Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Bioinformatics ; 23(2): 184-90, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17105717

RESUMO

MOTIVATION: ANOVA is a technique, which is frequently used in the analysis of microarray data, e.g. to assess the significance of treatment effects, and to select interesting genes based on P-values. However, it does not give information about what exactly is causing the effect. Our purpose is to improve the interpretation of the results from ANOVA on large microarray datasets, by applying PCA on the individual variance components. Interaction effects can be visualized by biplots, showing genes and variables in one plot, providing insight in the effect of e.g. treatment or time on gene expression. Because ANOVA has removed uninteresting sources of variance, the results are much more interpretable than without ANOVA. Moreover, the combination of ANOVA and PCA provides a simple way to select genes, based on the interactions of interest. RESULTS: It is shown that the components from an ANOVA model can be summarized and visualized with PCA, which improves the interpretability of the models. The method is applied to a real time-course gene expression dataset of mesenchymal stem cells. The dataset was designed to investigate the effect of different treatments on osteogenesis. The biplots generated with the algorithm give specific information about the effects of specific treatments on genes over time. These results are in agreement with the literature. The biological validation with GO annotation from the genes present in the selections shows that biologically relevant groups of genes are selected. AVAILABILITY: R code with the implementation of the method for this dataset is available from http://www.cac.science.ru.nl under the heading "Software".


Assuntos
Algoritmos , Perfilação da Expressão Gênica/métodos , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Proteoma/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Simulação por Computador , Interpretação Estatística de Dados , Modelos Estatísticos , Análise de Componente Principal
2.
J Natl Cancer Inst ; 92(17): 1388-402, 2000 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-10974075

RESUMO

Signaling from transforming growth factor-beta (TGF-beta) through its unique transmembrane receptor serine-threonine kinases plays a complex role in carcinogenesis, having both tumor suppressor and oncogenic activities. Tumor cells often escape from the antiproliferative effects of TGF-beta by mutational inactivation or dysregulated expression of components in its signaling pathway. Decreased receptor function and altered ratios of the TGF-beta type I and type II receptors found in many tumor cells compromise the tumor suppressor activities of TGF-beta and enable its oncogenic functions. Recent identification of a family of intracellular mediators, the Smads, has provided new paradigms for understanding mechanisms of subversion of TGF-beta signaling by tumor cells. In addition, several proteins recently have been identified that can modulate the Smad-signaling pathway and may also be targets for mutation in cancer. Other pathways such as various mitogen-activated protein kinase cascades also contribute substantially to TGF-beta signaling. Understanding the interplay between these signaling cascades as well as the complex patterns of cross-talk with other signaling pathways is an important area of investigation that will ultimately contribute to understanding of the bifunctional tumor suppressor/oncogene role of TGF-beta in carcinogenesis.


Assuntos
Mutação , Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor/genética , Humanos , Neoplasias/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad2 , Proteína Smad3 , Proteína Smad4 , Transativadores/metabolismo , Fator de Crescimento Transformador beta/genética
3.
Adv Cancer Res ; 83: 1-54, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11665716

RESUMO

Transforming growth factor-beta (TGF-beta) has been implicated in oncogenesis since the time of its discovery almost 20 years ago. The complex, multifunctional activities of TGF-beta endow it with both tumor suppressor and tumor promoting activities, depending on the stage of carcinogenesis and the responsivity of the tumor cell. Dysregulation or alteration of TGF-beta signaling in tumorigenesis can occur at many different levels, including activation of the ligand, mutation or transcriptional suppression of the receptors, or alteration of downstream signal transduction pathways resulting from mutation or changes in expression patterns of signaling intermediates or from changes in expression of other proteins which modulate signaling. New insights into signaling from the TGF-beta receptors, including the identification of Smad signaling pathways and their interaction with mitogen-activated protein (MAP) kinase pathways, are providing an understanding of the changes involved in the change from tumor suppressor to tumor promoting activities of TGF-beta. It is now appreciated that loss of sensitivity to inhibition of growth by TGF-beta by most tumor cells is not synonymous with complete loss of TGF-beta signaling but rather suggests that tumor cells gain advantage by selective inactivation of the tumor suppressor activities of TGF-beta with retention of its tumor promoting activities, especially those dependent on cross talk with MAP kinase pathways and AP-1.


Assuntos
Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Diferenciação Celular , Genes Supressores de Tumor , Humanos , Modelos Biológicos , Mutação , Isoformas de Proteínas , Transdução de Sinais , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/genética
4.
Cell Signal ; 13(7): 483-90, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11516623

RESUMO

Transforming growth factor-beta (TGF-beta) isoforms regulate numerous cellular functions through binding to receptors with intrinsic serine/threonine kinase activity that transduce the intracellular signals via activation of Smad proteins. In this study, we examined the signalling pathways involved in TGF-beta1-mediated growth inhibition and migration in a human mast cell line, HMC-1. TGF-beta1 evoked optimal migration at 40 fM, whereas maximal growth inhibition was obtained at 400 pM. Protein tyrosine kinase inhibitors completely inhibited TGF-beta1-mediated migration, without affecting the antimitogenic response. Smad2 was phosphorylated upon TGF-beta1 treatment, both in the absence and presence of genistein. The mitogen-induced extracellular kinase (MEK) inhibitor, PD98059, blocked the migratory response without affecting growth inhibition. In contrast, the p38 MAP kinase inhibitor, SB203580, had no significant effect on either migration or growth inhibition. These results indicate that different signalling pathways mediate TGF-beta1-induced migration and growth inhibition in HMC-1 cells, where the migration involves MEK activity.


Assuntos
Movimento Celular/efeitos dos fármacos , MAP Quinase Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases , Mastócitos/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Genisteína/farmacologia , Humanos , Mastócitos/citologia , Mastócitos/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosforilação , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteína Smad2 , Transativadores/metabolismo , Fator de Crescimento Transformador beta1 , Proteínas Quinases p38 Ativadas por Mitógeno
5.
J Leukoc Biol ; 67(3): 350-6, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10733095

RESUMO

Mast cells are known to accumulate at sites of inflammation, however, the chemotaxins involved remain largely undefined. Transforming growth factor-beta (TGF-beta) isoforms regulate numerous cellular functions, including cell growth and differentiation, formation of extracellular matrix, and the immune response. In this study we have compared the potency of different members of the TGF-beta family as human mast cell chemotaxins, and analyzed the expression of TGF-beta binding proteins on human mast cells. We were able to demonstrate that the maximal chemotactic response was attained at approximately 40 fM for the three TGF-beta isoforms, with TGF-beta3 being more effective than TGF-beta1 and TGF-beta2 at this concentration. This effect was observed in both the HMC-1 human mast cell line and in cultured primary mast cells. In addition, TGF-beta1, TGF-beta2, and less efficiently, TGF-beta3 inhibited the proliferation of HMC-1 cells. The migratory response is probably mediated through interaction with the TGF-beta serine/threonine type I and II receptors that were found to be expressed on the cells. No expression of TGF-beta type III receptor, endoglin, or the endothelial TGF-beta type I receptor ALK-1 could be detected. These results provide evidence that TGF-beta isoforms are highly potent chemotaxins for human mast cells and can play an important role in the recruitment of mast cells in inflammatory reactions.


Assuntos
Receptores de Ativinas Tipo I , Quimiotaxia/efeitos dos fármacos , Mastócitos/citologia , Mastócitos/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Anticorpos/imunologia , Anticorpos/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Reagentes de Ligações Cruzadas , Relação Dose-Resposta a Droga , Expressão Gênica , Humanos , Mastócitos/metabolismo , Testes de Precipitina , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
6.
J Bone Miner Res ; 13(3): 383-92, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9525338

RESUMO

Cartilage-derived morphogenetic proteins-1 and -2 (CDMP-1 and CDMP-2) are members of the bone morphogenetic protein (BMP) family, which play important roles in embryonic skeletal development. We studied the biological activities of recombinant CDMP-1 and CDMP-2 in chondrogenic and osteogenic differentiation and investigated their binding properties to type I and type II serine/threonine kinase receptors. In vivo, CDMP-1 and CDMP-2 were capable of inducing dose-dependently de novo cartilage and bone formation in an ectopic implantation assay. In vitro studies using primary chondrocyte cultures showed that both CDMP-1 and CDMP-2 stimulated equally de novo synthesis of proteoglycan aggrecan in a concentration-dependent manner. This activity was equipotent when compared with osteogenic protein-1 (OP-1). In contrast, CDMPs were less stimulatory than OP-1 in osteogenic differentiation as evaluated by alkaline phosphatase activity and expression levels of bone markers in ATDC5, ROB-C26, and MC3T3-E1 cells. CDMP-2 was the least osteogenic in these assays. Receptor binding studies of CDMP-1 and CDMP-2 revealed that both have affinity for the BMP receptor type IB (BMPR-IB) and BMPR-II, and weakly for BMPR-IA. Moreover, using a promoter/reporter construct, transcriptional activation signal was transduced by BMPR-IB in the presence of BMPR-II upon CDMP-1 and CDMP-2 binding. Our data show that distinct members of the BMP family differentially regulate the progression in the osteogenic lineage, and this may be due to their selective affinity for specific receptor complexes.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Cartilagem/fisiologia , Proteínas da Matriz Extracelular , Substâncias de Crescimento/farmacologia , Osteogênese/efeitos dos fármacos , Proteínas Serina-Treonina Quinases , Receptores de Fatores de Crescimento Transformadores beta , Agrecanas , Fosfatase Alcalina/metabolismo , Animais , Desenvolvimento Ósseo/efeitos dos fármacos , Proteína Morfogenética Óssea 7 , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Fator 5 de Diferenciação de Crescimento , Substâncias de Crescimento/biossíntese , Humanos , Lectinas Tipo C , Camundongos , Proteoglicanas/biossíntese , RNA/genética , RNA/isolamento & purificação , Receptores de Fatores de Crescimento/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia
7.
Endocrinology ; 139(3): 1306-13, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9492066

RESUMO

The rat TRH precursor contains five copies of TRH separated by connecting peptides. Previous studies have shown that the decapeptide prepro-TRH (160-169; Ps4) potentiates the effect of TRH on TSH secretion. In the present study, we have characterized Ps4 receptors in the rat pituitary by in vitro autoradiography using [125I-Tyr0]Ps4 as a radioligand, and we have investigated the evolution of receptor density during ontogenesis. Incubation of rat pituitary slices with [125I-Tyr0]Ps4 revealed intense binding in the anterior lobe and virtually no binding in the neurointermediate lobe. Biochemical characterization of the Ps4-binding sites suggested the existence of a single class of sites exhibiting high affinity for [Tyr0]Ps4 (IC50 = 8.3 +/- 1.2 nM) and a much lower affinity for Ps4 (IC50 = 9.3 +/- 1.2 microM). Emulsion-coated cytoautoradiography performed on cultured anterior pituitary cells showed that only 26% of the cells possessed [125I-Tyr0]Ps4-binding sites. Immunocytochemical analysis using antibodies against the different anterior pituitary hormones indicated that the cells possessing [125I-Tyr0]Ps4-binding sites did not correspond to TSH-, PRL-, GH-, ACTH-, or LH-secreting cells. In contrast, cells expressing Ps4 receptors were immunoreactive for the S-100 protein, a marker of folliculo-stellate cells. During postnatal development, a 4-fold increase in the concentration of [125I-Tyr0]Ps4-binding sites occurred from birth to weaning in the pituitary, with a marked and transient increase at the time of weaning. Thereafter, the density of sites declined gradually until day 60. In conclusion, the present study shows that folliculo-stellate cells express [125I-Tyr0]Ps4-binding sites in the anterior pituitary, and that these sites are developmentally regulated. The present data suggest that the potentiating effect of Ps4 on TRH-induced TSH secretion is mediated by folliculo-stellate cells.


Assuntos
Fragmentos de Peptídeos/metabolismo , Adeno-Hipófise/metabolismo , Precursores de Proteínas/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Fatores Etários , Animais , Sítios de Ligação , Células Cultivadas , Imuno-Histoquímica , Radioisótopos do Iodo , Masculino , Adeno-Hipófise/citologia , Ratos , Ratos Wistar
8.
Endocrinology ; 140(9): 4300-10, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10465304

RESUMO

Thyroid growth and function are intricately regulated by both positive and negative factors. In the present study, we have investigated the expression of transforming growth factor-beta (TGF-beta) super-family members and their receptors in normal porcine thyroid follicle cells. In tissue sections of porcine thyroids, we observed an expression of TGF-beta1, activin A, and bone morphogenetic protein (BMP)-7 proteins. The staining was localized to the follicular epithelium. In affinity cross-linking experiments, TGF-beta1 was found to bind to heteromeric complexes of TGF-beta type I and type II receptors, and activin A bound most efficiently to heteromeric complexes of activin type IB and type II receptors. We were unable to detect any BMP receptors (BMPRs) in attempts to perform affinity cross-linking with BMP-7. However, expression of BMPR-IA and BMPR-II messenger RNA (mRNA) was detected by Northern blot analysis. Both TGF-beta1 and activin A, but not BMP-7, increased the phosphorylation of Smad2, induced nuclear translocation of Smad2, Smad3, and Smad4, and inhibited thyrocyte cell growth as well as TSH-stimulated cAMP response. TGF-beta1 was more potent, compared with activin A, to induce these cellular responses. Taken together, our findings indicate a role for several members of the TGF-beta family in regulation of thyroid growth and function.


Assuntos
Inibinas/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Glândula Tireoide/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Receptores de Ativinas , Ativinas , Animais , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Divisão Celular/fisiologia , Células Cultivadas , Proteínas de Ligação a DNA/fisiologia , Ligantes , Valores de Referência , Transdução de Sinais/fisiologia , Proteína Smad2 , Proteína Smad3 , Suínos , Glândula Tireoide/citologia , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/fisiologia , Transativadores/fisiologia
9.
Chest ; 120(1 Suppl): 43S-47S, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11451911

RESUMO

Transforming growth factor (TGF)-beta plays a central role in fibrosis, contributing both to the influx and activation of inflammatory cells, as well as to activation of fibroblasts to elaborate extracellular matrix. In the past few years, new insight has been gained into signal transduction pathways downstream of the TGF-beta receptor serine-threonine kinases with the identification of a family of evolutionarily conserved Smad proteins. Two receptor-activated Smad proteins, Smad2 and Smad3, are phosphorylated by the activated TGF-beta type I receptor kinase, after which they partner with the common mediator, Smad4, and are translocated to the nucleus to where they participate in transcriptional complexes to control expression of target genes. We have shown in wound healing studies of mice null for Smad3, that loss of this key signaling intermediate interferes with the chemotaxis of inflammatory cells to TGF-beta as well as with their ability to autoinduce TGF-beta. Moreover, studies with mouse embryo fibroblasts null for Smad3 show that TGF-beta-dependent induction of c-Jun and c-Fos, important in induction of collagen as well as in autoinduction of TGF-beta, is mediated by Smad3. Based on these observations, we hypothesize that loss of Smad3 will confer resistance to fibrosis and result in reduced inflammatory cell infiltrates, reduced autoinduction of TGF-beta, important to sustain the process, and reduced elaboration of collagen. Preliminary observations in a model of radiation-induced fibrosis confirm this hypothesis and suggest that inhibitors of Smad3 might have clinical application both to improve wound healing and to reduce fibrosis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Fibrose Pulmonar/fisiopatologia , Transdução de Sinais/fisiologia , Transativadores/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Cicatrização/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Fosforilação , Proteína Smad3
10.
J Mammary Gland Biol Neoplasia ; 6(1): 67-82, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11467453

RESUMO

Ligands of the TGF-beta superfamily are unique in that they signal through transmembrane receptor serine-threonine kinases, rather than tyrosine kinases. The receptor complex couples to a signal transduction pathway involving a novel family of proteins, the Smads. On phosphorylation, Smads translocate to the nucleus where they modulate transcriptional responses. However, TGF-betas can also activate the mitogen-activated protein kinase (MAPK)4 pathway, and the different biological responses to TGF-beta depend to varying degrees on activation of either or both of these two pathways. The Smad pathway is a nexus for cross-talk with other signal transduction pathways and for modulation by many different interacting proteins. Despite compelling evidence that TGF-beta has tumor suppressor activity in the mammary gland, neither TGF-beta receptors nor Smads are genetically inactivated in human breast cancer, though receptor expression is reduced. Possible reasons are discussed in relation to the dual role of TGF-beta as tumor suppressor and oncogene.


Assuntos
Mama/crescimento & desenvolvimento , Neoplasias Mamárias Animais/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Humanos , Fator de Crescimento Transformador beta/genética
11.
FASEB J ; 13(15): 2105-24, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10593858

RESUMO

Transforming growth factor-beta (TGF-beta) superfamily members are multifunctional cell-cell signaling proteins that play pivotal roles in tissue homeostasis and development of multicellular animals. They mediate their pleiotropic effects from membrane to nucleus through distinct combinations of type I and type II serine/threonine kinase receptors and their downstream effectors, known as Smad proteins. Certain Smads, termed receptor-regulated Smads, become phosphorylated by activated type I receptors and form heteromeric complexes with a common-partner Smad4, which translocates into the nucleus to control gene transcription. In addition to these signal transducing Smads, inhibitory Smads have been identified that inhibit the activation of receptor-regulated Smads. In contrast to the still growing TGF-beta superfamily (with approximately 30 members in mammals), relatively few type I and type II receptors as well as Smads have been identified. We will focus on recent insights into the molecular mechanisms by which signaling specificity between different TGF-beta superfamily members is achieved and regulated, and how a single family member can elicit a broad scala of biological responses.-Piek, E., Heldin, C.-H., ten Dijke, P. Specificity, diversity, and regulation in TGF-beta superfamily signaling.


Assuntos
Proteínas Serina-Treonina Quinases , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento Transformadores beta , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos , Isoformas de Proteínas/metabolismo , Fatores de Transcrição/metabolismo
12.
J Cell Physiol ; 173(3): 447-59, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9369958

RESUMO

We have characterized a 60-kDa transforming growth factor-beta (TGF-beta) binding protein that was originally identified on LNCaP adenocarcinoma prostate cells by affinity cross-linking of cell surface proteins by using 125I-TGF-beta 1. Binding of 125I-TGF-beta 1 to the 60-kDa protein was competed by an excess of unlabeled TGF-beta 1 but not by TGF-beta 2, TGF-beta 3, activin, or osteogenic protein-1 (OP-1), also termed bone morphogenetic protein-7 (BMP-7). In addition, no binding of 125I-TGF-beta 2 and 125I-TGF-beta 3 to the 60-kDa binding protein on LNCaP cells could be demonstrated by using affinity labeling techniques. The 60-kDa TGF-beta binding protein showed no immunoreactivity with antibodies against the known type I and type II receptors for members of the TGF-beta superfamily. Treatment of LNCaP cells with 0.25 M NaCl, 1 microgram/ml heparin, or 10% glycerol caused a release of the 60-kDa protein from the cell surface. In addition, we found that the previously described TGF-beta type IV receptor on GH3 cells, which does not form a heterometric complex with TGF-beta receptors, could be released from the cell surface by these same treatments. This suggests that the 60-kDa protein and the similarly sized TGF-beta type IV receptor are related proteins. The eluted 60-kDa LNCaP protein was shown to interfere with the binding of TGF-beta to the TGF-beta receptors. Thus, the cell surface-associated 60-kDa TGF-beta binding protein may play a role in regulating TGF-beta binding to TGF-beta receptors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Fator de Crescimento Transformador beta/metabolismo , Adenocarcinoma , Sítios de Ligação , Proteínas de Transporte/isolamento & purificação , Divisão Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Humanos , Cinética , Proteínas de Ligação a TGF-beta Latente , Ligantes , Masculino , Peso Molecular , Neoplasias da Próstata , Fator de Crescimento Transformador beta/farmacologia , Células Tumorais Cultivadas
13.
J Cell Sci ; 112 ( Pt 24): 4557-68, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10574705

RESUMO

The capacities of different transforming growth factor-(beta) (TGF-(beta)) superfamily members to drive epithelial to mesenchymal transdifferentiation of the murine mammary epithelial cell line NMuMG were investigated. TGF-(beta)1, but not activin A or osteogenic protein-1 (OP-1)/bone morphogenetic protein-7 (BMP-7), was able to induce morphological transformation of NMuMG cells as shown by reorganisation of the actin cytoskeleton and relocalisation/downregulation of E-cadherin and (beta)-catenin, an effect that was abrogated by the more general serine/threonine kinase and protein kinase C inhibitor, staurosporine. TGF-(beta)1 bound to TGF-(beta) type I receptor (T(beta)R-I)/ALK-5 and T(beta)R-II, but not to activin type I receptor (ActR-I)/ALK-2. Activin A bound to ActR-IB/ALK-4 and ActR-II, and BMP-7 bound to ActR-I/ALK-2, BMP type I receptor (BMPR-I)/ALK-3, ActR-II and BMPR-II. TGF-(beta)1 and BMP-7 activated the Smad-binding element (SBE)(4) promoter with equal potency, whereas activin A had no effect. Transfection of constitutively active (CA)-ALK-4 activated the 3TP promoter to the same extent as TGF-(beta)1 and CA-ALK-5 indicating that activin signalling downstream of type I receptors was functional in NMuMG cells. In agreement with this, activin A induced low levels of plasminogen activator inhibitor I expression compared to the high induction by TGF-(beta)1. In contrast to activin A and BMP-7, TGF-(beta)1 strongly induced Smad2 phosphorylation. Consistent with these findings, TGF-(beta)1 induced the nuclear accumulation of Smad2 and/or Smad3. In addition, NMuMG cells transiently infected with adenoviral vectors expressing high level CA-ALK-5 exhibited full transdifferentiation. On the other hand, infections with low level CA-ALK-5, which alone did not result in transdifferentiation, together with Smad2 and Smad4, or with Smad3 and Smad4 led to transdifferentiation. In conclusion, TGF-(beta)1 signals potently and passes the activation threshold to evoke NMuMG cell transdifferentiation. The TGF-(beta) type I receptor (ALK-5) and its effector Smad proteins mediate the epithelial to mesenchymal transition. Activin A does not induce mesenchymal transformation, presumably because the number of activin receptors is limited, while BMP-7-initiated signalling cannot mediate transdifferentiation.


Assuntos
Receptores de Ativinas Tipo I , Diferenciação Celular/fisiologia , Glândulas Mamárias Animais/citologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transativadores/fisiologia , Ativinas , Animais , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Inibinas/fisiologia , Luciferases/genética , Mesoderma/citologia , Camundongos , Inibidores de Proteínas Quinases , Receptor do Fator de Crescimento Transformador beta Tipo I , Fator de Crescimento Transformador beta/fisiologia , Células Tumorais Cultivadas
14.
Dev Biol ; 264(1): 275-88, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14623248

RESUMO

Pericytes have been suggested to play a role in regulation of vessel stability; one mechanism for this stabilization may be via pericyte-derived vascular endothelial growth factor (VEGF). To test the hypothesis that differentiation of mesenchymal cells to pericytes/smooth muscle cells (SMC) is accompanied by VEGF expression, we used endothelial cell (EC) and mesenchymal cell cocultures to model cell-cell interactions that occur during vessel development. Coculture of EC and 10T1/2 cells, multipotent mesenchymal cells, led to induction of VEGF expression by 10T1/2 cells. Increased VEGF expression was dependent on contact between EC-10T1/2 and was mediated by transforming growth factorbeta (TGFbeta). A majority of VEGF produced in coculture was cell- and/or matrix-associated. Treatment of cells with high salt, protamine, heparin, or suramin released significant VEGF, suggesting that heparan sulfate proteoglycan might be sequestering some of the VEGF. Inhibition of VEGF in cocultures led to a 75% increase in EC apoptosis, indicating that EC survival in cocultures is dependent on 10T1/2-derived VEGF. VEGF gene expression in developing retinal vasculature was observed in pericytes contacting newly formed microvessels. Our observations indicate that differentiated pericytes produce VEGF that may act in a juxtacrine/paracrine manner as a survival and/or stabilizing factor for EC in microvessels.


Assuntos
Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Endotélio Vascular/fisiologia , Pericitos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C3H , Pericitos/citologia , Proteínas Recombinantes de Fusão/metabolismo , Retina/anatomia & histologia , Retina/crescimento & desenvolvimento , Retina/metabolismo , Proteína Smad3 , Transativadores/genética , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
15.
Proc Natl Acad Sci U S A ; 98(12): 6686-91, 2001 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-11390996

RESUMO

Epithelial-mesenchymal transitions (EMTs) are an essential manifestation of epithelial cell plasticity during morphogenesis, wound healing, and tumor progression. Transforming growth factor-beta (TGF-beta) modulates epithelial plasticity in these physiological contexts by inducing EMT. Here we report a transcriptome screen of genetic programs of TGF-beta-induced EMT in human keratinocytes and propose functional roles for extracellular response kinase (ERK) mitogen-activated protein kinase signaling in cell motility and disruption of adherens junctions. We used DNA arrays of 16,580 human cDNAs to identify 728 known genes regulated by TGF-beta within 4 hours after treatment. TGF-beta-stimulated ERK signaling mediated regulation of 80 target genes not previously associated with this pathway. This subset is enriched for genes with defined roles in cell-matrix interactions, cell motility, and endocytosis. ERK-independent genetic programs underlying the onset of EMT involve key pathways and regulators of epithelial dedifferentiation, undifferentiated transitional and mesenchymal progenitor phenotypes, and mediators of cytoskeletal reorganization. The gene expression profiling approach delineates complex context-dependent signaling pathways and transcriptional events that determine epithelial cell plasticity controlled by TGF-beta. Investigation of the identified pathways and genes will advance the understanding of molecular mechanisms that underlie tumor invasiveness and metastasis.


Assuntos
Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/fisiologia , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Integrinas/fisiologia , Queratinócitos/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia
16.
Int J Cancer ; 80(5): 756-63, 1999 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10048979

RESUMO

A panel of 6 human glioma cell lines was examined for TGF-beta1 responsiveness. U-178 MG and U-251 MG AgCl1 were significantly inhibited by TGF-beta1, while U-343 MGa 31L and U-343 MGa 35L were potently stimulated to proliferate. TGF-beta1 induced endogenous PAI-1 protein synthesis, Smad binding element/(CAGA)12-luciferase-reporter activity, as well as mRNA expression of Smad6 and Smad7 in all gliomas. Interestingly, TGF-beta1 differentially stimulated or inhibited the expression of TbetaR-I and TbetaR-II mRNA in the gliomas. Affinity cross-linking studies using 125I-TGF-beta1 revealed that the gliomas expressed TGF-beta-type-I(TbetaR-I) and -type-II(TbetaR-II) receptors, although binding to TbetaR-II in U-343 MGa 31L and U-251 MG AgCl1 was low to undetectable. Smad2 protein was abundantly present in U-178 MG, U-343 MG, and U-343 MGa 35L, while Smad3 was readily detectable in U-178 MG, U-343 MG, U-343 MGa 35L and U-251 MG AgCl1. In all gliomas, TGF-beta1 induced phosphorylation of Smad2. The level to which TGF-beta1 could activate the pathway leading to induction of the (CAGA)12-luciferase reporter seemed to correlate to the expression levels of TGF-beta receptors, Smad3 and Smad4 proteins. However, despite the plethora of data regarding TGF-beta1 signalling in the different glioma cell lines, the mechanism underlying the differential growth effects mediated by TGF-beta1 is still unclear. The results suggest that a complex balance between several components in the TGF-beta signalling pathway controls glioma responsiveness to TGF-beta1, and extend reports indicating that distinct signal transduction pathways are involved in growth inhibition and other cellular responses.


Assuntos
Proteínas de Ligação a DNA/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta/farmacologia , DNA de Neoplasias/biossíntese , Genes Reporter , Glioblastoma , Glioma , Humanos , Luciferases/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteína Smad2 , Proteína Smad3 , Proteína Smad4 , Transativadores/genética , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas
17.
J Cell Physiol ; 180(2): 141-9, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10395283

RESUMO

Activin A and osteogenic protein-1 (OP-1) exerted antagonistic effects on each other's responses on the human Tera-2 embryonal carcinoma cell line. OP-1 dose dependently inhibited activin A-induced activation of p3TP-Lux transcriptional reporter, containing part of the human plasminogen activator inhibitor-1 (PAI-1) promoter, while activin A inhibited OP-1-mediated alkaline phosphatase induction. Approximately equimolar concentrations of both growth factors resulted in 50% inhibition of the respective biological responses. Affinity cross-linking studies using 125I-activin A or 125I-OP-1 followed by receptor-immunoprecipitations revealed that both ligands bound to the activin type II receptor (ActR-II), but recruited different type I receptors. In addition, OP-1 competed with binding of 125I-activin A, and activin A competed with binding of 125I-OP-1 to ActR-II. Transient transfection studies showed that competition between activin A and OP-1 also occurred at the type I receptor (ActR-1) level; constitutively active (CA)-ActR-I inhibited CA-ActR-IB-mediated p3TP-Lux reporter induction. There was no competition between activin A and OP-1 for availability of Smad4, indicating that the concentration of this common signal transducer is not limiting for generating the observed biological responses. Overexpression of ActR-II abolished the inhibitory effect of OP-1 on activin A-induced p3TP-Lux activation and, surprisingly, led to OP-1-induced transcriptional reporter activity. Whereas the exact mechanism of competition is unclear, the role of ActR-II in the competition between activin A and OP-1 is discussed in light of the observed interference in downstream signaling by CA-ActR-I and CA-ActR-IB.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Substâncias de Crescimento/metabolismo , Inibinas/metabolismo , Células-Tronco Neoplásicas/enzimologia , Receptores de Ativinas Tipo I , Receptores de Activinas Tipo II , Ativinas , Fosfatase Alcalina/metabolismo , Northern Blotting , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/análise , Proteínas Morfogenéticas Ósseas/genética , Proteínas de Ligação a DNA/genética , Células-Tronco de Carcinoma Embrionário , Ativação Enzimática/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Reporter , Substâncias de Crescimento/análise , Substâncias de Crescimento/genética , Humanos , Inibinas/análise , Inibinas/genética , Células-Tronco Neoplásicas/química , Inibidor 1 de Ativador de Plasminogênio/genética , Regiões Promotoras Genéticas/fisiologia , Ligação Proteica/fisiologia , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/análise , Receptores de Fatores de Crescimento/metabolismo , Proteína Smad6 , Proteína Smad7 , Transativadores/genética , Transcrição Gênica/fisiologia , Fator de Crescimento Transformador beta/análise , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/enzimologia
18.
J Biol Chem ; 276(23): 19945-53, 2001 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-11262418

RESUMO

A prominent pathway of transforming growth factor (TGF)-beta signaling involves receptor-dependent phosphorylation of Smad2 and Smad3, which then translocate to the nucleus to activate transcription of target genes. To investigate the relative importance of these two Smad proteins in TGF-beta1 signal transduction, we have utilized a loss of function approach, based on analysis of the effects of TGF-beta1 on fibroblasts derived from mouse embryos deficient in Smad2 (S2KO) or Smad3 (S3KO). TGF-beta1 caused 50% inhibition of cellular proliferation in wild-type fibroblasts as assessed by [(3)H]thymidine incorporation, whereas the growth of S2KO or S3KO cells was only weakly inhibited by TGF-beta1. Lack of Smad2 or Smad3 expression did not affect TGF-beta1-induced fibronectin synthesis but resulted in markedly suppressed induction of plasminogen activator inhibitor-1 by TGF-beta1. Moreover, TGF-beta1-mediated induction of matrix metalloproteinase-2 was selectively dependent on Smad2, whereas induction of c-fos, Smad7, and TGF-beta1 autoinduction relied on expression of Smad3. Investigation of transcriptional activation of TGF-beta-sensitive reporter genes in the different fibroblasts showed that activation of the (Smad binding element)(4)-Lux reporter by TGF-beta1 was dependent on expression of Smad3, but not Smad2, whereas activation of the activin response element-Lux reporter was strongly suppressed in S2KO fibroblasts but, on the contrary, enhanced in S3KO cells. Our findings indicate specific roles for Smad2 and Smad3 in TGF-beta1 signaling.


Assuntos
Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Supressoras de Tumor , Animais , Divisão Celular , Inibidor de Quinase Dependente de Ciclina p15 , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Proteínas de Ligação a DNA/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Genes Precoces , Genes Reporter , Genes fos , Camundongos , Camundongos Knockout , Proteína Smad2 , Proteína Smad3 , Transativadores/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/biossíntese
19.
J Biol Chem ; 271(22): 12873-8, 1996 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-8662786

RESUMO

Expression of the platelet-derived growth factor alpha-receptor (PDGFalphaR) gene is tightly controlled in mammalian embryogenesis. A well established model system to study human embryogenesis is the embryonal carcinoma cell line Tera2. We have shown previously that retinoic acid-differentiated Tera2 cells express two PDGFalphaR transcripts of 6.4 kilobase pairs (kb) (encoding the full-length receptor) and 3.0 kb, respectively, whereas in contrast, undifferentiated Tera2 cells express PDFGalphaR transcripts of 1.5 kb and 5.0 kb. Here we show that this switch in PDGFalphaR expression pattern during differentiation of Tera2 cells results from alternative promoter use. In undifferentiated cells, a second promoter is used, which is located in intron 12 of the PDGFalphaR gene. Functional analysis shows that this promoter contains a consensus octamer motif, which can be bound by the POU domain transcription factor Oct-4. Oct-4 is expressed in undifferentiated Tera2 cells but not in retinoic acid-induced differentiated cells. Mutation of the octamer motif decreases promoter activity, while ectopic expression of Oct-4 in differentiated Tera2 cells specifically enhances the activity of this PDGFalphaR promoter. Therefore, we suggest that an important aspect in the maintenance of the undifferentiated state of human embryonal carcinoma cells results from Oct-4 expression, which thereupon activates this PDGFalphaR promoter.


Assuntos
Carcinoma Embrionário/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regiões Promotoras Genéticas , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , Fatores de Transcrição/metabolismo , Processamento Alternativo , Animais , Sequência de Bases , Sítios de Ligação , Carcinoma Embrionário/patologia , Sequência Consenso , Regulação Neoplásica da Expressão Gênica , Humanos , Dados de Sequência Molecular , Fator 3 de Transcrição de Octâmero , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA