Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 61(24): 8811-9, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11751403

RESUMO

A coordinated growth arrest during mammary involution completes the dramatic changes in mammary cell proliferation seen during pregnancy and lactation. Signals regulating this arrest are poorly understood, despite their potential relevance to oncogenesis. Here we report that the arrest involves a unique pulse of p16(INK4A) expression in vivo, which accompanies decreased cyclin D1 expression and a shift to an active repressor E2F4 complex. We used INK4A/ARF-/- mice as well as cyclin D1 and p16(INK4A) transgenic strains to examine the physiological significance of these patterns. p16(INK4A) directly regulated the in vivo transition from E2F3 to E2F4 as the major E2F DNA binding activity, and its contribution to growth arrest was independent of cyclin D1. Transgenic cyclin D1 expression prevented normal terminal differentiation by ablating the p16(INK4A) pulse, abolishing the shift from E2F3 to E2F4, derepressing E2F target genes, and expanding a stem cell population. The effects of cyclin D1 were reversed by restoring p16(INK4A) but were not seen in INK4A/ARF-/- mice. Our results indicate that cyclin D1 may contribute to tumorigenesis by altering cell differentiation and demonstrate a significant function for p16(INK4A) in development in vivo. These regulatory mechanisms used during mammary involution offer a potential explanation for the protective effect of pregnancy against breast cancer.


Assuntos
Ciclina D1/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Células 3T3 , Animais , Diferenciação Celular/genética , Divisão Celular/fisiologia , Ciclina D1/biossíntese , Ciclina D1/genética , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Vetores Genéticos/genética , Humanos , Lactação/metabolismo , Lactação/fisiologia , Glândulas Mamárias Animais/fisiologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Transgênicos , Gravidez , Células Tumorais Cultivadas
2.
Cell ; 107(6): 763-75, 2001 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-11747812

RESUMO

To identify functions of the IKKalpha subunit of IkappaB kinase that require catalytic activity, we generated an Ikkalpha(AA) knockin allele containing alanines instead of serines in the activation loop. Ikkalpha(AA/AA) mice are healthy and fertile, but females display a severe lactation defect due to impaired proliferation of mammary epithelial cells. IKKalpha activity is required for NF-kappaB activation in mammary epithelial cells during pregnancy and in response to RANK ligand but not TNFalpha. IKKalpha and NF-kappaB activation are also required for optimal cyclin D1 induction. Defective RANK signaling or cyclin D1 expression results in the same phenotypic effect as the Ikkalpha(AA) mutation, which is completely suppressed by a mammary specific cyclin D1 transgene. Thus, IKKalpha is a critical intermediate in a pathway that controls mammary epithelial proliferation in response to RANK signaling via cyclin D1.


Assuntos
Ciclina D1/metabolismo , Células Epiteliais/metabolismo , Glicoproteínas/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caseínas/genética , Caseínas/metabolismo , Células Cultivadas , Ciclina D1/genética , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/genética , Humanos , Quinase I-kappa B , Lactação/fisiologia , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/transplante , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Osteoprotegerina , Gravidez , Proteínas Serina-Treonina Quinases/genética , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Fator de Necrose Tumoral , Transplante de Tecidos , Transgenes , Fator de Necrose Tumoral alfa/farmacologia
3.
Transgenic Res ; 10(5): 471-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11708657

RESUMO

Overexpression of the cyclin D1 oncogene and inactivation of the p53 tumor suppressor have both been implicated in substantial proportions of sporadic human breast cancers. Transgenic mice with cyclin D1 overexpression targeted to mammary tissue by the MMTV enhancer-promoter have been shown to develop mammary cancers. To investigate the relationship between pathways driven by cyclin D1 overexpression and p53 loss during the development of breast cancers, we crossed MMTV-cyclin D1 mice with p53 heterozygous null (p53+/-) mice. In such crossed mice, cyclin D1-driven mammary neoplasia would need to be substantially accelerated by p53 loss in order for mammary tumors to develop prior to the expected onset of non-mammary tumors characteristic of the p53-deficient background alone. Instead, in mice heterozygous or homozygous for p53 deficiency and simultaneously carrying the MMTV-cyclin D1 transgene, only tumors typically found in p53-deficient mice developed and mammary tumors were not observed. Interestingly, MMTV-cyclin D1/p53+/- mice appeared to develop these non-mammary tumors more rapidly than p53+/- mice, and a majority of the sampled non-mammary tumors from MMTV-cyclin D1/p53+/- mice showed 'ectopic' expression of the MMTV-driven cyclin D1 transgene. Within the constraints of possible genetic background effects and limited sensitivity due to the early emergence of non-mammary tumors, these observations provide no evidence that inactivation of p53 confers a major additional selective advantage to mammary cells overexpressing cyclin D1 in this animal model of human breast cancer. Interestingly, the results do raise the possibility that p53 inactivation might complement or cooperate with cyclin D1 deregulation during the development of some types of non-mammary tumors.


Assuntos
Ciclina D1/genética , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/genética , Vírus do Tumor Mamário do Camundongo/genética , Proteína Supressora de Tumor p53/genética , Animais , Feminino , Glândulas Mamárias Animais/fisiologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neoplasias/genética , Neoplasias/patologia
4.
Proc Natl Acad Sci U S A ; 98(17): 9847-52, 2001 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-11493707

RESUMO

Hepatitis C virus (HCV) is a leading cause of chronic hepatitis, cirrhosis, and hepatocellular carcinoma. The absence of culture systems permissive for HCV replication has presented a major bottleneck to antiviral development. We sought to recapitulate the early steps in the life cycle of HCV by means of DNA-based expression of viral genomic sequences. Here we report expression of replicating HCV RNA by using a, to our knowledge, novel binary expression system in which cells were transfected with a T7 polymerase-driven full-length HCV cDNA plasmid containing a cis-acting hepatitis Delta ribozyme to control 3' cleavage, and infected with vaccinia-T7 polymerase. HCV genomic and replicative strand synthesis, in addition to protein synthesis, was detectable and depended on full-length HCV sequences. Moreover, the system was capable of generating HCV RNA quasispecies, consistent with the action of the low-fidelity HCV NS5B RNA polymerase. IFN-alpha, but not ribavirin, directly inhibited the viral replicative cycle in these cells, identifying the virus itself and not solely the immune system as a direct target of IFN action. The availability of a cell-based test for viral replication will facilitate screening of inhibitory compounds, analysis of IFN-resistance mechanisms, and analysis of virus-host cell interactions.


Assuntos
Antivirais/farmacologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Interferon-alfa/farmacologia , Replicação Viral/efeitos dos fármacos , Amantadina/farmacologia , Animais , Bacteriófago T7/genética , Carcinoma Hepatocelular/patologia , Chlorocebus aethiops , DNA Complementar/genética , Genes Sintéticos , Hepacivirus/fisiologia , Humanos , Neoplasias Hepáticas/patologia , Oligodesoxirribonucleotídeos Antissenso/síntese química , Oligodesoxirribonucleotídeos Antissenso/genética , Reação em Cadeia da Polimerase , RNA Catalítico/genética , RNA Viral/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribavirina/farmacologia , Regiões Terminadoras Genéticas , Transfecção , Células Tumorais Cultivadas , Proteínas Virais/biossíntese , Proteínas Virais/genética
5.
J Clin Invest ; 107(9): 1093-102, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11342573

RESUMO

The relationship between abnormal cell proliferation and aberrant control of hormonal secretion is a fundamental and poorly understood issue in endocrine cell neoplasia. Transgenic mice with parathyroid-targeted overexpression of the cyclin D1 oncogene, modeling a gene rearrangement found in human tumors, were created to determine whether a primary defect in this cell-cycle regulator can cause an abnormal relationship between serum calcium and parathyroid hormone response, as is typical of human primary hyperparathyroidism. We also sought to develop an animal model of hyperparathyroidism and to examine directly cyclin D1's role in parathyroid tumorigenesis. Parathyroid hormone gene regulatory region--cyclin D1 (PTH--cyclin D1) mice not only developed abnormal parathyroid cell proliferation, but also developed chronic biochemical hyperparathyroidism with characteristic abnormalities in bone and, notably, a shift in the relationship between serum calcium and PTH. Thus, this animal model of human primary hyperparathyroidism provides direct experimental evidence that overexpression of the cyclin D1 oncogene can drive excessive parathyroid cell proliferation and that this proliferative defect need not occur solely as a downstream consequence of a defect in parathyroid hormone secretory control by serum calcium, as had been hypothesized. Instead, primary deregulation of cell-growth pathways can cause both the hypercellularity and abnormal control of hormonal secretion that are almost inevitably linked together in this common disorder.


Assuntos
Adenoma/etiologia , Ciclina D1/biossíntese , Hiperparatireoidismo/etiologia , Hormônio Paratireóideo/metabolismo , Neoplasias das Paratireoides/etiologia , Animais , Osso e Ossos/patologia , Cálcio/sangue , Proteínas de Ligação ao Cálcio/isolamento & purificação , Aberrações Cromossômicas , Transtornos Cromossômicos , Ciclina D1/genética , Rearranjo Gênico , Humanos , Hiperparatireoidismo/genética , Camundongos , Camundongos Transgênicos , Hormônio Paratireóideo/sangue , Hormônio Paratireóideo/genética
6.
Clin Infect Dis ; 32(7): 1004-9, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11264027

RESUMO

Neisseria meningitidis is an important cause of serious bacterial infections in children. We undertook a study to identify meningococcal infections of the blood, cerebrospinal fluid, or both of children in a defined geographic area to describe the burden of disease and the spectrum of illness. We reviewed the medical records of all children aged <18 years who had meningococcal infections at the 4 pediatric referral hospitals in Boston, Massachusetts, from 1981 through 1996. We identified 231 patients with meningococcal disease; of these 231 patients, 194 (84%) had overt disease and 37 (16%) had unsuspected disease. Clinical manifestations included meningitis in 150 patients, hypotension in 26, and purpura in 17. Sixteen patients (7%) died. Although meningococcal disease is devastating to a small number of children, we found that the burden of pediatric disease that it caused at the 4 pediatric referral centers in this geographic region was limited; that patients with overt meningococcal disease are most likely to have meningitis; and that individual practitioners are unlikely to encounter a patient with unsuspected meningococcal disease.


Assuntos
Infecções Meningocócicas/epidemiologia , Neisseria meningitidis , Adolescente , Boston/epidemiologia , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Estudos Prospectivos
7.
Ophthalmology ; 107(8): 1450-3, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10919886

RESUMO

OBJECTIVE: To evaluate the effect of the introduction of the Haemophilus influenzae B (Hib) vaccine (introduced first in 1985, then extended in 1990 to children at least 2 months of age) on the epidemiologic features of periorbital and orbital cellulitis. DESIGN: Retrospective, comparative case series. PARTICIPANTS: Three hundred fifteen pediatric inpatients. METHODS: Children at Massachusetts General Hospital and Massachusetts Eye and Ear Infirmary with discharge diagnosis of periorbital or orbital cellulitis from 1980 through 1998 were reviewed. MAIN OUTCOME MEASURES: Case rate, culture-positive isolates, and associated conditions. RESULTS: A total of 297 cases of periorbital cellulitis and 18 cases of orbital cellulitis were reviewed. Before 1990, there were 27 cases of Hib-related cellulitis (11.7% of total in that period), whereas after 1990, there were only three (3.5% of total; P = 0.028). The number of cases per year was significantly lower after 1990 (21.2 +/- 10.4 vs. 8.7 +/- 3.9; P = 0.008), as were the number of positive culture isolates (for any organism) after 1990 (76 [33. 0%] vs. 9 [10.6%]; P < 0.001). The medical conditions most commonly associated with periorbital cellulitis were sinusitis (44 [14.5%]) and upper respiratory infections (73 [26.6%]). All cases of orbital cellulitis were associated with sinusitis. CONCLUSIONS: The introduction of the Hib vaccine coincided with a sharp decline not only in the number of periorbital and orbital cellulitis cases related to H. influenzae, but also in the annual case rate. These data are consistent with a facilitative role for H. influenzae in the development of cellulitis secondary to other pathogens. They also may support restriction of the spectrum of antibiotics used to manage these conditions.


Assuntos
Celulite (Flegmão)/prevenção & controle , Infecções Oculares Bacterianas/prevenção & controle , Infecções por Haemophilus/prevenção & controle , Vacinas Anti-Haemophilus/administração & dosagem , Doenças Orbitárias/prevenção & controle , Vacinação , Celulite (Flegmão)/epidemiologia , Celulite (Flegmão)/microbiologia , Criança , Pré-Escolar , Infecções Oculares Bacterianas/epidemiologia , Infecções Oculares Bacterianas/microbiologia , Feminino , Infecções por Haemophilus/epidemiologia , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/isolamento & purificação , Humanos , Lactente , Masculino , Massachusetts/epidemiologia , Doenças Orbitárias/epidemiologia , Doenças Orbitárias/microbiologia , Estudos Retrospectivos , Sinusite/epidemiologia , Sinusite/microbiologia , Sinusite/prevenção & controle
8.
Mol Cell Biol ; 20(12): 4462-73, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10825210

RESUMO

CDC37 encodes a 50-kDa protein that targets intrinsically unstable oncoprotein kinases including Cdk4, Raf-1, and v-src to the molecular chaperone Hsp90, an interaction that is thought to be important for the establishment of signaling pathways. CDC37 is required for proliferation in budding yeast and is coexpressed with cyclin D1 in proliferative zones during mouse development, a finding consistent with a positive role in cell proliferation. CDC37 expression may not only be required to support proliferation in cells that are developmentally programmed to proliferate but may also be required in cells that are inappropriately induced to initiate proliferation by oncogenes. Here we report that mouse mammary tumor virus (MMTV)-CDC37 transgenic mice develop mammary gland tumors at a rate comparable to that observed previously in MMTV-cyclin D1 mice. Moreover, CDC37 was found to collaborate with MMTV-c-myc in the transformation of multiple tissues, including mammary and salivary glands in females and testis in males, and also collaborates with cyclin D1 to transform the female mammary gland. These data indicate that CDC37 can function as an oncogene in mice and suggests that the establishment of protein kinase pathways mediated by Cdc37-Hsp90 can be a rate-limiting event in epithelial cell transformation.


Assuntos
Proteínas de Ciclo Celular/genética , Transformação Celular Neoplásica , Ciclina D1/genética , Proteínas de Drosophila , Genes myc , Chaperonas Moleculares , Animais , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Transgênicos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo
9.
J Bacteriol ; 181(24): 7618-20, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10601222

RESUMO

The cdc64-1 mutation causes G(1) arrest in Saccharomyces cerevisiae corresponding to a type II Start phenotype. We report that CDC64 encodes Ala1p, an alanyl-tRNA synthetase. Thus, cdc64-1 might affect charging of tRNA(Ala) and thereby initiation of cell division.


Assuntos
Alanina-tRNA Ligase/genética , Genes Fúngicos , Saccharomyces cerevisiae/enzimologia , Divisão Celular/genética , Fase G1/genética , Fenótipo , Saccharomyces cerevisiae/genética
10.
Oncogene ; 18(19): 2988-96, 1999 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-10378694

RESUMO

Cell division is coupled to cell growth. Since some c-myc target genes are regulators of cell growth while others function in cell division pathways, c-myc is apparently poised at the interface of these processes. Cell culture systems have shown specific myc-associated growth phenotypes. Increased cell growth precedes DNA synthesis after myc activation in cells expressing myc-estrogen receptor fuson constructs and cells lacking c-myc exhibit a marked loss of protein synthesis. A number of candidate c-myc target genes regulate processes required for cell growth including rRNA transcription and processing, ribosomal protein transcription and translation, and translation initiation. These interactions all have the potential to account for the growth phenotypes in c-myc mutant cells. The ability of translation initiation factors, including eIF4E, to transform cells makes them particularly interesting targets of c-myc. Further evaluation of these target genes will provide important insights into growth control and c-myc's functions in cellular proliferation.


Assuntos
Divisão Celular/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Animais , Divisão Celular/genética , Regulação da Expressão Gênica , Humanos
11.
Curr Opin Genet Dev ; 9(1): 76-80, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10072360

RESUMO

Proliferating cells must increase their mass coordinately with cell division. Recent evidence suggests that coupling of cell growth with cell division might be achieved by making synthesis of activators of cell division particularly sensitive to the capacity of the cell's protein synthesis machinery.


Assuntos
Divisão Celular/genética , Replicação do DNA/genética , Animais , Proteínas de Ciclo Celular/genética , Regulação da Expressão Gênica , Humanos , Mitose/genética , Leveduras
12.
Mol Cell Biol ; 18(10): 5621-33, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9742079

RESUMO

Regulation of the mRNA cap binding protein (eIF4E) is critical to the control of cellular proliferation since this protein is the rate-limiting factor in translation initiation and transforms fibroblasts and since eIF4E mutants arrest budding yeast in the G1 phase of the cell cycle (cdc33). We previously demonstrated regulation of eIF4E by altered transcription of its mRNA in serum-stimulated fibroblasts and in response to c-myc. To identify additional factors regulating eIF4E transcription, we used linker-scanning constructs to characterize sites in the promoter of the eIF4E gene required for its expression. Promoter activity was dependent on sites at -5, -25, -45, and -75; the site at -75 included a previously described myc box. Electrophoretic mobility shift assays identified DNA-protein interactions at -25 and revealed a binding site (TTACCCCCCCTT) that is unique to the eIF4E promoter. Proteins of 68 and 97 kDa bound this site in UV cross-linking and Southwestern experiments. Levels of 4E regulatory factor activities correlated with c-Myc levels, eIF4E expression levels, and protein synthesis in differentiating U937 and HL60 cells, suggesting that these activities may function to regulate protein synthesis rates during differentiation. Since the eIF4E promoter lacked typical TATA and initiator elements, further studies of this novel initiator-homologous element should provide insights into mechanisms of transcription initiation and growth regulation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Iniciação de Peptídeos/genética , Regiões Promotoras Genéticas , Capuzes de RNA , Animais , Sequência de Bases , Sítios de Ligação , Southern Blotting , Western Blotting , Diferenciação Celular , Reagentes de Ligações Cruzadas , DNA Complementar , Regulação para Baixo , Fator de Iniciação 4E em Eucariotos , Células HL-60 , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Pirimidinas/metabolismo , RNA Mensageiro/genética , Transcrição Gênica , Células Tumorais Cultivadas , Raios Ultravioleta
13.
Genes Dev ; 11(19): 2522-31, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9334317

RESUMO

The eukaryotic cell cycle is driven by a cascade of cyclins and kinase partners including the G1 cyclin Cln3p in yeast. As the first step in this cascade, Cln3p is uniquely positioned to determine the critical growth-rate threshold for division. To analyze factors regulating CLN3 expression, we identified a short upstream open reading frame (uORF) in the 5' leader of CLN3 mRNA as a translational control element. This control element is critical for the growth-dependent regulation of Cln3p synthesis because it specifically represses CLN3 expression during conditions of diminished protein synthesis or slow growth. Inactivation of the uORF accelerates the completion of Start and entry into the cell cycle suggesting that translational regulation of CLN3 provides a mechanism coupling cell growth and division.


Assuntos
Divisão Celular/fisiologia , Ciclinas/genética , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Antifúngicos/farmacologia , Divisão Celular/genética , Ciclinas/biossíntese , Replicação do DNA/genética , Citometria de Fluxo , Proteínas Fúngicas/biossíntese , Genes Reporter , Mutação , Fases de Leitura Aberta/genética , Fatores de Iniciação de Peptídeos/genética , Polienos/farmacologia , Polirribossomos/genética , Biossíntese de Proteínas/genética , RNA Fúngico/genética , RNA Fúngico/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Análise de Sequência de DNA , Sirolimo
14.
Hepatology ; 26(3): 598-604, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9303488

RESUMO

The human insulin receptor substrate-1 (hIRS-1) is a key intracellular protein involved in various cytokine signaling pathways associated with cell growth. We have previously demonstrated that stable transfection and overexpression of hIRS-1 in human hepatoblastoma cells in vitro leads to the constitutive activation of the mitogen-activated protein kinase (MAPK) cascade. In this setting, hIRS-1 acts as a dominant oncogene and will induce neoplastic transformation of NIH 3T3 cells. In the present study, the biologic effects of hIRS-1 overexpression in the liver was analyzed using both clinical tumor samples and a newly developed transgenic mouse model. We have found that approximately 40% of 22 human hepatocellular carcinoma (HCC) tumors had enhanced (>200%) hIRS-1 gene expression compared with adjacent non-involved liver tissue. There was a significant relationship between the level of hIRS-1 overexpression and the tumor size; this finding suggests a possible role for hIRS-1 in tumor progression. To determine if downstream signal transduction cascades were activated by overexpression of hIRS-1 in hepatocytes, we established a transgenic mouse model using an hIRS-1 construct driven by an albumin promoter/enhancer element to direct liver specific expression. The overexpressed hIRS-1 protein was found to be tyrosyl phosphorylated and interacted with downstream SH2-containing molecules such as the p85 subunit of phosphatidylinositol-3 kinase (PI3K), Grb2 adaptor, and SHP2 phosphatase proteins. The functional consequences of hIRS-1 overexpression were reflected by constitutive activation of both the MAPK and PI3K signal transduction cascades. More important, overexpression of hIRS-1 in the transgenic liver led to increased hepatocyte DNA synthesis. Our findings indicate that hIRS-1 overexpression induces downstream signaling molecules associated with hepatocyte growth and may potentially enhance tumor progression of HCC.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Fosfoproteínas/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Células 3T3 , Animais , Humanos , Proteínas Substratos do Receptor de Insulina , Rim/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Fosfatidilinositol 3-Quinases , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Reação em Cadeia da Polimerase , Proteínas Recombinantes/biossíntese , Baço/metabolismo
15.
Somat Cell Mol Genet ; 23(3): 221-3, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9330633

RESUMO

We recently cloned genomic sequences containing the promoter region for the messenger RNA cap binding protein (eIF4E). As the rate-limiting step in translation, eukaryotic initiation factor 4E is important in cellular growth control. Using oligonucleotide primers specific for the promoter region in polymerase chain reactions (PCR), we amplified the human gene in a chromosome 4-specific human/rodent somatic cell panel. This panel mapped single copy genomic sequences for eIF4E in the region 4q21 to 4q25.


Assuntos
Mapeamento Cromossômico , Cromossomos Humanos Par 4/química , Genes , Fatores de Iniciação de Peptídeos/genética , Animais , Cricetinae , Fator de Iniciação 4E em Eucariotos , Humanos , Células Híbridas
16.
Hepatology ; 25(4): 1014-21, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9096613

RESUMO

Although hepatitis C virus (HCV) is a leading cause of morbidity and mortality worldwide, the role of viral cytopathic effects remains unclear. To study the biosynthesis of HCV structural proteins and their pathogenic role, we constructed transgenic mice, expressing type 1b HCV structural proteins (core, E1, and E2) in liver tissues. Two liver-specific promoters were used. The mouse major urinary protein (MUP) promoter has been shown to be developmentally regulated with little or no expression in utero but high-level expression after birth. The albumin (Alb) promoter provides constitutive, high levels of transgenes in live. Expression of both HCV transgenes was detected in several lines by Northern blots, HCV-specific reverse transcriptase-polymerase chain reactions (RT-PCR), and Western immunoblotting. Alb HCV lines showed higher levels of HCV expression than the MUP HCV lines. Immunohistochemical analysis revealed a predominantly cytoplasmic presence of core protein with occasional nuclear staining, and both cytoplasmic and membrane expression of the E2 protein in the transgenic livers. In both transgenes, the highest levels of both antigens were seen in perivenular hepatocytes, suggesting potential processing specificity in those cells. At six months of age, the livers of all transgenic lineages remained histologically normal. We concluded that HCV structural proteins are not directly cytopathic in this animal model.


Assuntos
Hepacivirus/genética , Proteínas Estruturais Virais/genética , Animais , Sequência de Bases , Efeito Citopatogênico Viral/genética , Primers do DNA/genética , Modelos Animais de Doenças , Expressão Gênica , Hepacivirus/metabolismo , Hepacivirus/patogenicidade , Hepatite C/etiologia , Hepatite C/genética , Hepatite C/virologia , Humanos , Imuno-Histoquímica , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Estruturais Virais/metabolismo , Virulência/genética
17.
Cancer Lett ; 113(1-2): 123-30, 1997 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-9065811

RESUMO

The cyclin D1/PRAD1 protooncogene is a key regulator of the G1 phase of the cell cycle and has been incriminated in the pathogenesis of a variety of primary human tumors. Recently, part of a novel alternatively spliced cyclin D1 transcript, called transcript b, has been identified. This variant transcript showed a failure of splicing at the 3' end of exon 4 and as a result, the expected protein product is altered at its C-terminus. Because of similar transcript sizes, previous Northern analyses would not have been expected to distinguish the two variants, and the relative levels of the two cyclin D1 transcripts in human tumors is unknown. To elucidate the role of cyclin D1 transcript b, we have isolated cDNA clones of this variant transcript from human breast cancer cell lines and report the sequence of the entire coding region of the cDNA. The protein predicted from the cDNA sequence consists of 274 amino acid residues and lacks a PEST sequence in its C-terminus. Examination of the levels of the two alternative cyclin D1 transcripts in primary breast cancers and breast cancer cell lines by Northern blot analysis and reverse transcription-polymerase chain reaction (RT-PCR) assays showed that the variant transcript b is indeed expressed in primary breast cancers and breast cancer cell lines, but the level of transcript b is dramatically lower than that of the originally reported transcript a of the cyclin D1 gene. In breast cancers, oncogenic overexpression of cyclin D1 mRNA appears to consist overwhelmingly of transcript a, and the role of transcript b, if any, in oncogenesis remains to be established. Science Ireland Ltd.


Assuntos
Neoplasias da Mama/genética , Ciclinas/genética , Proteínas Oncogênicas/genética , Processamento Alternativo , Sequência de Aminoácidos , Sequência de Bases , Northern Blotting , Neoplasias da Mama/metabolismo , Ciclina D1 , DNA Complementar/análise , Feminino , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Transcrição Gênica , Células Tumorais Cultivadas
19.
Mol Cell Biol ; 16(9): 4754-64, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8756633

RESUMO

The mRNA cap-binding protein (eukaryotic initiation factor 4E [eIF4E]) binds the m7 GpppN cap on mRNA, thereby initiating translation. eIF4E is essential and rate limiting for protein synthesis. Overexpression of eIF4E transforms cells, and mutations in eIF4E arrest cells in G, in cdc33 mutants. In this work, we identified the promoter region of the gene encoding eIF4E, because we previously identified eIF4E as a potential myc-regulated gene. In support of our previous data, a minimal, functional, 403-nucleotide promoter region of eIF4E was found to contain CACGTG E box repeats, and this core eIF4E promoter was myc responsive in cotransfections with c-myc. A direct role for myc in activating the eIF4E promoter was demonstrated by cotransfections with two dominant negative mutants of c-myc (MycdeltaTAD and MycdeltaBR) which equally suppressed promoter function. Furthermore, electrophoretic mobility shift assays demonstrated quantitative binding to the E box motifs that correlated with myc levels in the electrophoretic mobility shift assay extracts; supershift assays demonstrated max and USF binding to the same motif. cis mutations in the core or flank of the eIF4E E box simultaneously altered myc-max and USF binding and inactivated the promoter. Indeed, mutations of this E box inactivated the promoter in all cells tested, suggesting it is essential for expression of eIF4E. Furthermore, the GGCCACGTG(A/T)C(C/G) sequence is shared with other in vivo targets for c-myc, but unlike other targets, it is located in the immediate promoter region. Its critical function in the eIF4E promoter coupled with the known functional significance of eIF4E in growth regulation makes it a particularly interesting target for c-myc regulation.


Assuntos
Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Fatores de Iniciação de Peptídeos/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-myc/fisiologia , Sequências Repetitivas de Ácido Nucleico , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Fator de Iniciação 4E em Eucariotos , Fibroblastos , Células HeLa , Humanos , Modelos Genéticos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fatores de Iniciação de Peptídeos/biossíntese , Biossíntese de Proteínas/fisiologia , Capuzes de RNA/metabolismo , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção , Fatores Estimuladores Upstream
20.
Proc Natl Acad Sci U S A ; 93(18): 9577-82, 1996 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-8790372

RESUMO

Overexpression of the c-myc oncogene is associated with a variety of both human and experimental tumors, and cooperation of other oncogenes and growth factors with the myc family are critical in the evolution of the malignant phenotype. The interaction of hepatocyte growth factor (HGF) with c-myc during hepatocarcinogenesis in a transgenic mouse model has been analyzed. While sustained overexpression of c-myc in the liver leads to cancer, coexpression of HGF and c-myc in the liver delayed the appearance of preneoplastic lesions and prevented malignant conversion. Furthermore, tumor promotion by phenobarbital was completely inhibited in the c-myc/HGF double transgenic mice, whereas phenobarbital was an effective tumor promoter in the c-myc single transgenic mice. The results indicate that HGF may function as a tumor suppressor during early stages of liver carcinogenesis, and suggest the possibility of therapeutic application for this cytokine.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Animais , Apoptose , Divisão Celular , Regulação Neoplásica da Expressão Gênica , Genes myc , Humanos , Neoplasias Hepáticas Experimentais/induzido quimicamente , Camundongos , Camundongos Transgênicos , Fenobarbital/farmacologia , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA