exerting TR4 action. This finding suggests that TR4 may represent a biomarker related to PCa metastasis and targeting the TR4-CCL2/CCR2 axis may become a new therapeutic approach to battle PCa metastasis.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/physiology , Animals , Cell Line, Tumor , Cell Movement , Chemokine CCL2 , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Lymphatic Metastasis , Male , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Prostatic Neoplasms/pathology , Receptors, CCR2 , Signal Transduction , Transcription, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL