Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
2.
J Cell Biol ; 116(1): 187-96, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1730743

RESUMO

Transforming growth factor-beta 1 (TGF-beta 1) has previously been implicated as a potential negative autocrine or paracrine growth regulator of certain cell types (Arteaga, C. L., R. J. Coffey, Jr., T. C. Dugger, C. M. McCutchen, H. L. Moses, and R. M. Lyons. 1990. Cell Growth & Differ. 1:367-374; Hafez, M. M., D. Infante, S. Winawer, and E. Friedman. 1990. Cell Growth & Differ. 1:617-626; Glick, A. B., K. C. Flanders, D. Danielpour, S. H. Yuspa, and M. B. Sporn. 1989. Cell Regulation. 1:87-97). This is based mainly on experiments assessing the effects of exogenous TGF-beta 1 or neutralizing antibodies to TGF-beta 1 on normal or tumor cell proliferation in vitro. However, direct evidence demonstrating such a negative regulation of tumor cell growth in vivo is still lacking. To overcome this problem we have constructed and used an antisense expression vector for TGF-beta 1 as a means of regulating endogenous TGF-beta 1 expression in tumor cells. Antisense-transfected FET human colon carcinoma cells showed a fivefold reduction in TGF-beta 1 mRNA and 15-fold reduction in TGF-beta 1 secretion. Antisense mRNA was detected in transfected cells by an RNase protection assay. Compared to control cells, cultured antisense-transfected cells showed a reduction in lag phase time rather than a change in doubling time. Cloning efficiencies of transfected cells were four times greater than control cells in anchorage-independent assays. Control cells did not form tumors at 5 x 10(5) in athymic nude mice. Antisense-transfected cells formed tumors in 40% of animals injected. At higher inocula (1 x 10(6) cells) antisense-transfected cells formed tumors in 100% of animals injected, but control cells still failed to form tumors. These results show that TGF-beta 1 acts as a negative growth regulator of human colon carcinoma cells in vivo as well as in vitro. Acquisition of partial or full resistance to such inhibitory effects may therefore contribute to tumor development and progression.


Assuntos
Divisão Celular , Neoplasias do Colo/patologia , RNA Antissenso/genética , RNA Mensageiro/genética , Fator de Crescimento Transformador beta/fisiologia , Animais , Anticorpos , Linhagem Celular , Clonagem Molecular , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Plasmídeos , Transfecção , Fator de Crescimento Transformador beta/genética , Transplante Heterólogo
3.
Cytokine Growth Factor Rev ; 11(1-2): 23-35, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10708950

RESUMO

Normal signaling by TGFbeta, in the absence of serum or exogenous factors, involves a rapid activation of Ras, Erks, and Sapks in proliferating cultures of TGFbeta-sensitive untransformed epithelial cells and human carcinoma cells. Expression of either RasN17 or dominant-negative (DN) MKK4, or addition of the MEK1 inhibitor PD98059, can block the ability of TGFbeta to induce AP-1 complex formation at the TGFbeta(1) promoter and to autoinduce its own production. The primary components present in this TGFbeta-stimulated AP-1 complex are JunD and Fra-2, although c-Jun, and possibly Fos B, may also be present. While there are two potential Smad binding elements (SBE's) in the TGFbeta(1) promoter, supershift assays suggest that at least one of these does not bind Smad4, and the other is unable to bind factors activated by TGFbeta. In contrast, TGFbeta autoinduction is Smad3-dependent, as DN Smad3 inhibits the ability of TGFbeta to stimulate TGFbeta(1) promoter activity. Our results indicate that TGFbeta can activate both the MKK4/Sapk and MEK/Erk pathways, through Ras and TGFbeta R(I) and R(II), to induce TGFbeta(1) production; Smad4 does not appear to be involved, and Smad3 appears to function independently of this Smad4. We also demonstrate that activation of the Ras/Mapk pathway by TGFbeta positively modulates Smad1-signaling-pathway activation by TGFbeta. In addition, Smad1 could enhance TGFbeta activation of the SBE reporter SBE-luc and this effect could be blocked by co-expression of a DN TGFbeta R(I) receptor or by the MEK1 inhibitor PD98059. This cross-talk between the MEK/Erk and Smad1 pathways was mediated through the four Erk consensus phosphorylation sites in the linker region of Smad1. Mutation of these sites resulted in a loss of the ligand-dependence of both Smad1-Smad4 interactions and nuclear accumulation of Smad1, as well as a loss of the ability of Smad1 to enhance TGFbeta-mediated SBE activation. Our results provide evidence that Erk-mediated phosphorylation of Smad1 in response to TGFbeta is critical for regulating Smad1 subcellular localization; this may be a key determinant in maintaining TGFbeta-dependent transcriptional activation.


Assuntos
MAP Quinase Quinase 4 , MAP Quinase Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases , Quinases de Proteína Quinase Ativadas por Mitógeno , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Proteínas ras/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Smad3 , Proteína Smad4 , Transativadores/metabolismo , Fator de Transcrição AP-1/metabolismo
4.
Cancer Res ; 51(9): 2256-62, 1991 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-2015589

RESUMO

A previous report from this laboratory indicated that a transformed fibroblast cell line up-regulated c-myc by as much as 14-fold as cultures approached saturating densities, whereas the untransformed counterparts displayed little alteration in c-myc expression (Cancer Res., 49: 2320, 1989). The results suggested a mechanism for the growth advantage of the transformed cells at postconfluent densities. Similarly, the present results indicate that regulation of c-myc expression during establishment of a quiescent state markedly differed in poorly differentiated versus well-differentiated human colon carcinoma cells. While c-myc expression increased 2- to 3-fold during this period in the poorly differentiated cells, expression levels for this protooncogene showed little variation in the well-differentiated cells. There was, however, no correlation between degree of differentiation and c-myc mRNA levels in growing cultures (i.e., cells in late log phase). Another proliferation-associated mRNA, transforming growth factor alpha (TGF-alpha), was also differentially regulated in the two groups of colon carcinoma cells as cultures approached quiescence. Further, addition of exogenous growth-stimulatory factors (epidermal growth factor plus insulin plus transferrin) to quiescent, well-differentiated cells resulted in an up-regulation of TGF-alpha mRNA levels by 9-fold over a 24-h period. In contrast, poorly differentiated cells displayed little alteration in TGF-alpha mRNA levels under similar conditions. The results suggest that inappropriate kinetic regulation of c-myc and TGF-alpha mRNAs at quiescence may be related to the growth factor independence of the poorly differentiated colon carcinoma cells. Furthermore, altered temporal regulation of c-myc and TGF-alpha expression appears to be more relevant to differentiation status in human colon carcinoma cells than are absolute expression levels.


Assuntos
Carcinoma/genética , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica/genética , Genes myc , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador alfa/genética , Carcinoma/patologia , Neoplasias do Colo/patologia , Humanos , Fatores de Tempo , Células Tumorais Cultivadas , Regulação para Cima
5.
Cancer Res ; 57(4): 628-33, 1997 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9044838

RESUMO

Although transforming growth factor beta (TGF-beta) is known to be a potent growth inhibitor of breast cancer cells (BCCs), the signaling mechanisms mediating TGF-beta responses have not been defined. We have demonstrated previously that TGF-beta can activate Ras and extracellular signal-regulated kinase (ERK) 1 in untransformed epithelial cells (K. M. Mulder and S. L. Morris, J. Biol. Chem., 267: 5029-5031, 1992; M. T. Hartsough and K. M. Mulder, J. Biol. Chem., 270: 7117-7124, 1995). We have also shown that TGF-beta signaling is altered in epithelial cells when Ras activation is blocked (Hartsough et at., J. Biol. Chem., 271: 22368-22375). Here we demonstrate the ability of the TGF-beta3 isoform to activate the signaling component ERK2 in TGF-beta-sensitive BCCs but not in TGF-beta-resistant cells. The ERK2 isoform was activated by 6-fold within 10 min of TGF-beta3 addition to the TGF-beta-sensitive BCC line Hs578T. Moreover, the IC50 for inhibition of DNA synthesis by TGF-beta3 in this cell line correlated with the EC50 for TGF-beta3 activation of ERK2. In contrast, TGF-beta3 had little effect on either DNA synthesis or ERK2 activation in ZR-75 BCCs lacking the type-II TGF-beta receptors (R(II)), or in ZR-75 BCCs stably transfected with R(II) yet still TGF-beta resistant. In addition, our data demonstrate that TGF-beta3 affected a sustained activation of the stress-activated protein kinase/Jun N-terminal kinase (SAPK/JNK) type of mitogen-activated protein kinase (MAPK); maximal induction levels were 2.5-fold above basal values and were attained at 30 min after TGF-beta3 treatment. In contrast, TGF-beta3 did not increase SAPK/JNK activity in the TGF-beta-resistant ZR-75 R(II) BCCs. Our data provide the first evidence that TGF-beta activation of ERK2 and SAPK/JNK is associated with negative growth control of BCCs. This is also the first demonstration that TGF-beta can activate the SAPK/JNK type of MAPK and that the TGF-beta3 isoform can regulate MAPK activity.


Assuntos
Neoplasias da Mama/enzimologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , DNA de Neoplasias/biossíntese , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno , Proteínas Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteína Quinase 1 Ativada por Mitógeno , Fosforilação
6.
Cancer Res ; 49(9): 2320-6, 1989 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-2706619

RESUMO

The present report describes a transformed cell line (AKR-MCA) in which the c-myc proto-oncogene is up-regulated by as much as 14-fold as cultures approach stationary phase growth. The untransformed counterpart AKR-2B cells did not exhibit such an increase in c-myc expression at high cell densities, nor did chemically transformed derivatives of another murine fibroblast cell line (C3H 10T1/2). N,N-Dimethylformamide and retinoic acid reduced c-myc levels in confluent AKR-MCA cells in association with a loss of transformed morphology, a reduction in saturation density, and the formation of a contact-inhibited monolayer at confluency. These findings suggest that the high levels of c-myc in confluent AKR-MCA cells may interfere with the normal signals involved in density-dependent growth regulation in this cell system. The effects of N,N-dimethylformamide and retinoic acid were reversible and dose-related. The half-time for the early, rapid decline in c-myc mRNA was approximately 26 min in response to N,N-dimethylformamide and 38 min in response to retinoic acid, effects which preceded the alterations in morphology and saturation densities. Activation of the latent transforming growth factor-beta in serum-free medium conditioned by confluent AKR-MCA cells, followed by its addition to preconfluent AKR-MCA cells, resulted in an up-regulation of c-myc mRNA. However, addition of serum-containing conditioned medium under similar conditions did not require prior acidification to up-regulate c-myc. Thus, active transforming growth factor-beta may be present in conditioned medium from confluent AKR-MCA cells grown in serum-containing medium, or autocrine factors other than TGF-beta may produce the confluency-associated up-regulation of c-myc and the altered density-dependent growth regulation in AKR-MCA cells.


Assuntos
Proto-Oncogenes , Divisão Celular , Linhagem Celular Transformada , Dimetilformamida/farmacologia , Relação Dose-Resposta a Droga , RNA Mensageiro/análise , Fatores de Tempo , Fatores de Crescimento Transformadores/farmacologia , Tretinoína/farmacologia
7.
Cancer Res ; 58(20): 4752-7, 1998 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9788633

RESUMO

Previous results have suggested that Smad1 transduces signals in response to bone morphogenetic proteins (BMPs), but not in response to transforming growth factor beta (TGF-beta). Here we investigated the ability of TGF-beta to regulate Smad1 phosphorylation, hetero-oligomerization with Smad4, translocation to the nucleus, and transcriptional activation of 3TP-luciferase reporter activity in TGF-beta- and BMP-responsive Hs578T human breast cancer cells. We demonstrate that Smad1 was rapidly phosphorylated in vivo in response to both TGF-beta3 and BMP2 as determined using an antibody against the epitope-tagged Smad1 being expressed. In addition, both TGF-beta3 and BMP2 increased Smad1-Smad4 hetero-oligomerization in Hs578T cells. Visualization of Smad1 nuclear translocation with the aid of green fluorescent protein (GFP) in live cells demonstrated nuclear accumulation of GFP-Smad1 fluorescence in response to either TGF-beta or BMP2 stimulation. After ligand stimulation, approximately 60-70% of transfected cells displayed prominent nuclear fluorescence. Expression of Smad1 in Hs578T cells increased the activity of the TGF-beta-responsive reporter 3TP-Lux. Moreover, TGF-beta treatment further potentiated the effect of Smad1 on 3TP-luciferase activity. Collectively, our results demonstrate that TGF-beta as well as BMP can signal through Smad1.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Ligação a DNA/fisiologia , Transativadores/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Proteínas Morfogenéticas Ósseas/farmacologia , DNA/biossíntese , Feminino , Humanos , Luciferases/metabolismo , Fosforilação , Coelhos , Proteínas Smad , Proteína Smad1 , Proteína Smad4
8.
Cancer Res ; 55(10): 2056-62, 1995 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-7743502

RESUMO

Here we describe human colon carcinoma cell clones, isolated from a transforming growth factor beta (TGF-beta)-responsive parental cell line, which display differential sensitivities to TGF-beta 1 and TGF-beta 2 isoforms. In a monolayer proliferation assay, some clones were sensitive to both isoforms (IC50 = 0.1-0.6 ng/ml; S1S2) while others were resistant to both isoforms (IC50 > 5 ng/ml; R1R2). Still other clones (R1S2) were sensitive to TGF-beta 2 (IC50 = 0.1-0.2 ng/ml), but were resistant to TGF-beta 1 (IC50 > or = 5 ng/ml). In S1S2 cells, both TGF-beta isoforms resulted in a repression of c-myc mRNA expression, a concentration-dependent increase in fibronectin levels, and an enhanced production of the colon cell differentiation marker carcinoembryonic antigen. In contrast, R1R2 cells did not display these responses, or did so only to a limited extent. In R1S2 cells, TGF-beta 2 elicited these responses, yet TGF-beta 1 was essentially without effect. Receptor cross-linking experiments indicated that TGF-beta resistance in this model system was not generally associated with a complete lack of expression of either type I or II receptors. Moreover, the R1S2 type clones were heterogeneous, although the majority of them displayed binding to type I receptors by TGF-beta 2 but not by TGF-beta 1. These data suggest that either the TGF-beta 1 and TGF-beta 2 isoforms differ with respect to their ability to interact with the type I and II classes of receptors, or the TGF-beta 1 and TGF-beta 2 isoforms can interact with distinct receptor proteins of the type I and II classes in this model system.


Assuntos
Carcinoma/metabolismo , Neoplasias do Colo/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Antígeno Carcinoembrionário/metabolismo , Carcinoma/genética , Carcinoma/patologia , Diferenciação Celular , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Resistência a Medicamentos , Fibronectinas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes myc/efeitos dos fármacos , Humanos , Proteínas Proto-Oncogênicas c-myc/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Células Tumorais Cultivadas
9.
Cancer Res ; 50(23): 7581-6, 1990 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-1701348

RESUMO

Previously, we reported that exponentially proliferating cultures of well-differentiated human colon carcinoma cells responded to transforming growth factor beta 1 (TGF-beta) with growth inhibition, alterations in morphology, and increased secretion of the differentiation marker, carcinoembryonic antigen. Poorly differentiated cultures were unresponsive. Here we show that TGF-beta was ineffective in repressing nutrient-stimulated mitogenesis in quiescent, poorly differentiated cells. However, in quiescent, well-differentiated cells, TGF-beta repressed the mitogenic responses to both nutrients alone (by 90%) and to nutrients plus the exogenous stimulatory factors epidermal growth factor (E), insulin (I), and transferrin (T) (by 55-65%). Thymidine incorporation experiments indicated that TGF-beta reduced both the onset and peak mitogenic response to growth factors and/or nutrients in the well-differentiated cells. Additionally, TGF-beta repressed the growth factor (E + I + T)-stimulated upregulation of expression of both c-myc and of transforming growth factor alpha (TGF-alpha) mRNAs in quiescent, well-differentiated cells. TGF-beta also elicited a rapid (t1/2 approximately 1h) down-regulation of c-myc expression in the absence of prior growth factor (E + I + T) stimulation. In contrast, TGF-beta had no effect on c-myc or TGF-alpha mRNA expression in the poorly differentiated cells. The results suggest that TGF-beta exerts rapid inhibitory effects on proliferation-associated genes in quiescent and restimulated, well-differentiated cells. Expression of these genes (c-myc and TGF-alpha) may otherwise (in the absence of TGF-beta) play roles in the cellular signaling of mitogenic responses by growth stimulatory factors in well-differentiated colon carcinoma cells.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Mitose/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Fator de Crescimento Transformador alfa/biossíntese , Fator de Crescimento Transformador beta/farmacologia , Northern Blotting , Linhagem Celular , Regulação para Baixo , Fator de Crescimento Epidérmico/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Insulina/farmacologia , RNA/isolamento & purificação , Transferrina/farmacologia
10.
Cancer Res ; 48(24 Pt 1): 7120-5, 1988 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-3191488

RESUMO

Previous work indicated that transforming growth factor-beta (TGF-beta) elicits proliferation-inhibitory effects in the human colon carcinoma cell line MOSER. This paper describes the isolation and characterization of spontaneously arising subclones from this TGF-beta-sensitive parental line which were relatively refractory to the inhibitory effects of TGF-beta. While the parental cell line responded to TGF-beta with an inhibition of cellular proliferation in monolayer culture and in soft agarose, an increase in extracellular fibronectin, and a down-regulation of c-myc protooncogene expression, these responses were absent or attenuated in the sublines. However, the resistant clones retained the ability to specifically bind TGF-beta. N,N-Dimethylformamide and retinoic acid, two other agents associated with induction of a partial differentiation-like response in the MOSER parental cells (similar to that elicited by TGF-beta), inhibited the monolayer proliferation of both the parental cells and the TGF-beta-resistant sublines. Thus, the refractoriness observed in the isolated clones was relatively specific for TGF-beta.


Assuntos
Neoplasias do Colo/patologia , Fatores de Crescimento Transformadores/farmacologia , Linhagem Celular , Células Clonais/efeitos dos fármacos , Dimetilformamida/farmacologia , Resistência a Medicamentos , Humanos , Oncogenes , Tretinoína/farmacologia
11.
Oncogene ; 17(1): 47-55, 1998 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-9671313

RESUMO

Our previous data demonstrated that Ras activation is necessary and sufficient for transforming growth factor beta (TGFbeta)-mediated Erk1 activation, and is partially required for the inhibition of cyclin-dependent kinase 2 (Cdk2) activity, cyclin A expression and DNA synthesis by TGFbeta (KM Mulder and SL Morris, J. Biol. Chem., 267: 5029-5031, 1992; MT Hartsough and KM Mulder, J. Biol. Chem., 270: 7117-7124, 1995; and MT Hartsough et al., J. Biol. Chem., 271: 22368-22375, 1996). Here, we examined the kinetics and role of Ras in TGFbeta3-mediated effects on specific G1 cell cycle components in TGFbeta-sensitive (4-1) and TGFbeta-resistant (4-6) intestinal epithelial cells (IEC's). Our results indicate that inactivation of Ras by stable, inducible expression of a dominant-negative mutant of Ras (RasN17) completely abrogated the ability of TGFbeta3 to up-regulate both CKI's. In contrast, the ability of TGFbeta3 to up-regulate p27Kip1 and p21Cip1 was maintained in ZnCl2-treated control cells. Inactivation of Ras also completely blocked the rapid TGFbeta-mediated increase in new synthesis of p27Kip1 protein. Moreover, up-regulation of p21Cip1 protein levels and new synthesis of p27Kip1, as well as the association of these CKI's with Cdk2, preceded the decrease in Cdk2 activity by TGFbeta. Collectively, our results suggest that p21Cip1 and p27Kip1 are upstream effectors of the TGFbeta-mediated inhibition of Cdk2 activity in IEC 4-1 cells, and demonstrate that Ras activation is obligatory for TGFbeta-mediated up-regulation of these CKIs in untransformed epithelial cells.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Ciclinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Supressoras de Tumor , Regulação para Cima , Proteínas ras/fisiologia , Linhagem Celular , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , DNA/biossíntese , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Cinética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteínas ras/genética
12.
Oncogene ; 18(11): 2033-7, 1999 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-10208426

RESUMO

Our previous data demonstrated that Ras activation was necessary and sufficient for transforming growth factor-beta (TGFbeta)-mediated Erk1 activation, and was required for TGFbeta up-regulation of the Cdk inhibitors (CKI's) p27(Kip1) and p21(Cip1) (KM Mulder and SL Morris, J. Biol. Chem., 267, 5029-5031, 1992; MT Hartsough and KM Mulder, J. Biol. Chem., 270, 7117-7124, 1995; MT Hartsough et al., J. Biol. Chem., 271, 22368-22375, 1996 and J Yue et al., Oncogene, 17, 47-55, 1998). Here we examined the role of Ras in TGFbeta-mediated effects on a rat homolog of Smad1 (termed RSmad1). We demonstrate that both TGFbeta and bone morphogenetic protein (BMP) can induce endogenous Smad1 phosphorylation in intestinal epithelial cells (IECs). The combination of transient expression of RSmad1 and TGFbeta treatment had an additive effect on induction of the TGFbeta-responsive reporter 3TP-lux. Either inactivation of Ras by stable, inducible expression of a dominant-negative mutant of Ras (RasN17) or addition of MAP and ERK kinase (MEK) inhibitor PD98059 to cells significantly decreased the ability of both TGFbeta and BMP to induce phosphorylation of endogenous Smad1 in IECs. Moreover, either inactivation of Ras or addition of PD98059 to IEC 4-1 cells inhibited the ability of RSmad1 to regulate 3TP luciferase activity in both the presence and absence of TGFbeta. Collectively, our data indicate that TGFbeta can regulate RSmad1 function in epithelial cells, and that the Ras/MEK pathway is partially required for TGFbeta-mediated regulation of RSmad1.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Regulação da Expressão Gênica , Genes Reporter , Luciferases/genética , MAP Quinase Quinase 1 , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Ratos , Proteínas Smad , Proteína Smad1
13.
Pharmacol Ther ; 91(1): 1-34, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11707292

RESUMO

Transforming growth factor (TGF)-beta is a natural and potent growth inhibitor of a variety of cell types, including epithelial, endothelial, and hematopoietic cells. The ability of TGF-beta to potently inhibit the growth of many solid tumors of epithelial origin, including breast and colon carcinomas, is of particular interest. However, many solid tumor cells become refractory to the growth inhibitory effects of TGF-beta due to defects in TGF-beta signaling pathways. In addition, TGF-beta may stimulate the invasiveness of tumor cells via the paracrine effects of TGF-beta. Accordingly, in order to develop more effective anticancer therapeutics, it is necessary to determine the TGF-beta signal transduction pathways underlying the growth inhibitory effects and other cellular effects of TGF-beta in normal epithelial cells. Thus far, two primary signaling cascades downstream of the TGF-beta receptors have been elucidated, the Sma and mothers against decapentaplegic homologues and the Ras/mitogen-activated protein kinase pathways. The major objective of this review is to summarize TGF-beta signaling in epithelial cells, focusing on recent advances involving the Sma and mothers against decapentaplegic homologues and Ras/mitogen-activated protein kinase pathways. This review is particularly timely in that it provides a comprehensive summary of both signal transduction mechanisms and the cell cycle effects of TGF-beta.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas Repressoras , Transdução de Sinais/fisiologia , Fatores de Transcrição , Fator de Crescimento Transformador beta/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Proteínas de Ligação a DNA/química , Células Epiteliais , Proteínas de Helminto/química , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/fisiologia
14.
Pharmacol Ther ; 75(1): 21-41, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9364579

RESUMO

Transforming growth factor (TGF)-beta is a potent growth suppressor of epithelial cells. Resistance to TGF-beta, however, occurs frequently in solid tumors of epithelial origin and contributes to the uncontrolled growth of these tumors. Although mutant receptor proteins contribute to TGF-beta insensitivity, deregulation of TGF-beta signaling cascades represents an equally important mechanism underlying TGF-beta resistance. Identification of abnormal regulation of signaling components in tumor epithelial cells will lead to the development of selective therapeutic approaches to repair the relevant signaling cascade(s) and reverse the growth anomaly. Within the past few years, great strides have been made in defining signaling pathways for TGF-beta. For example, our laboratory has demonstrated a direct correlation between TGF-beta-mediated growth inhibition of epithelial cells and activation of Ras and three members of the mitogen-activated protein kinase (MAPK) superfamily. The TGF-beta signaling events were sustained, dose-dependent, and absent in TGF-beta-resistant cells. Further, up-regulation of both p27Kip1 and p21Cip1, nuclear events important for the growth inhibitory effect of TGF-beta, are completely dependent upon the activation of Ras. However, Ras-independent pathways are also activated simultaneously with the Ras/MAPK pathways to mediate the final TGF-beta growth inhibitory outcome. One such pathway includes the SMAD signaling components that control TGF-beta-mediated gene transcription, currently under active study by a number of laboratories, including our own. Future efforts in this field will focus on defining the significance of these signaling proteins and pathways in mediating specific TGF-beta responses. Moreover, additional novel signaling proteins are sure to be identified.


Assuntos
Proteínas de Drosophila , Células Epiteliais/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Alquil e Aril Transferases/metabolismo , Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/enzimologia , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas de Insetos/metabolismo , Neoplasias/metabolismo , Sinais Direcionadores de Proteínas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas de Ligação a Tacrolimo , Fator de Crescimento Transformador beta/fisiologia
15.
Mol Endocrinol ; 3(8): 1215-22, 1989 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2674694

RESUMO

We demonstrate the differential sensitivity of poorly differentiated and well differentiated human colon carcinoma cells to nutrients alone or to nutrients and polypeptide growth factors under completely serum-free conditions. 3H-Thymidine incorporation into trichloroacetic acid precipitable material and autoradiographic analysis indicated that nutrient replenishment alone was sufficient to initiate DNA synthesis in quiescent poorly differentiated cells, whereas defined polypeptide growth factors produced no additional effect. In contrast, well differentiated cells were mitogenically stimulated to a much greater extent by growth factors (epidermal growth factor + insulin + transferrin), than by nutrient replenishment alone. Expression of the c-myc protooncogene was increased approximately 5-fold after growth factor addition to the well differentiated cells. Maximal expression of c-myc occurred at 4 h post stimulation. In contrast, nutrients resulted in only a slight up-regulation of c-myc (1.8-fold) at approximately 90 min after addition. Addition of nutrients and/or growth factors to the poorly differentiated colon carcinoma cells resulted in an initial decline in c-myc expression (90 min), presumably due to removal of endogenous growth stimulators. Expression of c-myc returned to baseline levels by 24 h after additions. The results indicate that differential sensitivity to polypeptide growth factors is related to differentiation status in this model system and suggest that the insensitivity of poorly differentiated cells to exogenous growth factors may be due to a greater production of autocrine growth stimulators.


Assuntos
Neoplasias do Colo/metabolismo , Proteínas Proto-Oncogênicas/genética , Carcinoma/metabolismo , Diferenciação Celular , Divisão Celular/efeitos dos fármacos , Meios de Cultura/farmacologia , Replicação do DNA/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Insulina/farmacologia , Cinética , Proteínas Proto-Oncogênicas c-myc , RNA Neoplásico/metabolismo , Transferrina/farmacologia , Células Tumorais Cultivadas
16.
Cancer Lett ; 117(1): 41-50, 1997 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9233830

RESUMO

We demonstrate herein the ability of transforming growth factor-beta-2 (TGFbeta2) to potently activate extracellular signal-regulated kinase 2 (ERK2) in the highly TGFbeta-sensitive breast cancer cell (BCC) line Hs578T. The ERK2 isoform was activated by 3-fold within 5 min of TGFbeta2 addition to Hs578T cells. However, TGFbeta2 only slightly activated ERK2 (1.5-fold) in the partially TGFbeta-responsive BCC line MDA-MB-23 1. The magnitude of the difference in activation of ERK2 by TGFbeta2 in the two cell lines paralleled the difference in the IC50 values for TGFbeta inhibition of DNA synthesis; the IC50 value in the MDA-MB-231 cells was 32-fold greater than that in the Hs578T cells. Further, our data demonstrate that TGFbeta2 activated the stress-activated protein kinase/Jun N-terminal kinase (SAPK/JNK) type of mitogen-activated protein kinases (MAPKs); maximal induction levels were 2.5-fold above basal values and were attained at 30 min after TGFbeta2 treatment. Transient co-transfection of a luciferase reporter construct (3TP-Lux) containing three AP-1 sites and the plasminogen activator inhibitor-1 (PAI-1) promoter, in conjunction with a construct that directs expression of a dominant-negative mutant ERK2 (TAYF) protein, did not block the ability of TGFbeta to induce AP-1 or PAI-1 activity. In contrast, TAYF ERK2 was able to block EGF and insulin-induced 3TP-Lux-reporter activity. These results indicate that in these BCCs, the activation of ERK2 by TGFbeta is more tightly linked to the ability of TGFbeta to inhibit DNA synthesis than to the ability to stimulate promoter regions important for TGFbeta production and control of the extracellular matrix. In addition, this is the first demonstration that TGFbeta can activate the SAPK/JNK type of MAPK in TGFbeta-sensitive human BCCs.


Assuntos
Neoplasias da Mama/enzimologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Fator de Crescimento Transformador beta/farmacologia , DNA de Neoplasias/biossíntese , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteína Quinase 1 Ativada por Mitógeno , Fator de Transcrição AP-1/metabolismo , Células Tumorais Cultivadas
17.
J Anal Toxicol ; 9(1): 31-5, 1985.
Artigo em Inglês | MEDLINE | ID: mdl-2858603

RESUMO

The present studies demonstrate the instability of glutathione (GS) in urine and plasma and illustrate the importance of developing procedures to stabilize GS in biological fluids. The rapid loss of urinary GSH was not prevented by inhibition of gamma-glutamyltranspeptidas (gamma-GTP) with L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid (AT-125), by EDTA, or by sodium citrate. GS in urine could be stabilized for extended periods of time by acidification of samples immediately after collection. In order to accurately measure urine and plasma GS concentrations, samples were analyzed by a known additions technique. The relevance of these analytical procedures to metal excretion studies is discussed.


Assuntos
Glutationa/análise , Metais/urina , Animais , Glutationa/sangue , Glutationa/urina , Humanos , Concentração de Íons de Hidrogênio , Camundongos , gama-Glutamiltransferase/fisiologia
20.
J Cell Physiol ; 138(3): 450-8, 1989 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2647770

RESUMO

We report the continuous growth maintenance of untransformed and chemically transformed fibroblasts (AKR-2B, AKR-MCA cells) in low concentrations of serum (0.1% FBS). The cell lines established (AKR-0.1F, MCA-0.1F) proliferated at rates comparable to cells maintained under high serum conditions (10% FBS). Complete removal of serum from the cells did not induce quiescence. The MCA-0.1F cells were more similar to the untransformed AKR-2B fibroblasts in their morphology, saturation density, inability to form colonies under anchorage-independent conditions, steady-state level of c-myc expression, and kinetics of induction of c-myc in response to specific growth factors. This report demonstrates the utility of this cell line as a nonquiescent model system for investigating growth factor-specific effects in serum-free, cycling cells. Addition of transforming growth factor-beta (TGF-beta) (5 ng/ml) to proliferating MCA-0.1F cells, in the absence of any serum, induced a multilayered growth pattern at confluency, similar to that of AKR-MCA cells maintained in 10% FBS. Other growth factors tested did not elicit this effect. The induction of this growth pattern by TGF-beta was associated with a sustained induction of the c-myc proto-oncogene at confluency, but not with a restoration of anchorage-independent growth. The data suggest that TGF-beta may play a role in the up-regulation of c-myc at confluency previously described for AKR-MCA cells maintained in 10% serum.


Assuntos
Ciclo Celular , Fibroblastos/citologia , Substâncias de Crescimento/fisiologia , Animais , Adesão Celular , Linhagem Celular , Meios de Cultura , Regulação da Expressão Gênica , Camundongos , Fenótipo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-myc , Fatores de Crescimento Transformadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA