Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Endocrinology ; 141(2): 606-14, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10650941

ABSTRACT

TSH stimulates proliferation and maintains differentiated function in thyroid follicular cells. The mitogenic activity and the stimulatory effects of TSH on thyroid-specific gene expression are impaired by interferon-gamma (IFNgamma); however, the mechanisms for these effects have not been elucidated in detail. We examined the effects of IFNgamma on acute responses to TSH in rat thyroid cells. IFNgamma did not impair TSH-stimulated p70/p85 ribosomal protein S6 kinase (p70/p85s6k) activity or cAMP response element (CRE)-regulated gene expression, although it inhibited DNA synthesis and thyroglobulin expression, effects measured over a more prolonged time course than those on kinase activity and reporter gene expression. Unexpectedly, when cells were chronically exposed to IFNgamma, CRE-lacZ promoter activity was decreased, whereas other cAMP-mediated signals, such as p70/p85s6k activity and CRE-binding protein phosphorylation, were unaffected. Activating protein-1-regulated promoters were also impaired by IFNgamma treatment, but with kinetics that differed from those of CRE-regulated promoters. Neither acute nor chronic treatment with interleukin-1beta impaired cAMP signaling, indicating that the effects of IFNgamma are specific. These studies identify CRE- and activating protein-1-regulated promoters as targets of IFNgamma in thyroid cells and fibroblasts. IFNgamma-mediated inhibition of these promoters, in addition to those containing thyroid-specific transcription factor-1-binding sites, may contribute to the profound effects of IFNgamma on thyroid cells.


Subject(s)
Cyclic AMP/metabolism , Gene Expression Regulation/physiology , Genes, MHC Class I , Interferon-gamma/pharmacology , Thyroid Gland/physiology , Thyrotropin/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/genetics , Interleukin-1/pharmacology , Kinetics , Rats , Rats, Wistar , Recombinant Proteins , Ribosomal Protein S6 Kinases/metabolism , Thyroid Gland/cytology , Thyroid Gland/drug effects , Transcription Factor AP-1/metabolism
2.
Endocrinology ; 141(1): 146-52, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10614633

ABSTRACT

Several reports have indicated that protein kinase C (PKC) is an important regulator of proliferation in thyroid cells. Unlike TSH, the mitogenic effects of phorbol esters are accompanied by de-differentiation. The role of individual PKC isoforms in thyroid cell proliferation and differentiation has not been examined. Recent studies have implicated the atypical PKCzeta, a phorbol ester-unresponsive isozyme, in cell proliferation, death, and survival. We overexpressed PKCzeta in Wistar rat thyroid (WRT) cells and determined that PKCzeta conferred TSH-independent DNA synthesis and cell proliferation. Cells overexpressing PKCzeta show higher levels of phosphorylated p42/p44 MAPK compared with vector-transfected cells. Experiments using a luciferase reporter for Elk-1 revealed that PKCzeta overexpressing cells exhibit higher basal Elk-1 transcriptional activity than vector-transfected control cells. Interestingly, stimulation of Elk-1 transcriptional activity by MEK1, a p42/p44 MAPK kinase, was significantly enhanced in cells overexpressing PKCzeta. Strikingly, TSH retained the ability to stimulate Tg expression in cells expressing PKCzeta. These results suggest that PKCzeta stimulates TSH-independent mitogenesis through a p42/p44 MAPK-dependent pathway. Unlike overexpression of Ras or phorbol ester treatment, PKC overexpression does not impair thyroglobulin (Tg) expression.


Subject(s)
DNA-Binding Proteins , Protein Kinase C/physiology , Thyroid Gland/cytology , Thyrotropin/physiology , Animals , Blotting, Western , Cell Division , Cells, Cultured , DNA/biosynthesis , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/physiology , Genetic Vectors/genetics , Mitogen-Activated Protein Kinase 1/physiology , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Proto-Oncogene Proteins/genetics , Rats , Rats, Wistar , Transcription Factors/genetics , ets-Domain Protein Elk-1
3.
Mol Cell Biol ; 19(9): 5882-91, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454535

ABSTRACT

The effects of cyclic AMP (cAMP) on cell proliferation are cell type specific. Although the growth-inhibitory effects of cAMP have been well studied, much less is known regarding how cAMP stimulates proliferation. We report that cAMP stimulates proliferation through both protein kinase A (PKA)-dependent and PKA-independent signaling pathways and that phosphatidylinositol 3-kinase (PI3K) is required for cAMP-stimulated mitogenesis. In cells where cAMP is a mitogen, cAMP-elevating agents stimulate membrane ruffling, Akt phosphorylation, and p70 ribosomal S6 protein kinase (p70s6k) activity. cAMP effects on ruffle formation and Akt were PKA independent but sensitive to wortmannin. In contrast, cAMP-stimulated p70s6k activity was repressed by PKA inhibitors but not by wortmannin or microinjection of the N-terminal SH2 domain of the p85 regulatory subunit of PI3K, indicating that p70s6k and Akt can be regulated independently. Microinjection of highly specific inhibitors of PI3K or Rac1, or treatment with the p70s6k inhibitor rapamycin, impaired cAMP-stimulated DNA synthesis, demonstrating that PKA-dependent and -independent pathways contribute to cAMP-mediated mitogenesis. Direct elevation of PI3K activity through microinjection of an antibody that stimulates PI3K activity or stable expression of membrane-localized p110 was sufficient to confer hormone-independent DNA synthesis when accompanied by elevations in p70s6k activity. These findings indicate that multiple pathways contribute to cAMP-stimulated mitogenesis, only some of which are PKA dependent. Furthermore, they demonstrate that the ability of cAMP to stimulate both p70s6k- and PI3K-dependent pathways is an important facet of cAMP-regulated cell cycle progression.


Subject(s)
Cell Division/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Protein Serine-Threonine Kinases , Signal Transduction/physiology , 3T3 Cells , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Amino Acid Sequence , Animals , Cell Division/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/ultrastructure , DNA/biosynthesis , Enzyme Inhibitors/pharmacology , Mice , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects
4.
Mol Cell Biol ; 18(7): 3718-26, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9632754

ABSTRACT

Ras mutants with the ability to interact with different effectors have played a critical role in the identification of Ras-dependent signaling pathways. We used two mutants, RasS35 and RasG37, which differ in their ability to bind Raf-1, to examine Ras-dependent signaling in thyroid epithelial cells. Wistar rat thyroid cells are dependent upon thyrotropin (TSH) for growth. Although TSH-stimulated mitogenesis requires Ras, TSH activates protein kinase A (PKA) and downregulates signaling through Raf and the mitogen-activated protein kinase (MAPK) cascade. Cells expressing RasS35, a mutant which binds Raf, or RasG37, a mutant which binds RalGDS, exhibited TSH-independent proliferation. RasS35 stimulated morphological transformation and anchorage-independent growth. RasG37 stimulated proliferation but not transformation as measured by these indices. TSH exerted markedly different effects on the Ras mutants and transiently repressed MAPK phosphorylation in RasS35-expressing cells. In contrast, TSH stimulated MAPK phosphorylation and growth in cells expressing RasG37. The Ras mutants, in turn, exerted differential effects on TSH signaling. RasS35 abolished TSH-stimulated changes in cell morphology and thyroglobulin expression, while RasG37 had no effect on these activities. Together, the data indicate that cross talk between Ras and PKA discriminates between distinct Ras effector pathways.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cell Line , Cyclic AMP/metabolism , Gene Expression Regulation , Mutagenesis, Site-Directed , Phosphorylation , Rats , Rats, Wistar , Thyroglobulin/genetics , Thyroid Gland , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL