Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Cell Biol ; 125(3): 557-71, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-8175881

RESUMO

The Golgi apparatus is a dynamic organelle whose structure is sensitive to vesicular traffic and to cell cycle control. We have examined the potential role for rab1a, a GTPase previously associated with ER to Golgi and intra-Golgi transport, in the formation and maintenance of Golgi structure. Bacterially expressed, recombinant rab1a protein was microinjected into rat embryonic fibroblasts, followed by analysis of Golgi morphology by fluorescence and electron microscopy. Three recombinant proteins were tested: wild-type rab, mutant rab1a(S25N), a constitutively GDP-bound form (Nuoffer, C., H. W. Davidson, J. Matteson, J. Meinkoth, and W. E. Balch, 1994. J. Cell Biol. 125: 225-237), and mutant rab1a(N124I) defective in guanine nucleotide binding. Microinjection of wild-type rab1a protein or a variety of negative controls (injection buffer alone or activated ras protein) did not affect the appearance of the Golgi, as visualized by immunofluorescence of alpha-mannosidase II (Man II), used as a Golgi marker. In contrast, microinjection of the mutant forms promoted the disassembly of the Golgi stacks into dispersed vesicular structures visualized by immunofluorescence. When S25N-injected cells were analyzed by EM after immunoperoxidase labeling, Man II was found in isolated ministacks and large vesicular elements that were often surrounded by numerous smaller unlabeled vesicles resembling carrier vesicles. Golgi disassembly caused by rab1a mutants differs from BFA-induced disruption, since beta-COP remains membrane associated, and Man II does not redistribute to the ER. BFA can still cause these residual Golgi elements to fuse and disperse, albeit at a slower rate. Moreover, BFA recovery is incomplete in the presence of rab1 mutants or GTP gamma S. We conclude that GTP exchange and hydrolysis by GTPases, specifically rab1a, are required to form and maintain normal Golgi stacks. The similarity of Golgi disassembly seen with rab1a mutants to that occurring during mitosis, may point to a molecular basis involving rab1a for fragmentation of the Golgi apparatus during cell division.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Complexo de Golgi/ultraestrutura , Animais , Brefeldina A , Células Cultivadas , Proteína Coatomer , Ciclopentanos/farmacologia , GTP Fosfo-Hidrolases/metabolismo , GTP Fosfo-Hidrolases/farmacologia , Proteínas de Ligação ao GTP/genética , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Técnicas In Vitro , Manosidases/metabolismo , Microinjeções , Microscopia Eletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Ratos , Proteínas Recombinantes , Proteínas rab1 de Ligação ao GTP
2.
Mol Cell Biol ; 13(8): 4477-84, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8336696

RESUMO

Microinjection of a dominant interfering mutant of Ras (N17 Ras) caused a significant reduction in thyrotropin (thyroid-stimulating hormone [TSH])-stimulated DNA synthesis in rat thyroid cells. A similar reduction was observed following injection of the heat-stable protein kinase inhibitor of the cyclic AMP-dependent protein kinase. Coinjection of both inhibitors almost completely abolished TSH-induced DNA synthesis. In contrast to TSH, overexpression of cellular Ras protein did not stimulate the expression of a cyclic AMP response element-regulated reporter gene. Similarly, injection of N17 Ras had no effect on TSH-stimulated reporter gene expression. Moreover, overexpression of cellular Ras protein stimulated similar levels of DNA synthesis in the presence or absence of the heat-stable protein kinase inhibitor. Together, these results suggest that in Wistar rat thyroid cells, a full mitogenic response to TSH requires both Ras and cyclic APK-dependent protein kinase.


Assuntos
Divisão Celular/efeitos dos fármacos , DNA/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Glândula Tireoide/citologia , Tireotropina/antagonistas & inibidores , Animais , Proteínas de Transporte/farmacologia , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Microinjeções , Proteínas Proto-Oncogênicas p21(ras)/administração & dosagem , Ratos , Transdução de Sinais
3.
Mol Cell Biol ; 11(3): 1759-64, 1991 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1847503

RESUMO

We constructed cell lines containing various enhancer elements cloned upstream from a marker gene. By microinjection of specific antibodies directed against transcriptional activator proteins into these cell lines, we have developed a functional assay for factors which regulate the activity of target promoters. Here we show that microinjection of a highly specific antibody to the cyclic AMP enhancer element-binding (CREB) protein diminishes gene expression in response to cyclic AMP in living fibroblasts.


Assuntos
AMP Cíclico/fisiologia , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Animais , Linhagem Celular , Clonagem Molecular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Elementos Facilitadores Genéticos , Técnicas Imunológicas , Microinjeções , Ratos
4.
Mol Cell Biol ; 21(6): 1921-9, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238928

RESUMO

Originally identified as an antagonist of Ras action, Rap1 exhibits many Ras-independent effects, including a role in signaling pathways initiated by cyclic AMP (cAMP). Since cAMP is a critical mediator of the effects of thyrotropin (TSH) on cell proliferation and differentiation, we examined the regulation of Rap1 by TSH in a continuous line of rat thyroid-like cells. Both cAMP and protein kinase A (PKA) contribute to the regulation of Rap1 activity and signaling by TSH. TSH activates Rap1 through a cAMP-mediated and PKA-independent mechanism. TSH phosphorylates Rap1 in a PKA-dependent manner. Interference with PKA activity blocked phosphorylation but not the activation of Rap1. Rather, PKA inhibitors prolonged Rap1 activation, as did expression of a Rap1A mutant lacking a PKA phosphorylation site. These results indicate that PKA elicits negative feedback regulation on cAMP-stimulated Rap1 activity in some cells. The dual regulation of Rap1 by cAMP and PKA extends to downstream effectors. The ability of TSH to stimulate Akt phosphorylation was markedly enhanced by the expression of activated Rap1A and was repressed in cells expressing a putative dominant-negative Rap1A mutant. Although the expression of activated Rap1A was sufficient to stimulate wortmannin-sensitive Akt phosphorylation, TSH further increased Akt phosphorylation in a phosphatidylinositol 3-kinase- and PKA-dependent manner. The ability of TSH to phosphorylate Akt was impaired in cells expressing a Rap1A mutant that could be activated but not phosphorylated. These findings indicate that dual signals, Rap1 activation and phosphorylation, contribute to TSH-stimulated Akt phosphorylation. Rap1 plays an essential role in cAMP-regulated differentiation. TSH effects on thyroid-specific gene expression, but not its effects on proliferation, were markedly enhanced in cells expressing activated Rap1A and repressed in cells expressing a dominant-negative Rap1A mutant. These findings reveal complex regulation of Rap1 by cAMP including PKA-independent activation and PKA-dependent negative feedback regulation. Both signals appear to be required for TSH signaling to Akt.


Assuntos
Diferenciação Celular/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas Serina-Treonina Quinases , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Genes ras , Mutação , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Wistar , Glândula Tireoide/efeitos dos fármacos , Tireotropina/metabolismo , Tireotropina/farmacologia , Proteínas rap1 de Ligação ao GTP/genética
5.
Mol Cell Biol ; 19(9): 5882-91, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10454535

RESUMO

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.


Assuntos
Divisão Celular/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas Serina-Treonina Quinases , Transdução de Sinais/fisiologia , Células 3T3 , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Sequência de Aminoácidos , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/ultraestrutura , DNA/biossíntese , Inibidores Enzimáticos/farmacologia , Camundongos , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Mol Cell Biol ; 18(7): 3718-26, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9632754

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Transdução de Sinais , Proteínas ras/metabolismo , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Divisão Celular , Linhagem Celular , AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Mutagênese Sítio-Dirigida , Fosforilação , Ratos , Ratos Wistar , Tireoglobulina/genética , Glândula Tireoide , Proteínas ras/genética
7.
Mol Cell Biol ; 13(4): 2104-12, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8384305

RESUMO

The involvement of serine/threonine protein phosphatases in signaling pathways which modulate the activity of the transcription factor AP-1 was examined. Purified protein phosphatase types 1 (PP1) and 2A (PP2A) were microinjected into cell lines containing stably transfected lacZ marker genes under the control of an enhancer recognized by AP-1. Microinjection of PP2A potentiated serum-stimulated beta-galactosidase expression from the AP-1-regulated promoter. Similarly, transient expression of the PP2A catalytic subunit with c-Jun resulted in a synergistic transactivation of an AP-1-regulated reporter gene. PP2A, but not PP1, potentiated serum-induced c-Jun expression, which has been previously shown to be autoregulated by AP-1 itself. Consistent with these results, PP2A dephosphorylated c-Jun on negative regulatory sites in vitro, suggesting one possible direct mechanism for the effects of PP2A on AP-1 activity. Microinjection of PP2A had no effect on cyclic AMP (cAMP)-induced expression of a reporter gene containing a cAMP-regulated promoter, while PP1 injection abolished cAMP-induced gene expression. Taken together, these results suggest a specific role for PP2A in signal transduction pathways that regulate AP-1 activity and c-Jun expression.


Assuntos
Regulação da Expressão Gênica , Fosfoproteínas Fosfatases/fisiologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun/metabolismo , Animais , Sequência de Bases , Bovinos , Técnicas In Vitro , Microinjeções , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos/química , Fosforilação , Proteína Fosfatase 2 , Ativação Transcricional
8.
Mol Cell Biol ; 7(5): 1740-50, 1987 May.
Artigo em Inglês | MEDLINE | ID: mdl-2885742

RESUMO

In a previous study (G. M. Wahl, B. Robert de Saint Vincent, and M. L. De Rose, Nature (London) 307:516-520, 1984), we used gene transfer of a CAD cosmid to demonstrate that gene position profoundly affects amplification frequency. One transformant, T5, amplified the donated CAD genes at a frequency at least 100-fold higher than did the other transformants analyzed. The CAD genes in T5 and two drug-resistant derivatives were chromosomally located. In this report, we show that a subclone of T5 gives rise to an extrachromosomal molecule (CAD episome) containing the donated CAD genes. Gel electrophoresis indicated that the CAD episome is approximately 250 to 300 kilobase pairs, and a variety of methods showed that it is a covalently closed circle. We show that the CAD episome replicates semiconservatively and approximately once per cell cycle. Since the CAD cosmid, which comprises most of the CAD episome, does not replicate autonomously when transfected into cells, our results indicate that either the process which generated the episome resulted in a cellular origin of DNA replication being linked to the CAD sequences or specific rearrangements within the episome generated a functional origin. The implications of these results for mechanisms of gene amplification and the genesis of minute chromosomes are discussed.


Assuntos
Replicação do DNA , Amplificação de Genes , Genes Reguladores , Complexos Multienzimáticos/genética , Plasmídeos , Proteínas/genética , Animais , Aspartato Carbamoiltransferase/genética , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/genética , Mapeamento Cromossômico , Cosmídeos , Cricetinae , Enzimas de Restrição do DNA , Di-Hidro-Orotase/genética , Cariotipagem , Transfecção
9.
Mol Biol Cell ; 4(10): 993-1002, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8298196

RESUMO

The catalytic (C) subunit of cyclic AMP (cAMP) dependent protein kinase (PKA) has previously been shown to enter and exit the nucleus of cells when intracellular cAMP is raised and lowered, respectively. To determine the mechanism of nuclear translocation, fluorescently labeled C subunit was injected into living REF52 fibroblasts either as free C subunit or in the form of holoenzyme (PKA) in which the catalytic and regulatory subunits were labeled with fluorescein and rhodamine, respectively. Quantification of nuclear and cytoplasmic fluorescence intensities revealed that free C subunit nuclear accumulation was most similar to that of macromolecules that diffuse into the nucleus. A glutathione S-transferase-C subunit fusion protein did not enter the nucleus following cytoplasmic microinjection. Puncturing the nuclear membrane did not decrease the nuclear concentration of C subunit, and C subunit entry into the nucleus did not appear to be saturable. Cooling or depleting cells of energy failed to block movement of C subunit into the nucleus. Photobleaching experiments showed that even after reaching equilibrium at high [cAMP], individual molecules of C subunit continued to leave the nucleus at approximately the same rate that they had originally entered. These results indicate that diffusion is sufficient to explain most aspects of C subunit subcellular localization.


Assuntos
Núcleo Celular/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Animais , Transporte Biológico Ativo/fisiologia , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/imunologia , Citoplasma/metabolismo , Difusão , Ativação Enzimática/fisiologia , Fibroblastos , Imunofluorescência , Cobaias , Histonas/metabolismo , Camundongos , Membrana Nuclear/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Albumina Sérica/metabolismo , Espectrometria de Fluorescência/métodos , Inibidores da Tripsina/metabolismo
10.
Oncogene ; 20(50): 7334-41, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11704863

RESUMO

Ras mutations occur at high frequency in thyroid cancer. In vitro, the effects of Ras in thyroid cells are pleiotropic in that expression of activated Ras has been reported to stimulate proliferation and apoptosis. An understanding of the factors that contribute to the survival versus demise of Ras-transformed cells is essential to our understanding of the contribution of Ras to thyroid neoplasia and other cancers. Constitutive expression of oncogenic H-Ras sensitized Wistar rat thyroid (WRT) cells to apoptosis stimulated by multiple insults. When deprived of matrix attachment, Ras-transformed cells perished by apoptotic cell death at a high frequency. In contrast, parental cells were more resistant to suspension-induced cell death. Ras effects on anchorage-independent cell death were reproduced by a mutant protein that signals selectively to Raf-1, but not by mutant Ras that preferentially binds to RalGDS. Expression of a Ras mutant that selectively activates PI3K resulted in substantial protection from detachment-induced cell death. MAPK activity was increased in adherent Ras12V- and Ras12V35S-expressing cells, but abolished upon detachment. Interestingly, impaired MAPK activity was sufficient to stimulate apoptosis in adherent Ras-transformed cells, but not in parental cells. Treatment with a PI3K inhibitor also stimulated apoptosis selectively in Ras-transformed cells. These results demonstrate that constitutive expression of activated Ras elicits differential effects on the survival of thyroid cells. Moreover, Ras expression results in a greater dependence of thyroid cells on MAPK and PI3K activity for their survival.


Assuntos
Apoptose/genética , Transformação Celular Neoplásica/genética , Genes ras , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Glândula Tireoide/citologia , Substituição de Aminoácidos , Animais , Adesão Celular , Técnicas de Cultura de Células/métodos , Linhagem Celular Transformada/citologia , Meios de Cultura Livres de Soro , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Humanos , Sistema de Sinalização das MAP Quinases , Mutagênese Sítio-Dirigida , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Mutação Puntual , Proteínas Proto-Oncogênicas c-raf/fisiologia , Ratos , Ratos Wistar , Suspensões , Fator ral de Troca do Nucleotídeo Guanina/fisiologia
11.
Oncogene ; 19(7): 924-32, 2000 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-10702801

RESUMO

Hormones are specialized mitogens that stimulate proliferation in their differentiated target cells. Thyrotropin (TSH), the physiologic regulator of thyroid cells, stimulates cAMP-mediated proliferation and thyroid-specific gene expression. The mitogenic effects of TSH require Ras, therefore Ras activation should be compatible with the maintenance of thyroid differentiation. However, expression of activated Ras extinguishes the differentiated phenotype of thyroid cells. One explanation for this apparent paradox is the selective utilization of Ras effector pathways. We tested the hypothesis that Ras signaling through PI3K mediates the mitogenic effects of TSH in cells which retain their differentiated character. Expression of a Ras effector mutant (RasV12S35) that signals preferentially through Raf-1, although sufficient to confer TSH-independent proliferation, abolished hormone-regulated expression of thyroglobulin and the sodium/iodide symporter. In contrast, expression of a Ras mutant (RasV12C40) that binds selectively to PI3K conferred TSH-independent proliferation without marked effects on thyroid-specific gene expression. Unlike the inhibitory effects of TSH on the proliferation of RasV12S35-expressing cells, TSH enhanced RasV12C40-stimulated proliferation by further increasing the activity of p70s6k, an important mediator of the mitogenic effects of TSH and RasV12C40. These results demonstrate that channeling Ras-dependent signals to PI3K confers TSH with the ability to stimulate proliferation in differentiated cells. Oncogene (2000) 19, 924 - 932.


Assuntos
Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/fisiologia , Tireotropina/fisiologia , Proteínas ras/fisiologia , Animais , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , DNA/biossíntese , Mitógenos/metabolismo , Ratos , Ratos Wistar , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo
12.
Oncogene ; 19(32): 3609-15, 2000 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-10951566

RESUMO

In addition to protein kinase A (PKA), cAMP regulates the activity of cAMP-gated channels and Rap1-specific guanine nucleotide exchange factors. We tested the hypothesis that the targets of cAMP might also include regulators of the Ras protooncogene. In rat thyroid cells, thyrotropin (TSH) stimulates proliferation through a cAMP-mediated pathway that requires Ras activity. Interference with Ras impairs DNA synthesis stimulated by TSH as well as cAMP elevating agents and analogs, demonstrating that the requirement for Ras lies down-stream of cAMP. Although cAMP stimulates proliferation, microinjection of the purified PKA catalytic subunit failed to do so, suggesting that factors in addition to PKA are required for cAMP-stimulated cell cycle progression. When added to thyroid cells expressing human Ha-Ras, TSH rapidly and markedly increased the proportion of GTP-bound Ras. Ras activity was increased within 1 min of TSH addition, maximal at 5-15 min, and declined to basal levels 30-60 min after hormone treatment. Cyclic AMP elevating agents elicited similar effects on Ras, indicating that TSH activates Ras through a cAMP-mediated pathway. Although cAMP-mediated, Ras activation by TSH and cAMP was independent of PKA activity. Moreover, cAMP-stimulated Ras activation was not impaired by tyrosine kinase inhibitors. These results indicate that cAMP activates targets in addition to PKA in thyroid cells, and that these targets may include regulators of Ras. The ability of cAMP elevating agents to activate Ras in addition to PKA may explain the inability of the PKA catalytic subunit to stimulate DNA synthesis in thyroid cells.


Assuntos
AMP Cíclico/metabolismo , Proteínas ras/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Linhagem Celular , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , DNA/biossíntese , Humanos , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Wistar , Glândula Tireoide/citologia , Tireotropina/farmacologia , Proteínas ras/genética
13.
Oncogene ; 13(6): 1305-14, 1996 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-8808705

RESUMO

Raf-1 is a serine/threonine specific kinase that integrates signaling by a large number of mitogens to elicit a transcriptional response in the nucleus. Activated Raf-1 phosphorylates and activates MAPK/ERK kinase Mek), thus initiating the Mek--> MAP kinase cascade, which ultimately results in the phosphorylation and activation of transcription factors by MAP kinase. Here we have characterized the mechanism by which monoclonal antibody URP26K, which binds to an epitope in the Raf-1 kinase domain, inhibits intracellular signal transduction. This antibody preferentially immunoprecipitated the underphosphorylated, non-activated form of Raf-1 from quiescent cells. Baculovirus-expressed Raf-1 immunoprecipitated with URP26K was largely refractory to phosphorylation and activation mediated by protein kinase C (PKC)alpha or the tyrosine kinase Lck. In addition, URP26K reduced the binding of Raf-1 to its substrate Mek in vitro, but did not disturb the association of Raf-1 with Ras. Microinjection of URP26K into Rat-1 cells blocked DNA synthesis initiated by serum, insulin and various purified growth factors, but it did not block DNA synthesis initiated by v-ras. Microinjected URP26K also impaired the expression of stably transfected beta-galactosidase reporter genes regulated by minimal promoter elements. These results demonstrate, (i) that the URP26K monoclonal antibody inhibits Raf-1 by preventing activating Raf-1 phosphorylation and/or association with its substrate Mek, (ii) that inhibition of Raf-1 by URP26K does not interfere with Ras-induced DNA synthesis. In contrast to dominant negative Raf-1 mutants, which also block Ras signaling by binding to the Ras effector domain, antibody mediated Raf-1 inhibition thus reveals a branchpoint of mitogenic signaling at the level of Ras.


Assuntos
Anticorpos Monoclonais/farmacologia , MAP Quinase Quinase Quinase 1 , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Transdução de Sinais/fisiologia , Células 3T3/efeitos dos fármacos , Células 3T3/metabolismo , Células 3T3/fisiologia , Animais , Anticorpos Monoclonais/metabolismo , Sequência Conservada , DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática/efeitos dos fármacos , Epitopos/metabolismo , Substâncias de Crescimento/farmacologia , Camundongos , Microinjeções , Fosforilação , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-raf , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/metabolismo , Proteínas ras/fisiologia
14.
Endocrinology ; 139(4): 1991-8, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9528986

RESUMO

cAMP exerts differential effects on mitogenic signaling pathways. In many cells, cAMP inhibits growth factor-stimulated MAPK activity and proliferation. In others, cAMP promotes growth. TSH stimulates proliferation through elevations in cAMP in thyroid follicular cells. This mitogenic pathway is dependent upon both protein kinase A and Ras, but not upon Raf-1, mitogen-activated protein kinase kinase, or mitogen-activated protein kinase. We report that TSH, acting through cAMP, activates pp70s6k and that this activity is required for TSH-stimulated DNA synthesis. A similar role for pp70s6k in cAMP-mediated mitogenesis was observed in secondary rat Schwann cells and in Swiss3T3 fibroblasts, two additional cell types that respond to cAMP with growth. In contrast, cAMP elevation did not activate pp70s6k in NIH3T3 or REF52 fibroblasts, cells in which cAMP fails to stimulate proliferation. Together, these results suggest that pp70s6k plays an important and general role in cAMP-mediated proliferation.


Assuntos
AMP Cíclico/farmacologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Tireotropina/farmacologia , Células 3T3/citologia , Células 3T3/efeitos dos fármacos , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , DNA/biossíntese , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Camundongos , Polienos/farmacologia , Ratos , Ratos Wistar , Proteínas Quinases S6 Ribossômicas/antagonistas & inibidores , Células de Schwann/citologia , Células de Schwann/efeitos dos fármacos , Sirolimo , Glândula Tireoide/citologia , Glândula Tireoide/efeitos dos fármacos
15.
Endocrinology ; 135(3): 1212-9, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8070365

RESUMO

The rapid and transient induction of immediate early gene expression accompanies growth factor stimulation. TSH and insulin-like growth factor I (IGF-I) are important regulators of the thyroid follicular cell and stimulate both proliferation and differentiation. The signaling pathways induced by TSH and IGF-I are at least partially distinct. TSH uses cAMP as a second messenger, whereas the IGF-I receptor possesses protein tyrosine kinase activity. Although both agents stimulate DNA synthesis and proliferation in Wistar rat thyroid cells, they induce dramatically different patterns of immediate early gene expression. IGF-I stimulates the expression of c-fos, c-jun, junB, and egr1. In contrast, TSH stimulates c-fos and junB but not egr1 expression. TSH inhibits basal levels of c-jun expression in quiescent cells and represses serum and IGF-I-stimulated c-jun, c-fos, and egr1 expression. Consistent with these results, TSH represses serum- and phorbol ester-stimulated AP-1 activity. Although TSH and IGF-I individually stimulate DNA synthesis in thyroid cells, they exert opposing effects on the expression of some immediate early genes, including c-jun and egr1.


Assuntos
Expressão Gênica/efeitos dos fármacos , Genes Precoces , Proteínas Imediatamente Precoces , Fator de Crescimento Insulin-Like I/farmacologia , Glândula Tireoide/efeitos dos fármacos , Tireotropina/farmacologia , Animais , Sequência de Bases , Bovinos/sangue , Bovinos/embriologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína 1 de Resposta de Crescimento Precoce , Sangue Fetal , Sondas Moleculares/genética , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Acetato de Tetradecanoilforbol/farmacologia , Glândula Tireoide/citologia , Glândula Tireoide/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Endocrinology ; 137(1): 96-104, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8536648

RESUMO

We previously reported that microinjection of purified Ras protein stimulated DNA synthesis in quiescent Wistar rat thyrocytes and that TSH (TSH)-stimulated DNA synthesis was Ras-dependent. In contrast to these results, microinjection of cellular or oncogenic Ras significantly reduced TSH-stimulated thyroglobulin (Tg) expression, a marker of thyrocyte differentiation. Microinjection of a dominant inhibitory Ras mutant had no effect on TSH-stimulated Tg expression. As the Tg promoter is cAMP-responsive and Ras was previously reported to interfere with entry of catalytic (C) subunit of the cAMP-dependent protein kinase into the nucleus, experiments were performed to assess the effects of Ras on cAMP-mediated signaling. Microinjection of either cellular or oncogenic Ras had no effect on TSH-stimulated entry of C subunit into the nucleus. Consistent with these data, Ras did not reduce TSH-stimulated cAMP response element binding protein phosphorylation, or cAMP response element-regulated gene expression. These results demonstrate that Ras exerts differential effects on TSH signaling; Ras increases TSH-stimulated DNA synthesis and decreases TSH-induced Tg expression. Moreover, the mechanism through which Ras induces Tg expression lies distal to entry of C subunit into the nucleus, cAMP response element binding protein phosphorylation, and cAMP response element-regulated gene expression.


Assuntos
AMP Cíclico/fisiologia , Transdução de Sinais/efeitos dos fármacos , Tireoglobulina/antagonistas & inibidores , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Proteínas ras/farmacologia , Animais , Catálise , Bovinos , Linhagem Celular , Núcleo Celular/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Microinjeções , Ratos , Ratos Wistar , Tireoglobulina/metabolismo , Glândula Tireoide/citologia , Tireotropina/farmacologia , Fatores de Tempo
17.
Endocrinology ; 141(2): 606-14, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10650941

RESUMO

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.


Assuntos
AMP Cíclico/metabolismo , Regulação da Expressão Gênica/fisiologia , Genes MHC Classe I , Interferon gama/farmacologia , Glândula Tireoide/fisiologia , Tireotropina/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Células Cultivadas , Colforsina/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Interleucina-1/farmacologia , Cinética , Ratos , Ratos Wistar , Proteínas Recombinantes , Proteínas Quinases S6 Ribossômicas/metabolismo , Glândula Tireoide/citologia , Glândula Tireoide/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo
18.
Endocrinology ; 141(1): 146-52, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10614633

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA , Proteína Quinase C/fisiologia , Glândula Tireoide/citologia , Tireotropina/fisiologia , Animais , Western Blotting , Divisão Celular , Células Cultivadas , DNA/biossíntese , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Vetores Genéticos/genética , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase C/biossíntese , Proteína Quinase C/genética , Proteínas Proto-Oncogênicas/genética , Ratos , Ratos Wistar , Fatores de Transcrição/genética , Proteínas Elk-1 do Domínio ets
20.
Proc Natl Acad Sci U S A ; 75(9): 4252-6, 1978 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-212741

RESUMO

Peking duck cell nuclear DNA has no complementarity to RNA of Prague C Rous sarcoma virus (RSV). Upon infection of Peking duck cells by Prague C RSV, polynucleotide sequences complementary to Peking duck cell nuclear DNA can be detected in the high molecular weight RNA from progeny Peking duck cell passaged RSV (RSVPD) by RNA.DNA molecular hybridazation. When 3T3 cells are subsequently infected by RSVPD, polynucleotide sequences complementary to Peking duck cell nuclear DNA can be detected in the 3T3 cell nuclear DNA by RNA.DNA molecular hybridization. The potential consequences of the transfer of the Peking duck cell nuclear DNA from the avian to the murine cells are discussed.


Assuntos
Vírus do Sarcoma Aviário/genética , DNA Recombinante , Transdução Genética , Animais , Sequência de Bases , Linhagem Celular , Patos , Hibridização de Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA