Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Steroid Biochem Mol Biol ; 86(3-5): 477-86, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14623547

RESUMO

Long term exposure to estradiol increases the risk of breast cancer in a variety of animal species, as well as in women. The mechanisms responsible for this effect have not been firmly established. The prevailing theory proposes that estrogens increase the rate of cell proliferation by stimulating estrogen receptor-mediated transcription and thereby the number of errors occurring during DNA replication. An alternative hypothesis proposes that estradiol can be metabolized to quinone derivatives which can react with DNA and then remove bases from DNA through a process called depurination. Error prone DNA repair then results in point mutations. We postulate that these two processes, increased cell proliferation and genotoxic metabolite formation, act in an additive or synergistic fashion to induce cancer. If correct, aromatase inhibitors would block both processes whereas anti-estrogens would only inhibit receptor-mediated effects. Accordingly, aromatase inhibitors would be more effective in preventing breast cancer than use of anti-estrogens. Our studies initially demonstrated that catechol estrogen (CE) quinone metabolites are formed in MCF-7 human breast cancer cells in culture. Measurement of estrogen metabolites and conjugates involved utilization of an HPLC separation coupled with an electrochemical detector. We then utilized an animal model that allows dissociation of estrogen receptor-mediated function from that of the effects of estradiol metabolites. Wnt-1 transgenic mice harboring a knock-out of ERalpha provides a means of examining the effect of estrogen deprivation in the absence of the ER in animals with a high incidence of breast tumors. ERbeta was shown to be absent in the breast tissue of these animals by RNase protection assay. In the breast tissue of these estrogen receptor alpha knock-out (ERKO)/Wnt-1 transgenic mice, we demonstrated formation of genotoxic estradiol metabolites. The ERKO/Wnt-1 breast extracts contained picomole amounts of the 4-catechol estrogens, but not their methoxy conjugates nor the 2-CE or their methoxy conjugates. The 4-CE conjugates with glutathione or its hydrolytic products (cysteine and N-acetylcysteine) were detected in picomole amounts in both tumors and hyperplastic mammary tissue, demonstrating the formation of CE-3,4-quinones. These results are consistent with the hypothesis that mammary tumor development is primarily initiated by metabolism of estrogens to 4-CE and, then, to CE-3,4-quinones, which may react with DNA to induce oncogenic mutations. The next set of experiments examined the incidence of tumors formed in Wnt-1 transgenic mice bearing wild type ERalpha (ER+/+), the heterozygous combination of genes (ER+/ER-) or ERalpha knock-out (ER-/-). To assess the effect of estrogens in the absence of ER, half of the animals were oophorectomized on day 15 and the other half were sham operated. Castration reduced the incidence of breast tumors in all animal groups and demonstrated the dependence of tumor formation upon estrogens. A trend toward reduction in tumor number (not statistically significant at this interim analysis) occurred in the absence of functional ER since the number of tumors was markedly reduced in ERKO animals which were castrated early in life. In aggregate, our results support the concept that metabolites of estradiol may act in concert with ER mediated mechanisms to induce breast cancer.


Assuntos
Neoplasias da Mama/induzido quimicamente , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Estradiol/metabolismo , Estradiol/toxicidade , Neoplasias Mamárias Animais/induzido quimicamente , Animais , Aromatase/genética , Aromatase/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Catecol O-Metiltransferase/genética , Divisão Celular/genética , Estrona/análogos & derivados , Estrona/metabolismo , Humanos , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Mutação , Polimorfismo Genético , Fatores de Risco
2.
Breast Cancer Res Treat ; 68(2): 139-46, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11688517

RESUMO

We have previously shown that ornithine decarboxylase (ODC) overexpression enhances the transforming effects of HER-2neu and epidermal growth factor (EGF) in normal MCF-10A human breast epithelial cells. Our data suggest that such potentiation may be mediated by activation of the mitogen-activated protein kinase (MAPK) pathway and, possibly, STAT signalling. To further explore the interaction between the polyamine pathway and EGF/HER-2neu signalling in this system, we inhibited endogenous ODC activity with alpha-difluoromethylornithine (DFMO) and assessed the effects of this blockade on the expression of EGF receptors (EGFR) and HER-2neu as well as activation of downstream EGF target genes. We found that DFMO administration to MCF-10A cells increased EGF-R mRNA and protein levels in a dose-response fashion, while HER-2neu expression was not affected. The effect of DFMO was mediated through polyamine depletion since it could be reversed by exogenous putrescine administration. Our results also indicated that the increase in EGFR induced by DFMO was not a non-specific consequence of inhibition of cell proliferation. The upregulated EGFRs were functional since they could be phosphorylated by EGF and they were able to promote phosphorylation of downstream signalling molecules including ERK, STAT-3, and STAT-5. We propose that physiologic levels of ODC activity may be critical for regulation of a yet undefined signalling pathway, whose blockade by DFMO leads to a compensatory increase in functional EGFR.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Eflornitina/farmacologia , Receptores ErbB/metabolismo , Poliaminas/metabolismo , Receptor ErbB-2/metabolismo , Northern Blotting , Western Blotting , Neoplasias da Mama/patologia , Linhagem Celular , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Receptores ErbB/genética , Humanos , Hidroxiureia/farmacologia , Nocodazol/farmacologia , Ornitina Descarboxilase/metabolismo , Inibidores da Ornitina Descarboxilase , Proteínas Serina-Treonina Quinases/metabolismo , Receptor ErbB-2/genética , Transdução de Sinais , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
3.
Brain Res Mol Brain Res ; 84(1-2): 106-14, 2000 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-11113537

RESUMO

The native opioid growth factor (OGF), [Met(5)]-enkephalin, is a tonic inhibitory peptide that modulates cell proliferation and tissue organization during development, cancer, cellular renewal, wound healing, and angiogenesis. OGF action is mediated by a receptor mechanism. The receptor for OGF, OGFr, has been cloned and sequenced in humans and rats. Using primers based on the rat OGFr cDNA, and a mouse embryo expressed sequence tag, the full-length 2.1 kb mouse OGFr cDNA was sequenced. The open reading frame was found to encode a protein of 634 amino acids, and 14 imperfect repeats of 9 amino acids each were a prominent feature. The molecular weight of OGFr was calculated as 70679, and the isoelectric point was 4.5. Northern blot analysis revealed a 2.1 kb OGFr mRNA transcript in adult mouse brain, heart, lung, liver, kidney, and triceps surae muscle. The amino acids for mouse and rat OGFr were 93% similar and 91% identical, but the mouse and human shared only a 70% similarity and a 58% identity. These results emphasize the molecular validity of OGFr, and explain the interaction of OGF with respect to normal and abnormal growth in mouse cells and tissues.


Assuntos
Receptores Opioides/análise , Receptores Opioides/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Sequência Consenso , Sequência Conservada , Perfilação da Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Peso Molecular , RNA Mensageiro/análise , RNA Mensageiro/genética , Receptores Opioides/genética , Sequências Repetitivas de Aminoácidos , Alinhamento de Sequência , Análise de Sequência de DNA
4.
Int J Oncol ; 17(5): 1053-61, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11029512

RESUMO

The native opioid growth factor (OGF), [Met5]-enkephalin, is a tonic inhibitory peptide that modulates cell proliferation and migration, as well as tissue organization, during development, cancer, homeostatic cellular renewal, wound healing, and angiogenesis. OGF action is mediated by the OGF receptor (OGFr). To investigate the target of OGF as to cell proliferation, the effects of excess OGF, and a deprivation of OGF-OGFr interaction by an opioid antagonist, naltrexone (NTX), were examined in 3 human cancer cell lines: pancreatic (BxPC-3), colon (HT-29), and head and neck (CAL-27). OGF exposure decreased growth, DNA synthesis, and mitosis, and increased the doubling time from control levels. FACS analysis revealed a marked increase in cells in the G0/G1 phase and compensatory reduction in cells in S and G2/M phases. Consistent with this observation, the percentage of labeled mitosis (PLM) analysis showed a notable increase in the time of the G0/G1 phase. Receptor blockade with NTX increased the rate of growth, length of DNA synthesis and mitotic phases, and decreased doubling time from control values. FACS analysis indicated an increase in the proportion of cells in S and G2/M phases, and a decrease in the number of cells in the G0/G1 phase. PLM evaluation demonstrated a shortening of the length of the S and G2 phases in the 3 cell lines, and decreases in the M and G0/G1 phases in some cancers. These results indicate that OGF action is directed at the G0/G1 phase, but interruption of OGF-OGFr interfacing has widespread repercussions on the cell cycle. The data on blockade of OGF-OGFr during log phase growth suggest a requisite escorting of the growth peptide and its receptor through the cell cycle.


Assuntos
Ciclo Celular/fisiologia , Encefalina Metionina/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/patologia , Receptores Opioides/fisiologia , Adenocarcinoma/patologia , Carcinoma de Células Escamosas/patologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Replicação do DNA/efeitos dos fármacos , Encefalina Metionina/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neoplasias/metabolismo , Neoplasias Pancreáticas/patologia , Receptores Opioides/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
5.
Cancer Res ; 60(11): 2764-9, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10850410

RESUMO

We previously showed that introduction of a normal, neomycin-tagged human chromosome 11 reduces the metastatic capacity of MDA-MB-435 (435) human breast carcinoma cells by 70-90% without affecting tumorigenicity, suggesting the presence of one or more metastasis suppressor genes encoded on human chromosome 11. To identify the gene(s) responsible, differential display comparing chromosome 11-containing (neo11/ 435) and parental, metastatic cells was done. We describe the isolation and functional characterization of a full-length cDNA for one of the novel genes, designated breast-cancer metastasis suppressor 1 (BRMS1), which maps to human chromosome 11q13.1-q13.2. Stably transfected MDA-MB-435 and MDA-MB-231 breast carcinoma cells still form progressively growing, locally invasive tumors when injected into mammary fat pads but are significantly less metastatic to lungs and regional lymph nodes. These data provide compelling functional evidence that breast-cancer metastasis suppressor 1 is a novel mediator of metastasis suppression in human breast carcinoma.


Assuntos
Neoplasias da Mama/genética , Carcinoma Ductal de Mama/genética , Cromossomos Humanos Par 11/genética , Proteínas de Neoplasias , Proteínas/genética , Supressão Genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Mapeamento Cromossômico , DNA Complementar/metabolismo , Feminino , Humanos , Hibridização in Situ Fluorescente , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Modelos Genéticos , Dados de Sequência Molecular , Metástase Neoplásica , Transplante de Neoplasias , Proteínas Repressoras , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas
6.
Brain Res ; 856(1-2): 75-83, 2000 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-10677613

RESUMO

The native opioid growth factor (OGF), [Met(5)]-enkephalin, is a tonic inhibitory peptide that modulates cell proliferation and tissue organization during development, cancer, cellular renewal, wound healing, and angiogenesis. OGF action is mediated by a receptor mechanism. We have cloned and sequenced cDNAs encoding multiple spliced forms of a human OGF receptor. The open reading frame in the longest cDNA was found to encode a protein of 697 amino acids, and 8 imperfect repeats of 20 amino acids each were a prominent feature. Altogether, five alternatively spliced forms were observed. The cDNA hybridized to mRNA from a variety of normal and neoplastic cells and tissues. Functional studies using antisense oligonucleotides to OGFr demonstrated an enhancement in cell growth. Fluorescent in situ hybridization (FISH) experiments showed the chromosomal location to be 20q13.3. This OGF receptor has no homology to classical opioid receptors. These results provide molecular validity for the interaction of OGF and OGF receptor in the regulation of growth processes in humans.


Assuntos
Encéfalo/metabolismo , Cromossomos Humanos Par 20 , Receptores Opioides/genética , Receptores Opioides/metabolismo , Adulto , Processamento Alternativo , Sequência de Aminoácidos , Animais , Mapeamento Cromossômico , Clonagem Molecular , DNA Complementar , Feminino , Feto , Humanos , Masculino , Dados de Sequência Molecular , Neuroblastoma , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Fases de Leitura Aberta , Placenta/metabolismo , Gravidez , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos , Receptores Opioides/química , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sequências Repetitivas de Aminoácidos , Alinhamento de Sequência , Células Tumorais Cultivadas
7.
Clin Exp Metastasis ; 18(8): 683-93, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11827072

RESUMO

Introduction of normal, neomycin-tagged human chromosome 11 (neo11) reduces the metastatic capacity of MDA-MB-435 human breast carcinoma cells by 70-90% without affecting tumorigenicity. Differential display comparing MDA-MB-435 and neo11/435 led to the discovery of a human breast carcinoma metastasis suppressor gene, BRMS1, which maps to chromosome 11q13.1-q13.2. Stable transfectants of MDA-MB-435 and MDA-MB-231 breast carcinoma cells with BRMS1 cDNA still form progressively growing, locally invasive tumors when injected in mammary fat pads of athymic mice but exhibit significantly lower metastatic potential (50-90% inhibition) to lungs and regional lymph nodes. To begin elucidating the mechanism(s) of action, we measured the ability of BRMS1 to perturb individual steps of the metastatic cascade modeled in vitro. Consistent differences were not observed for adhesion to extracellular matrix components (laminin, fibronectin, type IV collagen, type I collagen, Matrigel); growth rates in vitro or in vivo; expression of matrix metalloproteinases, heparanase, or invasion. Likewise. BRMS1 expression did not up regulate expression of other metastasis suppressors, such as NM23, Kai1, KiSS1 or E-cadherin. Motility of BRMS1 transfectants was modestly inhibited (30-60%) compared to parental and vector-only transfectants. Ability to grow in soft agar was also decreased in MDA-MB-435 cells by 80-89%, but the decrease for MDA-MB-231 was less (13-15% reduction). Also, transfection and re-expression of BRMS1 restored the ability of human breast carcinoma cells to form functional homotypic gap junctions. Collectively, these data suggest that BRMS1 suppresses metastasis of human breast carcinoma by complex, atypical mechanisms.


Assuntos
Neoplasias Pulmonares/prevenção & controle , Neoplasias Mamárias Experimentais/prevenção & controle , Proteínas de Neoplasias , Proteínas/fisiologia , Animais , Northern Blotting , Southern Blotting , Primers do DNA/química , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Nus , Fosforilação , RNA Mensageiro/metabolismo , Proteínas Repressoras , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas/metabolismo
8.
Brain Res ; 849(1-2): 147-54, 1999 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-10592296

RESUMO

The native opioid growth factor (OGF), [Met(5)]enkephalin, is a tonic inhibitory peptide that modulates cell proliferation and tissue organization during development, cancer, cellular renewal, wound healing and angiogenesis. OGF action is mediated by a receptor mechanism. We have cloned and sequenced a 2.1-kilobase (kb) cDNA for a receptor to OGF (OGFr). The open reading frame was found to encode a protein of 580 amino acids, and eight imperfect repeats of nine amino acids each were a prominent feature. The protein encoded by this cDNA exhibited the pharmacological, temporal and spatial characteristics of the OGFr. Functional studies using antisense technology demonstrated an enhancement in cell growth. The molecular organization of the OGFr has no homology to classical opioid receptors. These results provide molecular validity for the interaction of OGF and OGFr in the regulation of growth processes.


Assuntos
Encéfalo/metabolismo , Encefalina Metionina/metabolismo , Regulação da Expressão Gênica , Receptores Opioides/genética , Receptores Opioides/fisiologia , Envelhecimento , Sequência de Aminoácidos , Animais , Sequência de Bases , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Clonagem Molecular , DNA Complementar , Regulação da Expressão Gênica/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Ratos , Receptores Opioides/química , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Tionucleotídeos
9.
Cancer Lett ; 135(1): 107-12, 1999 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-10077228

RESUMO

Human pancreatic cancer is stimulated by the autocrine production of gastrin. In this study, the effects of administration of antisense oligonucleotides to gastrin on growth of pancreatic cancer were evaluated in vitro and in vivo. Log phase BxPC-3 human pancreatic cancer cells in culture were exposed to increasing concentrations (0.5-10 microM) of a synthetic 20-mer antisense phosphorothioate oligonucleotide to gastrin for 48 h and growth was assessed by the cellular proliferation assay. Growth was inhibited up to 88% by anti-gastrin oligonucleotides in a dose-related fashion compared to cells treated with diluent or a randomized sequence with the same composition as the anti-gastrin oligonucleotide. In vivo nude mice bearing BxPC-3 xenografts were treated daily for 14 days with a 0.1-ml intratumoral injection of either anti-gastrin (5 microM), the scrambled sequence control phosphorothioate oligonucleotide (5 microM), or buffer. Tumors from the anti-gastrin-treated mice were significantly smaller in volume and weight and had less gastrin detected by radioimmunoassay than either controls. These results support the role of gastrin as a stimulatory peptide for growth of human pancreatic cancer. Antisense oligonucleotide to gastrin may have a role in the future treatment of patients with pancreatic cancer.


Assuntos
Antineoplásicos/farmacologia , Gastrinas/genética , Inibidores do Crescimento/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Animais , Antineoplásicos/administração & dosagem , Inibidores do Crescimento/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Oligonucleotídeos Antissenso/administração & dosagem , Neoplasias Pancreáticas/genética , Transplante Heterólogo , Células Tumorais Cultivadas
10.
Int J Mol Med ; 2(3): 309-15, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9855703

RESUMO

Gastrin has been shown to stimulate growth of human pancreatic cancer, and does so in an autocrine fashion. In this study, a relationship between gastrin mRNA, peptide, and gastrin receptors were studied in a variety of human pancreatic tissues. Low levels of gastrin mRNA were detected in normal human pancreas by quantitative reverse transcription polymerase chain reaction, but gastrin peptide was not present using radioimmunoassay. Pancreatic adenocarcinoma cells and tissues had 34- to 530-fold higher gastrin mRNA and peptide levels than normal pancreas. Gastrin mRNA and peptide levels were 8,000- and 15,000-fold, respectively, greater in a pancreatic islet cell gastrinoma tumor than in normal pancreas. In comparison to age-matched controls, fasting gastrin plasma levels were 2-fold higher in patients with pancreatic adenocarcinoma and 131-fold greater in subjects with gastrinomas. Receptor binding assays revealed that pancreatic cancer cells had a binding capacity 200-fold greater than gastrinoma tumors, and 10-fold greater than normal pancreas; no differences in K(d) values were recorded between specimens. In contrast to the normal pancreas and gastrinoma tumor, the aggressive behavior of pancreatic adenocarcinoma may be attributed to the autocrine production of gastrin and to the presence of its growth-related receptor.


Assuntos
Gastrinoma/química , Gastrinas/análise , Pâncreas/química , Neoplasias Pancreáticas/química , RNA Mensageiro/análise , Idoso , Animais , Northern Blotting , Linhagem Celular Tumoral , Feminino , Gastrinas/sangue , Gastrinas/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Transplante de Neoplasias , RNA Mensageiro/sangue , Radioimunoensaio , Receptor de Colecistocinina B/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
11.
Int J Cancer ; 76(4): 563-70, 1998 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-9590135

RESUMO

Our experiments were designed to test the cooperativity between the polyamine pathway and HER-2neu in inducing transformation of human mammary epithelial cells in culture. Using the MCF-10A breast epithelial cell line, we observed that induction of overexpression of ornithine decarboxylase (ODC) (the first rate-limiting enzyme in polyamine biosynthesis) markedly potentiated the anchorage-independent growth stimulating effect of the beta2 isoform of neu differentiating factor (NDF) known to activate HER-2neu in MCF-10A cells. ODC overexpression, on the other hand, did not enhance growth in liquid culture, thus pointing to a specific effect on transformation rather than proliferation. ODC-overexpressing MCF-10A cells exhibited increased MAPK phosphorylation in response to administration of NDF and/or epidermal growth factor (EGF). In contrast, the phosphorylation of the members of the stress-activated protein kinase cascade p38 and SEK were not affected by ODC overexpression. Of note, in the absence of EGF and NDF, ODC overexpression failed to induce both clonogenicity and MAPK activation. These results suggest that increased polyamine biosynthetic activity critically interacts with HER-2neu in promoting human mammary cell transformation in culture and that the MAPK cascade is an important mediator of this interaction. If confirmed in future in vivo studies, our results may identify important new targets for the chemoprevention of human breast cancer.


Assuntos
Neoplasias da Mama/etiologia , Mama/patologia , Transformação Celular Neoplásica , Receptores ErbB/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno , Poliaminas/metabolismo , Receptor ErbB-2/metabolismo , Mama/metabolismo , Neoplasias da Mama/enzimologia , Linhagem Celular , Células Clonais , Ativação Enzimática , Epitélio , Humanos , MAP Quinase Quinase 1 , Ornitina Descarboxilase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais
12.
J Virol ; 72(2): 1103-7, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9445005

RESUMO

About one-third of the MA protein in Rous sarcoma virus (RSV) is phosphorylated. Previous analyses of this fraction have suggested that serine residues 68 and 106 are the major sites of phosphorylation. As a follow-up to that study, we have characterized mutants which have these putative phosphorylation sites changed to alanine, either separately or together. None of the substitutions (S68A, S106A, or S68/106A) had an effect on the budding efficiency or infectivity of the virus. Upon examination of the 32P-labeled viral proteins, we found that the S68A substitution did not affect phosphorylation in vivo at all. In contrast, the S106A substitution prevented all detectable phosphorylation of MA, suggesting that there is only one major site of phosphorylation in MA. We also found that the RSV MA protein is phosphorylated on tyrosine, but the amount was low and detectable only with large numbers of virions and an antibody specific for phosphotyrosine.


Assuntos
Vírus do Sarcoma Aviário/fisiologia , Fosfoproteínas/genética , Proteínas da Matriz Viral/genética , Proteínas Virais/genética , Replicação Viral/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Células COS , Genoma Viral , Dados de Sequência Molecular , Mutação , Fosfoproteínas/metabolismo , Fosforilação , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/metabolismo
13.
Mol Biol Cell ; 8(5): 843-54, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9168470

RESUMO

Substrates critical for transformation by pp60v-src remain unknown, as does the precise role of the src homology 2 (SH2) domain in this process. To continue exploring the role of the SH2 domain in pp60v-src-mediated transformation, site-directed mutagenesis was used to create mutant v-src alleles predicted to encode proteins with overall structural integrity intact but with reduced ability to bind phosphotyrosine-containing peptides. Arginine-175, which makes critical contacts in the phosphotyrosine-binding pocket, was mutated to lysine or alanine. Unexpectedly, both mutations created v-src alleles that transform chicken cells with wild-type (wt) efficiency and are reduced for transformation of rat cells; these alleles are host dependent for transformation. Additionally, these alleles resulted in a round morphological transformation of chicken cells, unlike 12 of the 13 known host-dependent src SH2 mutations that result in a fusiform morphology. Analysis of phosphopeptide binding by the mutant SH2 domains reveal that the in vitro ability to bind phosphopeptides known to have a high affinity for wt src SH2 correlates with wt (round) morphological transformation in chicken cells and in vitro ability to bind phosphopeptides known to have a low affinity for wt src SH2 correlates with rat cell transformation. These results suggest that the search for critical substrates in rat cells should be among proteins that interact with pp60v-src with low affinity.


Assuntos
Transformação Celular Neoplásica , Proteína Oncogênica pp60(v-src)/fisiologia , Fosfopeptídeos/metabolismo , Domínios de Homologia de src , Animais , Linhagem Celular , Embrião de Galinha , Expressão Gênica , Mutação , Proteína Oncogênica pp60(v-src)/genética , Proteína Oncogênica pp60(v-src)/metabolismo , Ligação Proteica , Ratos , Especificidade da Espécie
14.
EMBO J ; 16(6): 1258-67, 1997 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-9135142

RESUMO

Two ligands for Eph-related receptor tyrosine kinases, RAGS and ELF-1, have been implicated in the control of development of the retinotectal projection. Both molecules are expressed in overlapping gradients in the tectum, the target area of retinal ganglion cell axons. In two in vitro assays ELF-1 is shown to have a repellent axon guidance function for temporal, but apparently not for nasal axons. RAGS on the other hand is repellent for both types of axons, though to different degrees. Thus, RAGS and ELF-1 share some and differ in other properties. The biological activities of these molecules correlate with the strength of interaction with their receptors expressed on RGC axons. The meaning of these findings for guidance of retinal axons in the tectum is discussed.


Assuntos
Axônios/metabolismo , Proteínas de Ligação a DNA/metabolismo , Retina/embriologia , Células Ganglionares da Retina/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Células COS , Embrião de Galinha , Primers do DNA/genética , DNA Complementar/genética , Proteínas de Ligação a DNA/genética , Efrina-A2 , Regulação da Expressão Gênica no Desenvolvimento , Técnicas In Vitro , Camundongos , Proteínas Nucleares , RNA/genética , RNA/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Retina/citologia , Retina/metabolismo , Colículos Superiores/citologia , Colículos Superiores/embriologia , Colículos Superiores/metabolismo , Fatores de Transcrição/genética , Transfecção , Vias Visuais/citologia , Vias Visuais/embriologia , Vias Visuais/metabolismo
15.
Int J Cancer ; 70(2): 175-82, 1997 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-9009157

RESUMO

In these experiments we tested the hypothesis that constitutive activation of polyamine(PA) biosynthesis may contribute to mammary carcinogenesis. Spontaneously immortalized normal human MCF-10A breast epithelial cells were infected with the retroviral vector pLOSN containing a cDNA which codes for a truncated and more stable ornithine decarboxylase (ODC), the rate-limiting enzyme in PA synthesis. Upon chronic selective pressure with alpha-difluoromethyl-ornithine (DFMO) (an irreversible inhibitor of ODC), infected MCF-10A cells exhibited an approximately 250-fold increase in ODC activity, which persisted despite discontinuation of DFMO. ODC-over-expressing MCF-10A cells showed a modest decrease in S-adenosylmethionine decarboxylase and an increase in spermidine/spermineN1-acetyltransferase. Analysis of cellular PA profile revealed a selective accumulation of putrescine without alterations in spermidine and spermine contents. Lesser degrees of increased ODC activity were obtained reproducibly by re-exposing the cells to incremental small doses of DFMO. We observed a bell-shaped dose-related positive effect of ODC activity on clonogenicity in soft agar of MCF-10A cells. Since anchorage-dependent growth was actually reduced, such positive influence on this feature of transformation was not a non-specific consequence of a growth advantage provided by ODC over-expression. In addition, we observed a close parallelism between the dose-dependent effects of ODC expression on clonogenicity and activity of the ERK-2 kinase, a central element of the MAPK cascade. Our data demonstrate an interaction between PA and the MAPK signalling pathway and suggest that the latter may be involved in ODC-induced transformation of mammary epithelial cells.


Assuntos
Mama/citologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Transformação Celular Neoplásica/genética , Ornitina Descarboxilase/fisiologia , Acetiltransferases/metabolismo , Adenosilmetionina Descarboxilase/metabolismo , Mama/efeitos dos fármacos , Mama/enzimologia , Linhagem Celular Transformada , Ensaio de Unidades Formadoras de Colônias , DNA Complementar/genética , Eflornitina/farmacologia , Indução Enzimática , Células Epiteliais , Epitélio/efeitos dos fármacos , Epitélio/enzimologia , Feminino , Genes ras , Humanos , Proteína Quinase 1 Ativada por Mitógeno , Ornitina Descarboxilase/biossíntese , Ornitina Descarboxilase/química , Ornitina Descarboxilase/genética , Inibidores da Ornitina Descarboxilase , Poliaminas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Transfecção
18.
J Virol ; 70(4): 2664-8, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8642704

RESUMO

The Gag protein of Rous sarcoma virus (RSV) can direct particle assembly and budding at the plasma membrane independently of the other virus-encoded products. A previous deletion analysis has suggested that the first 86 amino acids of RSV Gag constitute a large membrane-binding domain that is absolutely required for these processes. To test this hypothesis, we inserted these residues in place of the N-terminal membrane-binding domain of the pp60v-src, a transforming protein whose biological activity requires plasma membrane localization. The ability of the Src chimera to induce cellular transformation suggests that the RSV sequence indeed contains an independent, functional domain.


Assuntos
Vírus do Sarcoma Aviário/metabolismo , Membrana Celular/metabolismo , Produtos do Gene gag/metabolismo , Células 3T3 , Animais , Vírus do Sarcoma Aviário/genética , Sequência de Bases , Sítios de Ligação , Membrana Celular/virologia , Transformação Celular Viral , DNA Viral , Deleção de Genes , Produtos do Gene gag/genética , Camundongos , Dados de Sequência Molecular , Proteína Oncogênica pp60(v-src)/genética , Proteína Oncogênica pp60(v-src)/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
19.
Mol Biol Cell ; 6(8): 953-66, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7579711

RESUMO

The biochemical properties of several pp60v-src substrates believed to participate in src-mediated transformation were examined in cells expressing a kinase-active, transformation-defective v-src allele (v-src-F172 delta/Y416F) and its parental allele, v-src-F172 delta, a host-range--dependent allele that transforms chicken cells to a fusiform morphology, but does not transform rat cells. Because pp60v-src-F172 delta is dependent on autophosphorylation for transforming ability, these alleles provide a unique opportunity to examine the role of pp60v-src autophosphorylation in regulating substrate interactions. Increased pp125FAK tyrosine phosphorylation and high levels of pp60v-src-associated phosphotidylinositol-3' kinase activity were detected specifically in chicken cells exhibiting round, refractile transformation but not in cells transformed to a fusiform morphology. Increased pp125FAK kinase activity, but not increased pp125FAK tyrosine-phosphorylation correlated with pp60v-src autophosphorylation and increased anchorage-independent growth. Thus, pp125FAK and PI3'K may participate in morphological transformation by v-src. Furthermore, association of phosphorylated SHC with the adapter GRB2 correlated with increased anchorage-independent growth (and autophosphorylation) in both rat and chicken cells independent of the morphological phenotype induced. Therefore, host-range dependence for transformation may be regulated through association of SHC with GRB2, thus implicating SHC as a crucial substrate for src-dependent transformation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Moléculas de Adesão Celular/metabolismo , Transformação Celular Neoplásica , Genes src , Proteína Oncogênica pp60(v-src)/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas/metabolismo , Alelos , Animais , Linhagem Celular Transformada , Galinhas , Cortactina , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Proteína Adaptadora GRB2 , Proteínas Ativadoras de GTPase , Proteínas dos Microfilamentos/análise , Proteínas dos Microfilamentos/metabolismo , Proteína Oncogênica pp60(v-src)/genética , Fosfatidilinositol 3-Quinases , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Ratos , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Tirosina/metabolismo
20.
J Biol Chem ; 270(8): 3467-70, 1995 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-7876076

RESUMO

We have isolated a murine cDNA encoding a ligand for the Cek7 receptor protein-tyrosine kinase (RPTK), a member of the Eph/Eck RPTK subfamily. Sequence analysis predicts an open reading frame of 209 amino acids with a predicted molecular mass of 24 kDa. The Cek7 ligand shows a 48% sequence identity at the protein level to B61, a ligand for the related Eck RPTK, 30% to the Cek5 ligand, 59% to the recently cloned Ehk1-L, and identity to ELF-1, a recently described ligand for the Mek4 and Sek RPTKs. The expressed Cek7 ligand is functionally active as it induces autophosphorylation of the Cek7 RPTK.


Assuntos
Proteínas Fetais/metabolismo , Proteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Linhagem Celular , Clonagem Molecular , Cricetinae , DNA Complementar , Ativação Enzimática , Efrina-A2 , Humanos , Ligantes , Dados de Sequência Molecular , Proteínas/genética , Receptor EphA4 , Receptor EphA5 , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA