Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell Death Differ ; 22(6): 959-73, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25361083

ABSTRACT

Mitochondrial dysfunction, often characterized by massive fission and other morphological abnormalities, is a well-known risk factor for Alzheimer's disease (AD). One causative mechanism underlying AD-associated mitochondrial dysfunction is thought to be amyloid-ß (Aß), yet the pathways between Aß and mitochondrial dysfunction remain elusive. In this study, we report that CR6-interacting factor 1 (Crif1), a mitochondrial inner membrane protein, is a key player in Aß-induced mitochondrial dysfunction. Specifically, we found that Crif1 levels were downregulated in the pathological regions of Tg6799 mice brains, wherein overexpressed Aß undergoes self-aggregation. Downregulation of Crif1 was similarly observed in human AD brains as well as in SH-SY5Y cells treated with Aß. In addition, knockdown of Crif1, using RNA interference, induced mitochondrial dysfunction with phenotypes similar to those observed in Aß-treated cells. Conversely, Crif1 overexpression prevented Aß-induced mitochondrial dysfunction and cell death. Finally, we show that Aß-induced downregulation of Crif1 is mediated by enhanced reactive oxygen species (ROS) and ROS-dependent sumoylation of the transcription factor specificity protein 1 (Sp1). These results identify the ROS-Sp1-Crif1 pathway to be a new mechanism underlying Aß-induced mitochondrial dysfunction and suggest that ROS-mediated downregulation of Crif1 is a crucial event in AD pathology. We propose that Crif1 may serve as a novel therapeutic target in the treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cell Cycle Proteins/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Nuclear Proteins/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival , Humans , Mice , Mitochondria/genetics , Nuclear Proteins/genetics , Reactive Oxygen Species/metabolism , Sp1 Transcription Factor/metabolism
2.
Oncogene ; 34(11): 1354-62, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-24681946

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that has a central role in the regulation of tumour metabolism under hypoxic conditions. HIF-1α stimulates glycolytic energy production and promotes tumour growth. Sirtuins are NAD(+)-dependent protein deacetylases that regulate cellular metabolism in response to stress; however, their involvement in the hypoxic response remains unclear. In this study, it is shown that SIRT2-mediated deacetylation of HIF-1α regulates its stability in tumour cells. SIRT2 overexpression destabilized HIF-1α under hypoxic conditions, whereas HIF-1α protein levels were high in SIRT2-deficient cells. SIRT2 directly interacted with HIF-1α and deacetylated Lys709 of HIF-1α. Deacetylation of HIF-1α by SIRT2 resulted in increased binding affinity for prolyl hydroxylase 2, a key regulator of HIF-1α stability, and increased HIF-1α hydroxylation and ubiquitination. Moreover, a pharmacological agent that increased the intracellular NAD(+)/NADH ratio led to the degradation of HIF-1α by increasing SIRT2-mediated deacetylation and subsequent hydroxylation. These findings suggest that SIRT2-mediated HIF-1α deacetylation is critical for the destablization of HIF-1α and the hypoxic response of tumour cells.


Subject(s)
Cell Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Sirtuin 2/metabolism , Animals , Cell Line, Tumor , Energy Metabolism/genetics , Female , HeLa Cells , Humans , Hydroxylation , Mice , Mice, Inbred BALB C , Mice, Nude , NAD/metabolism , Prolyl Hydroxylases/metabolism , Protein Binding , Protein Stability , RNA Interference , RNA, Small Interfering , Sirtuin 2/genetics , Ubiquitination
3.
Oncogenesis ; 2: e55, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23857250

ABSTRACT

The transcription coactivator Yes-associated protein 1 (YAP1) is regulated by the Hippo tumor suppressor pathway. However, the role of YAP1 in thyroid cancer, which is frequently associated with the BRAF(V600E) mutation, remains unknown. This study aimed to investigate the role of YAP1 in thyroid cancer. YAP1 was overexpressed in papillary (PTC) and anaplastic thyroid cancer, and nuclear YAP1 was more frequently detected in BRAF(V600E) (+) PTC. In the thyroid cancer cell lines TPC-1 and HTH7, which do not have the BRAF(V600E) mutation, YAP1 was cytosolic and inactive at high cell densities. In contrast, YAP1 was retained in the nucleus and its target genes were expressed in the thyroid cancer cells 8505C and K1, which harbor the BRAF(V600E) mutation, regardless of cell density. Furthermore, the nuclear activation of YAP1 in 8505C was not inhibited by RAF or MEK inhibitor. In vitro experiments, YAP1 silencing or overexpression affected migratory capacities of 8505C and TPC-1 cells. YAP1 knockdown resulted in marked decrease of tumor volume, invasion and distant metastasis in orthotopic tumor xenograft mouse models using the 8505C thyroid cancer cell line. Taken together, YAP1 is involved in the tumor progression of thyroid cancer and YAP1-mediated effects might not be affected by the currently used RAF kinase inhibitors.

4.
Diabetologia ; 55(5): 1482-94, 2012 May.
Article in English | MEDLINE | ID: mdl-22349108

ABSTRACT

AIMS/HYPOTHESIS: IL-6 is a proinflammatory cytokine associated with the pathogenesis of hepatic diseases. Metformin is an anti-diabetic drug used for the treatment of type 2 diabetes, and orphan nuclear receptor small heterodimer partner (SHP, also known as NR0B2), a transcriptional co-repressor, plays an important role in maintaining metabolic homeostasis. Here, we demonstrate that metformin-mediated activation of AMP-activated protein kinase (AMPK) increases SHP protein production and regulates IL-6-induced hepatic insulin resistance. METHODS: We investigated metformin-mediated SHP production improved insulin resistance through the regulation of an IL-6-dependent pathway (involving signal transducer and activator of transcription 3 [STAT3] and suppressor of cytokine signalling 3 [SOCS3]) in both Shp knockdown and Shp null mice. RESULTS: IL-6-induced STAT3 transactivation and SOCS3 production were significantly repressed by metformin, adenoviral constitutively active AMPK (Ad-CA-AMPK), and adenoviral SHP (Ad-SHP), but not in Shp knockdown, or with the adenoviral dominant negative form of AMPK (Ad-DN-AMPK). Chromatin immunoprecipitation (ChIP), co-immunoprecipitation (Co-IP) and protein localisation studies showed that SHP inhibits DNA binding of STAT3 on the Socs3 gene promoter via interaction and colocalisation within the nucleus. Upregulation of inflammatory genes and downregulation of hepatic insulin signalling by acute IL-6 treatment were observed in wild-type mice but not in Shp null mice. Finally, chronic IL-6 exposure caused hepatic insulin resistance, leading to impaired insulin tolerance and elevated gluconeogenesis, and these phenomena were aggravated in Shp null mice. CONCLUSIONS/INTERPRETATION: Our results demonstrate that SHP upregulation by metformin may prevent hepatic disorders by regulating the IL-6-dependent pathway, and that this pathway can help to ameliorate the pathogenesis of cytokine-mediated metabolic dysfunction.


Subject(s)
Hypoglycemic Agents/therapeutic use , Insulin Resistance , Interleukin-6/metabolism , Liver/drug effects , Metformin/therapeutic use , Orphan Nuclear Receptors/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , AMP-Activated Protein Kinases/metabolism , Animals , Insulin/metabolism , Liver/metabolism , Mice , Promoter Regions, Genetic , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
5.
Endocrinology ; 146(5): 2473-80, 2005 May.
Article in English | MEDLINE | ID: mdl-15705774

ABSTRACT

It is almost unanimously accepted that thyrocyte proliferation is synergistically activated by TSH and insulin/IGF-I. Moreover, it was recently suggested that p66Shc, which is an adaptor molecule of the IGF-I receptor, might play a critical role in this synergistic effect. In this study, we undertook to confirm the role and the mechanism underlying the regulation of p66Shc expression via TSH receptor in thyrocytes. We have found that p66Shc expression is elevated in proliferating human thyroid tissues, including adenomatous goiter, adenoma, Graves' disease, and thyroid cancer, but not in normal thyroid. Among growth factors, TSH increased p66Shc expression both in vivo and in vitro; however, IGF-I, epidermal growth factor, or insulin did not. TSH and Graves' Ig increased the p66Shc expression via the TSH receptor-G(s)-cAMP pathway. However, interestingly, IGF-I or epidermal growth factor increased the tyrosine phosphorylations of p66Shc, and this was enhanced by TSH pretreatment. A similar synergism was observed during the DNA synthesis. When we measured the p66Shc levels induced by individual Igs from 130 patients with Graves' disease, TSH receptor stimulating activity and goiter size showed a weak correlation. We conclude that the expression of p66Shc is regulated by signaling through the TSH receptor in proliferating thyroid cells and that p66Shc appears to be an important mediator of the synergistic effect between TSH and IGF-I with respect to thyrocyte proliferation. Moreover, we suggest that TSH potentiates the regulatory effect of IGF-I on thyrocyte growth, at least in part, by increasing the expression of p66Shc.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation/physiology , Receptors, Thyrotropin/physiology , Signal Transduction , Thyroid Gland/metabolism , Adult , Aged , Animals , Autoantibodies/blood , Cell Division/drug effects , Cell Line , Cells, Cultured , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/pharmacology , Female , GTP-Binding Protein alpha Subunits, Gs/physiology , Gene Expression Regulation/drug effects , Goiter/metabolism , Humans , Insulin-Like Growth Factor I/pharmacology , Male , Middle Aged , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Thyrotropin/immunology , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Thyroid Gland/chemistry , Thyroid Gland/cytology , Thyrotropin/pharmacology
6.
J Endocrinol Invest ; 26(2): 132-7, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12739740

ABSTRACT

Disulfide bonds are formed between the sulfhydryl groups in two cysteine residues of a protein. The formation of these bonds is necessary for the proper folding of a protein into its active three-dimensional form. In this study, the genes associated with disulfide bond formation of proteins from the rat thyroid cell line, FRTL-5 cell, were investigated using disulfide bond reducing agent of dithiothreitol (DTT), which prevented disulfide formation of newly synthesized proteins. The expression of six genes, they being the cAMP phosphodiesterase 7A1, neuronal cell death inducible putative kinase (NIPK), cytosolic LIM protein (Ajuba), Eker, early growth response 1 and the ferritin heavy chain, was specifically enhanced under both reductive conditions and various endoplasmic reticulum (ER) stresses inducing drugs such as Brefeldin A (BFA), calcium ionophore A23187 (A23187) and tunicamycin. These results suggest that a suitable redox environment is necessary for the correct disulfide bond conformation in thyrocytes in a complex system.


Subject(s)
Disulfides/metabolism , Gene Expression Regulation/physiology , Proteins/genetics , Thyroid Gland/physiology , Animals , Blotting, Northern , Brefeldin A/pharmacology , Calcimycin , Cell Line , Dithiothreitol/pharmacology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Homeodomain Proteins , Ionophores/pharmacology , LIM Domain Proteins , Nucleic Acid Hybridization , Polymerase Chain Reaction , Protein Conformation , Protein Folding , Protein Synthesis Inhibitors/pharmacology , Proteins/metabolism , RNA/chemistry , RNA/genetics , Rats , Thyroid Gland/metabolism , Tunicamycin/pharmacology
7.
Mol Cell Endocrinol ; 194(1-2): 77-84, 2002 Aug 30.
Article in English | MEDLINE | ID: mdl-12242030

ABSTRACT

The binding of thyrotropin (TSH) to the TSH receptor (TSHR) activates two signaling pathways: the cAMP-protein kinase A (PKA) and the protein kinase C (PKC) systems. We have recently demonstrated that TSH activates the Janus kinases (JAK)/signal transducer and activator of transcription (STAT) pathway via TSHR. This study aimed to investigate whether the cAMP/PKA or the PKC system is involved in STAT3 activation in response to TSH. Treatment with TSH activated STAT3 phosphorylation in FRTL-5 thyrocytes and human TSHR-expressing Chinese hamster ovary cells. TSH-induced STAT3 activation was inhibited by a blocking antibody directed against TSHR that was isolated from patients with primary myxoedema. Increased intracellular cAMP activated STAT3 but inhibition of PKA did not affect STAT3 activation. On the other hand, the PKC stimulant PMA induced STAT3 phosphorylation and the PKC inhibitors inhibited it. Moreover, inhibition of PKC blocked STAT3 activation induced by a stimulator of cAMP. Our data suggest that TSH activates STAT3 via TSHR and cAMP- and PKC-dependent pathways, and provide evidence that PKC may be involved in the pathway downstream from cAMP.


Subject(s)
DNA-Binding Proteins/metabolism , Protein Kinase C/metabolism , Receptors, Thyrotropin/metabolism , Thyroid Gland/cytology , Thyrotropin/physiology , Trans-Activators/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Rats , Receptors, Thyrotropin/genetics , STAT3 Transcription Factor , Signal Transduction , Transfection
8.
Mol Pharmacol ; 60(5): 972-80, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11641425

ABSTRACT

The antithyroid drug, methimazole (MMI) is used to treat patients with Graves' hyperthyroidism. The major action of MMI is to inhibit synthesis of thyroid hormone in the thyroid gland. However, MMI also has antioxidant and immunomodulatory effects on thyrocytes and/or immune cells. This study identifies novel antioxidant and immunomodulatory effects of MMI involving the interferon-gamma (IFN-gamma) signaling pathway in thyroid cells. MMI inhibits transcription of the intercellular adhesion molecule-1 (ICAM-1) gene by modulating the function of transcription factor STAT1 (signal transducer and activator of transcription 1), which binds to the IFN-gamma activated site of the ICAM-1 promoter. Furthermore, MMI rapidly eliminates H(2)O(2) produced by IFN-gamma treatment in thyroid cells and thus inhibits the H(2)O(2)-mediated phosphorylation of tyrosine 701 in STAT1. MMI also eliminates H(2)O(2) in vitro. MMI facilitates electron transfer from NADPH to H(2)O(2) using thioredoxin or glutathione, fulfilling a role similar to peroxiredoxin or glutathione peroxidase, respectively. MMI prevents the IFN-gamma and H(2)O(2)-mediated reversible inactivation of phosphatases. These effects inhibit full activation of the IFN-gamma-induced Janus kinase(JAK)/STAT signaling pathway in FRTL-5 thyroid cells. These results may in part explain the antioxidant and immunomodulatory effects of MMI in thyroid cells of Graves' disease patients.


Subject(s)
Antioxidants/pharmacology , Gene Expression/drug effects , Hydrogen Peroxide/metabolism , Interferon-gamma/pharmacology , Methimazole/pharmacology , Proteins , Proto-Oncogene Proteins , Repressor Proteins , Thyroid Gland/drug effects , Transcription Factors , Adjuvants, Immunologic/pharmacology , Animals , Carrier Proteins/biosynthesis , Cells, Cultured , DNA-Binding Proteins/metabolism , Drug Interactions , Electron Transport/drug effects , Hydrogen Peroxide/pharmacology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Interferon-gamma/antagonists & inhibitors , Janus Kinase 1 , Janus Kinase 2 , Kinetics , Peroxidases/pharmacology , Peroxiredoxins , Phosphorylation/drug effects , Protein Biosynthesis , Protein Inhibitors of Activated STAT , Protein-Tyrosine Kinases/metabolism , Rats , STAT1 Transcription Factor , STAT3 Transcription Factor , Signal Transduction/physiology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Thyroid Gland/cytology , Thyroid Gland/metabolism , Trans-Activators/metabolism , Tyrosine/metabolism
9.
Cell Physiol Biochem ; 11(5): 247-52, 2001.
Article in English | MEDLINE | ID: mdl-11684813

ABSTRACT

Thiol-specific antioxidant (TSA) plays an important role in regulating cell differentiation and proliferation by modulating the hydrogen peroxide (H2O2) mediated responses in a variety of mammalian cells. Thyroid cells are constantly exposed to the actions of reactive oxygen species (ROS), because they produce high levels of H2O2 in response to the physiological action of TSH (thyroid-stimulating hormone). Thyrocytes have several defense mechanisms against ROS, including TSA and SOD (superoxide anion dismutase). Using Northern blot hybridization, we tested the effects of TSH on TSA gene expression in FRTL-5 cells derived from rat thyroids. TSA mRNA expression increased following treatment of cells with TSH at concentrations greater than 10(-9) M. This effect was observed within 6 hours following treatment, and peaked at 8 hours. The effect was blocked by actinomycin D, but not by cycloheximide. The half-life of TSA mRNA was approximately 5.5 hours in the presence or absence of TSH, and that was not affected by TSA mRNA stability. The effects on TSA gene expression were specific to TSH. Other growth factors (e.g., insulin, transferrin and hydrocortisone) did not alter TSA expression. Our results are the first indication that TSH regulates the expression of TSA transcriptionally in thyrocytes.


Subject(s)
Peroxidases/biosynthesis , Thyroid Gland/metabolism , Thyrotropin/pharmacology , Transcriptional Activation , Animals , Cell Line , Colforsin/pharmacology , Hydrogen Peroxide/metabolism , Peroxidases/genetics , Peroxiredoxins , RNA Stability/drug effects , RNA, Messenger/biosynthesis , Rats , Thyroid Gland/cytology
10.
J Immunol ; 167(7): 4038-45, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564824

ABSTRACT

In this report, we describe seven mutations, including a novel single base pair substitution in intron 1, of the Bruton's tyrosine kinase (Btk) gene found in 12 Korean patients with X-linked agammaglobulinemia. Various mutations, including three novel genetic alterations, were discovered using single-strand conformation polymorphism analysis and direct DNA sequencing. The effect of the intron 1 point mutation (intron 1 +5G-->A) was further evaluated using reporter constructs. Using luciferase assay experiments, we showed that the transcriptional activity of the mutant was significantly lower than in normal counterparts, indicating that the intronic mutation was functional. In addition, DNase I footprinting analysis showed that a single protected region spanning the position +3 to +15 bp hybridized with a mutant-specific probe, but not with a wild-type probe. EMSA indicated that a distinct nuclear protein has the ability to bind the mutant oligonucleotides to produce a new DNA-protein complex. We also observed decreased expression of Btk proteins in monocytes of patients having the point mutation in intron 1. Taken together with the functional analysis, our results strongly suggest the existence of a novel cis-acting element, which might be involved in the down-regulation of Btk gene transcription. Precise definition of the regulatory defect in the Btk intron 1 may provide valuable clues toward elucidating the pathogenesis of X-linked agammaglobulinemia.


Subject(s)
Agammaglobulinemia/genetics , Introns , Mutation , Protein-Tyrosine Kinases/genetics , X Chromosome , Adolescent , Adult , Agammaglobulinaemia Tyrosine Kinase , Agammaglobulinemia/diagnosis , Cells, Cultured , Child , Child, Preschool , DNA Footprinting , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/chemistry , Flow Cytometry , Genes, Reporter , Genetic Linkage , Humans , Infant , Korea , Male , Monocytes/metabolism , Pedigree , Point Mutation , Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured
11.
J Immunol ; 166(7): 4481-9, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11254704

ABSTRACT

Fas ligand (FasL), perforin, TNF-alpha, IL-1, and NO have been considered as effector molecule(s) leading to beta cell death in autoimmune diabetes. However, the real culprit(s) in beta cell destruction have long been elusive, despite intense investigation. We and others have demonstrated that FasL is not a major effector molecule in autoimmune diabetes, and previous inability to transfer diabetes to Fas-deficient nonobese diabetic (NOD)-lpr mice was due to constitutive FasL expression on lymphocytes from these mice. Here, we identified IFN-gamma/TNF-alpha synergism as the final effector molecules in autoimmune diabetes of NOD mice. A combination of IFN-gamma and TNF-alpha, but neither cytokine alone, induced classical caspase-dependent apoptosis in insulinoma and pancreatic islet cells. IFN-gamma treatment conferred susceptibility to TNF-alpha-induced apoptosis on otherwise resistant insulinoma cells by STAT1 activation followed by IFN regulatory factor (IRF)-1 induction. IRF-1 played a central role in IFN-gamma/TNF-alpha-induced cytotoxicity because inhibition of IRF-1 induction by antisense oligonucleotides blocked IFN-gamma/TNF-alpha-induced cytotoxicity, and transfection of IRF-1 rendered insulinoma cells susceptible to TNF-alpha-induced cytotoxicity. STAT1 and IRF-1 were expressed in pancreatic islets of diabetic NOD mice and colocalized with apoptotic cells. Moreover, anti-TNF-alpha Ab inhibited the development of diabetes after adoptive transfer. Taken together, our results indicate that IFN-gamma/TNF-alpha synergism is responsible for autoimmune diabetes in vivo as well as beta cell apoptosis in vitro and suggest a novel signal transduction in IFN-gamma/TNF-alpha synergism that may have relevance in other autoimmune diseases and synergistic anti-tumor effects of the two cytokines.


Subject(s)
Apoptosis/immunology , DNA-Binding Proteins/physiology , Diabetes Mellitus, Type 1/immunology , Interferon-gamma/toxicity , Islets of Langerhans/pathology , Phosphoproteins/physiology , Signal Transduction/immunology , Trans-Activators/physiology , Tumor Necrosis Factor-alpha/toxicity , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Caspase 1/biosynthesis , Caspases/biosynthesis , Caspases, Initiator , Cell Death/immunology , Cells, Cultured , Cytotoxicity Tests, Immunologic , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/prevention & control , Drug Synergism , Enzyme Induction/immunology , Immune Sera/administration & dosage , Infusions, Intravenous , Interferon Regulatory Factor-1 , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Mice , Mice, Inbred ICR , Mice, Inbred NOD , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Phosphorylation , STAT1 Transcription Factor , Trans-Activators/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/immunology
12.
J Mol Endocrinol ; 26(2): 127-33, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11241164

ABSTRACT

This study was performed to evaluate the effects of thyroid-stimulating hormone (TSH) on phosphatidylinositol-4-phosphate 5-kinase type IIgamma (PIPKIIgamma) gene expression in the thyrocytes of FRTL-5 cells. Although PIPKIIgamma mRNA was expressed constantly in the absence of added TSH, its expression increased remarkably in the presence of 10(-9) M TSH. This increase started within 6 h of the addition of TSH, and reached a maximum at 8 h. The mRNA expression properties of PIPKIIgamma in the cells were identified using inhibitors. Actinomycin D blocked PIPKIIgamma transcription strongly, while cycloheximide did not. In an experiment using 5,6-dichlo-1-beta-d -ribofuranosylbenzimidaxole, the half-life of PIPKIIgamma mRNA was approximately 6 h in the presence or absence of TSH, and it was not affected by the stability of the PIPKIIgamma mRNA. The effects of TSH on PIPKIIgamma gene expression were specific, and other growth factors examined (transferrin, insulin and hydrocortisone) did not alter its expression. It is possible that the mechanism of PIPKIIgamma gene expression is involved in the permissive effect of the TSH-cAMP cascade proper. Our results indicate, for the first time, that the expression of PIPKIIgamma is regulated transcriptionally by TSH in thyrocytes.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Thyroid Gland/drug effects , Thyrotropin/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Blotting, Northern , Cell Line , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dichlororibofuranosylbenzimidazole/pharmacology , Enzyme Inhibitors/pharmacology , Hydrocortisone/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Thyroid Gland/cytology , Thyroid Gland/enzymology , Time Factors , Transferrin/pharmacology
13.
Eur J Biochem ; 267(20): 6180-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11012671

ABSTRACT

The Gadd45gamma (growth arrest and DNA damage-inducible) gene is activated transcriptionally by at least two kinds of agents: DNA damaging agent such as methyl methanesulfonate (MMS) and UV radiation, or cytokines such as interleukin (IL)-6, IL-2 and granulocyte colony-stimulating factor (G-CSF). To investigate the sequences and transcription factors involved in induction of Gadd45gamma after treatment with IL-6, the human gene was cloned and sequenced. We found C/EBP (CCAAT/enhancer-binding protein) family proteins, major transcription factors in the IL-6 signal transduction pathway, could regulate the transcriptional activity of the Gadd45gamma promoter. In addition, a noncanonical C/EBP-binding site within the Gadd45gamma promoter where C/EBPbeta and C/EBPdelta could bind, was identified by electrophoretic mobility shift assay (EMSA) and reporter gene analysis. Furthermore, we found a coordinated expression profile between Gadd45gamma mRNA and C/EBPs (beta and delta) protein during the differentiation of M1 cells: the amount of Gadd45gamma transcripts became maximal when both C/EBPbeta and C/EBPdelta levels were high, on day 1 of differentiation of M1 cells after treatment with IL-6. These findings suggest that mitotic growth arrest coupled to M1 cell differentiation is mediated by C/EBPs stimulation of growth arrest-associated genes such as Gadd45gamma.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Gene Expression Regulation , Promoter Regions, Genetic , Proteins/genetics , 3T3 Cells , Animals , Base Sequence , Binding Sites , Cloning, Molecular , DNA Damage , Gene Expression Regulation/drug effects , Humans , Interleukin-6/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Open Reading Frames , Protein Isoforms/metabolism , RNA, Messenger/genetics , Recombinant Proteins/biosynthesis , Transcription Factors/metabolism , Transcription, Genetic , Transfection , GADD45 Proteins
14.
Z Naturforsch C J Biosci ; 55(5-6): 449-54, 2000.
Article in English | MEDLINE | ID: mdl-10928558

ABSTRACT

The transcriptional expression of an ischemia responsive protein (irp94) in the hippocampus of rats was analyzed by Northern blotting. A transient forebrain ischemia was induced in the rats by temporary occluding of the bilateral common carotid arteries (CCAs) for various periods, and then reperfusion. Among the frontal, parietal, temporal and occipital lobes, and the cerebellum and hippocampus, the maximum mRNA expression of irp94 was at the occipital lobe, and the minimum was at the parietal lobe following ten min of forebrain ischemia. The irp94 mRNA expression reached a maximum fifteen min after the transient ischemia. From twenty min on after the ischemia its expression decreased. After a ten-min ischemia and the following reperfusion, irp94 mRNA expression gradually increased in the first twelve h, and then decreased. The expression pattern was like that of the endoplasmic reticulum chaperone, Erp72, but not that of the cytosol chaperone, hsp72. In addition, when intracellular ATP was depleted with antimycin A the mRNA level of irp94 increased in a thyrocyte cell culture model. The results suggest that irp94, like a molecular chaperone, may play a role in protecting the cell against external stimulation, especially after a transient forebrain ischemia. Although future studies of irp94 will be required to clarify the interactions with other intracellular factors inducing ischemia or showing molecular chaperone activity, what is offered here is an insight into its functional role as a component of stress response in neurons that should be considered as a new therapeutic approach for the treatment of ischemia.


Subject(s)
Brain/metabolism , Gene Expression Regulation , HSP70 Heat-Shock Proteins/genetics , Ischemic Attack, Transient/metabolism , Animals , Cells, Cultured , HSP110 Heat-Shock Proteins , Male , Prosencephalon , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic
15.
Endocrinology ; 141(6): 2090-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10830295

ABSTRACT

TSH is known as an important hormone that plays the major role not only in the maintenance of normal physiology but also in the regulation of immunomodulatory gene expression in thyrocytes. The adhesion molecule intercellular adhesion molecule-1 (ICAM-1) was identified as one of the proteins that are abnormally expressed in the thyroid gland during autoimmune thyroid diseases. In this study we found that TSH inhibits interferon-gamma (IFNgamma)-mediated expression of the ICAM-1 gene, and we investigated the involved mechanisms in rat FRTL-5 thyroid cells. After exposure to IFNgamma, ICAM-1 expression is positively regulated at the level of transcription. This effect occurs via the IFNgamma-activated site (GAS) element in the ICAM-1 promoter as a consequence of the activation of STAT1 (signal transducer and activator of transcription-1), but not of STAT3. On the other hand, after exposure to TSH plus IFNgamma, ICAM-1 transcription is negatively modulated. We found that this inhibitory effect of TSH also occurs via the GAS element. Electrophoretic mobility shift assays confirmed that the IFNgamma-induced DNA-binding activities of STAT1 were reduced by TSH. Furthermore, our results showed that the inhibitory effect of TSH on IFNgamma signaling is caused by inhibition of tyrosine phosphorylation on STAT1, Janus kinase-1 (Jak1), and IFNgamma receptor a, but not Jak2. In conclusion, we have identified a novel mechanism in which TSH modulates the IFNgamma-mediated Jak/STAT signaling pathway through the inhibition of Jak1 and STAT1.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression/drug effects , Intercellular Adhesion Molecule-1/genetics , Interferon-gamma/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Thyrotropin/pharmacology , Trans-Activators/metabolism , Animals , Cattle , Cell Line , DNA/metabolism , Enzyme Inhibitors/pharmacology , Janus Kinase 1 , Phosphotyrosine/metabolism , Rats , STAT1 Transcription Factor , Signal Transduction/drug effects , Thyroid Gland/metabolism
16.
J Biol Chem ; 275(24): 18266-70, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10849441

ABSTRACT

Peroxiredoxins (Prxs) play an important role in regulating cellular differentiation and proliferation in several types of mammalian cells. One mechanism for this action involves modulation of hydrogen peroxide (H(2)O(2))-mediated cellular responses. This report examines the expression of Prx I and Prx II in thyroid cells and their roles in eliminating H(2)O(2) produced in response to thyrotropin (TSH). Prx I and Prx II are constitutively expressed in FRTL-5 thyroid cells. Prx I expression, but not Prx II expression, is stimulated by exposure to TSH and H(2)O(2). In addition, methimazole induces a high level of Prx I mRNA and protein in these cells. Overexpression of Prx I and Prx II enhances the elimination of H(2)O(2) produced by TSH in FRTL-5 cells. Treatment with 500 micrometer H(2)O(2) causes apoptosis in FRTL-5 cells as evidenced by standard assays of apoptosis (i.e. terminal deoxynucleotidyl transferase deoxyuridine triphosphate-biotin nick end labeling, BAX expression, and poly(ADP-ribose) polymerase cleavage. Overexpression of Prx I and Prx II reduces the amount of H(2)O(2)-induced apoptosis measured by these assays. These results suggest that Prx I and Prx II are involved in the removal of H(2)O(2) in thyroid cells and can protect these cells from undergoing apoptosis. These proteins are likely to be involved in the normal physiological response to TSH-induced production of H(2)O(2) in thyroid cells.


Subject(s)
Apoptosis , Hydrogen Peroxide/metabolism , Peroxidases/physiology , Thyroid Gland/cytology , Animals , Cattle , Cells, Cultured , Iodides/pharmacology , Peroxidases/genetics , Peroxiredoxins , RNA, Messenger/metabolism , Rabbits , Rats , Thyrotropin/pharmacology
17.
FEBS Lett ; 475(1): 27-30, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10854851

ABSTRACT

This experiment was performed to evaluate the effect of thyroid-stimulating hormone (TSH) on the endoplasmic reticulum resident 29 kDa protein (ERp29) gene expression in the thyrocytes of FRTL-5 cells. Although ERp29 mRNA was constantly expressed, its expression began to increase remarkably from 10(-9) M TSH. On the other hand, the effect of TSH on the abundance of ERp29 mRNA started within 6 h and peaked at 8 h. Actinomycin D strongly blocked this effect while cycloheximide did not. The half-life of ERp29 mRNA was about 4-4.5 h in the presence or absence of TSH that was not affected by the stability of ERp29 mRNA. The effect of TSH on the ERp29 gene expression was specific, while other growth factors (transferrin, insulin and hydrocortisone) did not alter its expression. Our data indicate for the first time that the expression of ERp29 is regulated transcriptionally by TSH in thyrocytes.


Subject(s)
Gene Expression Regulation/drug effects , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Thyroid Gland/metabolism , Thyrotropin/pharmacology , Cell Line , Endoplasmic Reticulum/metabolism , Thyroid Gland/ultrastructure
18.
Mol Endocrinol ; 14(5): 662-70, 2000 May.
Article in English | MEDLINE | ID: mdl-10809230

ABSTRACT

TSH is an important physiological regulator of growth and function in thyroid gland. The mechanism of action of TSH depends on interaction with its receptor coupled to heterotrimeric G proteins. We show here that TSH induces the phosphorylation of tyrosine in the intracellular kinases Janus kinase 1 (JAK1) and -2 (JAK2) in rat thyroid cells and in Chinese hamster ovary (CHO) cells transfected with human TSH receptor (TSHR). The JAK family substrates STAT3 (signal transducers and activators of transcription) are rapidly tyrosine phosphorylated in response to TSH. We also find that JAK1, JAK2, and STAT3 coprecipitate with the TSHR, indicating that the TSHR may be able to signal through the intracellular phosphorylation pathway used by the JAK-STAT cascade. TSH increases STAT3-mediated promoter activity and also induces endogenous SOCS-1 (suppressor of cytokine signaling-1) gene expression, a known target gene of STAT3. The expression of a dominant negative form of STAT3 completely inhibited TSH-mediated SOCS-1 expression. These findings suggest that the TSHR is able to signal through JAK/STAT3 pathways.


Subject(s)
DNA-Binding Proteins/physiology , Intracellular Signaling Peptides and Proteins , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins , Repressor Proteins , Signal Transduction , Thyroid Gland/drug effects , Thyrotropin/physiology , Trans-Activators/physiology , Animals , CHO Cells , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cells, Cultured , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Dominant , Humans , Janus Kinase 1 , Janus Kinase 2 , Macromolecular Substances , Phosphorylation , Protein Processing, Post-Translational , Rats , Receptors, Thyrotropin/drug effects , Receptors, Thyrotropin/metabolism , Recombinant Fusion Proteins/physiology , STAT1 Transcription Factor , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Thyroid Gland/cytology , Thyrotropin/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic
19.
Mol Endocrinol ; 14(3): 440-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10707961

ABSTRACT

TSH has multiple physiological roles: it is required for growth, differentiation, and function of the thyroid gland, and it regulates transcription of interferon-gamma (IFN-gamma)-responsive genes in thyrocytes, including genes for the major histocompatibility complex and intercellular adhesion molecule-1. This report demonstrates that TSH induces the expression of suppressor of cytokine signaling (SOCS)-1 and -3 proteins and alters the phosphorylation state of signal transducer and activator of transcription (STAT) proteins STAT1 and STAT3. The expression of SOCS-1 and SOCS-3 and the phosphorylation state of STAT1 and STAT3 were examined after treatment with TSH or IFN-gamma in either TSH-sensitive FRTL-5 thyroid cells or TSH-insensitive FRT and buffalo rat liver (BRL) cells, which lack functional TSH receptors. SOCS-1 and SOCS-3 are constitutively expressed in FRTL-5 cells, but not in FRT and BRL cells. IFN-gamma up-regulated SOCS-1 and SOCS-3 RNA and protein in FRTL-5 cells, as reported previously for nonthyroid cells. Interestingly, TSH also significantly induced SOCS-1 and SOCS-3 in FRTL-5 cells, but not in FRT and BRL cells. When SOCS-1 or SOCS-3 was overexpressed in FRTL-5 cells, STAT1 phosphorylation at Y701 and STAT1/DNA complex formation in response to IFN-gamma were reduced. Furthermore, overexpression of either SOCS-1 or SOCS-3 significantly inhibited the IFN-gamma-mediated transactivation of the rat ICAM-1 (intercellular adhesion molecule-1) promoter. TSH and IFN-gamma had different effects on STAT1 and STAT3 phosphorylation. The phosphorylation of Y701 in STAT1, which is responsible for homodimer formation, nuclear translocation, and DNA binding, was specifically stimulated by IFN-gamma, but not by TSH or forskolin. However, the phosphorylation of S727 in STAT1 was induced by IFN-gamma, TSH, and forskolin. TSH induced phosphorylation of both Y705 and S727 in STAT3, while IFN-gamma phosphorylated only the Y705. In addition, we found that SOCS-3 was associated with JAK1 and JAK2 and that these associations were stimulated by TSH. These findings demonstrate that TSH induces SOCS in thyroid cells and provides the evidence of signal cross-talk between TSH and cytokines in thyroid cells.


Subject(s)
Carrier Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins , Protein Biosynthesis , Repressor Proteins , Signal Transduction/drug effects , Thyroid Gland/drug effects , Thyrotropin/pharmacology , Transcription Factors , Animals , Antithyroid Agents/pharmacology , Carrier Proteins/genetics , Cell Line , Colforsin/pharmacology , DNA-Binding Proteins/metabolism , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Interferon-gamma/pharmacology , Liver/drug effects , Liver/metabolism , Methimazole/pharmacology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/genetics , Rats , Recombinant Proteins , STAT1 Transcription Factor , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Thyroid Gland/metabolism , Trans-Activators/metabolism
20.
Z Naturforsch C J Biosci ; 54(7-8): 573-7, 1999.
Article in English | MEDLINE | ID: mdl-10488566

ABSTRACT

We established a relationship between the toxic effects of cadmium on the expression of the endoplasmic reticulum (ER) chaperone GRP94 (glucose regulated protein 94) and cell survival in cultured rat-thyrocytes of FRTL5 cells. There are no data reporting that the enhanced expression of GRP94 by Cd stimulation is detectable in thyrocytes. Western blot analysis revealed higher levels of GRP94 expression in those cells post-treated with low concentrations of Cd, following a step-down treatment method, than in Cd pre-treated cells or cells not treated with any Cd, due to changes in cellular sensitivity after pre-treatment with Cd and the possible induction of GRP94 expression after removal of a low concentration of Cd. Elevated GRP94 expression in thyrocytes post-treated with Cd confers a survival advantage by rendering them resistant to cytotoxic stress, and the existence in the thyrocytes of a Cd-specific pathway regulates the expression of stress proteins by Cd.


Subject(s)
Cadmium/pharmacology , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , Membrane Proteins/biosynthesis , Molecular Chaperones/biosynthesis , Thyroid Gland/drug effects , Cell Line , Thyroid Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...