Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Br J Cancer ; 107(10): 1684-91, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23099809

RESUMO

BACKGROUND: The aim of this study was to investigate the value of the cyclin D1 isoforms D1a and D1b as prognostic factors and their relevance as predictors of response to adjuvant chemotherapy with 5-fluorouracil and levamisole (5-FU/LEV) in colorectal cancer (CRC). METHODS: Protein expression of nuclear cyclin D1a and D1b was assessed by immunohistochemistry in 335 CRC patients treated with surgery alone or with adjuvant therapy using 5-FU/LEV. The prognostic and predictive value of these two molecular markers and clinicopathological factors were evaluated statistically in univariate and multivariate survival analyses. RESULTS: Neither cyclin D1a nor D1b showed any prognostic value in CRC or colon cancer patients. However, high cyclin D1a predicted benefit from adjuvant therapy measured in 5-year relapse-free survival (RFS) and CRC-specific survival (CSS) compared to surgery alone in colon cancer (P=0.012 and P=0.038, respectively) and especially in colon cancer stage III patients (P=0.005 and P=0.019, respectively) in univariate analyses. An interaction between treatment group and cyclin D1a could be shown for RFS (P=0.004) and CSS (P=0.025) in multivariate analysis. CONCLUSION: Our study identifies high cyclin D1a protein expression as a positive predictive factor for the benefit of adjuvant 5-FU/LEV treatment in colon cancer, particularly in stage III colon cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Ciclina D1/biossíntese , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Terapia Combinada/métodos , Intervalo Livre de Doença , Feminino , Fluoruracila/administração & dosagem , Seguimentos , Humanos , Imuno-Histoquímica/métodos , Levamisol/administração & dosagem , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Estudos Prospectivos , Recidiva , Resultado do Tratamento
2.
Oncogene ; 26(43): 6307-18, 2007 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-17452985

RESUMO

Functional inactivation of the retinoblastoma tumor suppressor gene product (RB) is a common event in human cancers. Classically, RB functions to constrain cellular proliferation, and loss of RB is proposed to facilitate the hyperplastic proliferation associated with tumorigenesis. To understand the repertoire of regulatory processes governed by RB, two models of RB loss were utilized to perform microarray analysis. In murine embryonic fibroblasts harboring germline loss of RB, there was a striking deregulation of gene expression, wherein distinct biological pathways were altered. Specifically, genes involved in cell cycle control and classically associated with E2F-dependent gene regulation were upregulated via RB loss. In contrast, a program of gene expression associated with immune function and response to pathogens was significantly downregulated with the loss of RB. To determine the specific influence of RB loss during a defined period and without the possibility of developmental compensation as occurs in embryonic fibroblasts, a second system was employed wherein Rb was acutely knocked out in adult fibroblasts. This model confirmed the distinct regulation of cell cycle and immune modulatory genes through RB loss. Analyses of cis-elements supported the hypothesis that the majority of those genes upregulated with RB loss are regulated via the E2F family of transcription factors. In contrast, those genes whose expression was reduced with the loss of RB harbored different promoter elements. Consistent with these analyses, we found that disruption of E2F-binding function of RB was associated with the upregulation of gene expression. In contrast, cells harboring an RB mutant protein (RB-750F) that retains E2F-binding activity, but is specifically deficient in the association with LXCXE-containing proteins, failed to upregulate these same target genes. However, downregulation of genes involved in immune function was readily observed with disruption of the LXCXE-binding function of RB. Thus, these studies demonstrate that RB plays a significant role in both the positive and negative regulations of transcriptional programs and indicate that loss of RB has distinct biological effects related to both cell cycle control and immune function.


Assuntos
Ciclo Celular , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/metabolismo , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Transcrição Gênica/genética , Animais , Células Cultivadas , Regulação para Baixo , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Retinoblastoma/genética , Retinoblastoma/imunologia , Proteína do Retinoblastoma/genética , Regulação para Cima
4.
Oncogene ; 25(11): 1620-8, 2006 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-16550162

RESUMO

The cyclin D1 proto-oncogene exercises powerful control over the mechanisms that regulate the mitotic cell cycle, and excessive cyclin D1 expression and/or activity is common in human cancers. Although somatic mutations of the cyclin D1 locus are rarely observed, mounting evidence demonstrates that a specific polymorphism of cyclin D1 (G/A870) and a protein product of a potentially related alternate splicing event (cyclin D1b) may influence cancer risk and outcome. Herein, we review the epidemiological and functional literatures that link these alterations of cyclin D1 to human tumor development and progression.


Assuntos
Processamento Alternativo , Ciclina D1/genética , Neoplasias/epidemiologia , Neoplasias/genética , Polimorfismo Genético , Ciclo Celular/fisiologia , Transformação Celular Neoplásica/genética , Ciclina D1/fisiologia , Progressão da Doença , Estudos Epidemiológicos , Humanos , Neoplasias/fisiopatologia , Proto-Oncogene Mas
5.
Mol Cell Biol ; 17(10): 5771-83, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9315635

RESUMO

The growth suppression function of RB is dependent on its protein binding activity. RB contains at least three distinct protein binding functions: (i) the A/B pocket, which binds proteins with the LXCXE motif; (ii) the C pocket, which binds the c-Abl tyrosine kinase; and (iii) the large A/B pocket, which binds the E2F family of transcription factors. Phosphorylation of RB, which is catalyzed by cyclin-dependent protein kinases, inhibits all three protein binding activities. We have previously shown that LXCXE binding is inactivated by the phosphorylation of two threonines (Thr821 and Thr826), while the C pocket is inhibited by the phosphorylation of two serines (Ser807 and Ser811). In this report, we show that the E2F binding activity of RB is inhibited by two sets of phosphorylation sites acting through distinct mechanisms. Phosphorylation at several of the seven C-terminal sites can inhibit E2F binding. Additionally, phosphorylation of two serine sites in the insert domain can inhibit E2F binding, but this inhibition requires the presence of the RB N-terminal region. RB mutant proteins lacking all seven C-terminal sites and two insert domain serines can block Rat-1 cells in G1. These RB mutants can bind LXCXE proteins, c-Abl, and E2F even after they become phosphorylated at the remaining nonmutated sites. Thus, multiple phosphorylation sites regulate the protein binding activities of RB through different mechanisms, and a constitutive growth suppressor can be generated through the combined mutation of the relevant phosphorylation sites in RB.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Proteínas de Ligação a DNA , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo , Animais , Divisão Celular , Células Cultivadas , Fatores de Transcrição E2F , Fase G1 , Humanos , Fosforilação , Ligação Proteica , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Ratos , Proteínas Recombinantes de Fusão , Proteína 1 de Ligação ao Retinoblastoma , Serina/metabolismo , Fator de Transcrição DP1
6.
Mol Cell Biol ; 18(7): 4032-42, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9632788

RESUMO

Growth suppression by the retinoblastoma protein (RB) is dependent on its ability to form complexes with transcription regulators. At least three distinct protein-binding activities have been identified in RB: the large A/B pocket binds E2F, the A/B pocket binds the LXCXE peptide motif, and the C pocket binds the nuclear c-Abl tyrosine kinase. Substitution of Trp for Arg 661 in the B region of RB (mutant 661) inactivates both E2F and LXCXE binding. The tumor suppression function of mutant 661 is not abolished, because this allele predisposes its carriers to retinoblastoma development with a low penetrance. In cell-based assays, 661 is shown to inhibit G1/S progression. This low-penetrance mutant also induces terminal growth arrest with reduced but detectable activity. We have constructed mutations that disrupt C pocket activity. When overproduced, the RB C-terminal fragment did not induce terminal growth arrest but could inhibit G1/S progression, and this activity was abolished by the C-pocket mutations. In full-length RB, the C-pocket mutations reduced but did not abolish RB function. Interestingly, combination of the C-pocket and 661 mutations completely abolished RB's ability to cause an increase in the percentage of cells in G1 and to induce terminal growth arrest. These results suggest that the A/B or C region can induce a prolongation of G1 through mechanisms that are independent of each other. In contrast, long-term growth arrest requires combined activities from both regions of RB. In addition, E2F and LXCXE binding are not the only mechanisms through which RB inhibits cell growth. The C pocket also contributes to RB-mediated growth suppression.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Divisão Celular , Proteínas de Ligação a DNA , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo , Sítios de Ligação , Clonagem Molecular , DNA/metabolismo , Fatores de Transcrição E2F , Fase G1 , Humanos , Mutagênese , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína do Retinoblastoma/genética , Proteína 1 de Ligação ao Retinoblastoma , Fase S , Fator de Transcrição DP1 , Fatores de Transcrição/genética , Células Tumorais Cultivadas
7.
Mol Cell Biol ; 20(20): 7751-63, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11003670

RESUMO

The retinoblastoma tumor suppressor protein (RB) is a potent inhibitor of cell proliferation. RB is expressed throughout the cell cycle, but its antiproliferative activity is neutralized by phosphorylation during the G(1)/S transition. RB plays an essential role in the G(1) arrest induced by a variety of growth inhibitory signals. In this report, RB is shown to also be required for an intra-S-phase response to DNA damage. Treatment with cisplatin, etoposide, or mitomycin C inhibited S-phase progression in Rb(+/+) but not in Rb(-/-) mouse embryo fibroblasts. Dephosphorylation of RB in S-phase cells temporally preceded the inhibition of DNA synthesis. This S-phase dephosphorylation of RB and subsequent inhibition of DNA replication was observed in p21(Cip1)-deficient cells. The induction of the RB-dependent intra-S-phase arrest persisted for days and correlated with a protection against DNA damage-induced cell death. These results demonstrate that RB plays a protective role in response to genotoxic stress by inhibiting cell cycle progression in G(1) and in S phase.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Dano ao DNA , Proteínas de Ligação a DNA , Proteína do Retinoblastoma/metabolismo , Fase S , Animais , Morte Celular/efeitos dos fármacos , Cisplatino/farmacologia , Ciclina A/antagonistas & inibidores , Ciclina A/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/deficiência , Ciclinas/genética , Ciclinas/fisiologia , DNA/biossíntese , Dano ao DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Fatores de Transcrição E2F , Etoposídeo/farmacologia , Fibroblastos , Citometria de Fluxo , Imunofluorescência , Deleção de Genes , Camundongos , Camundongos Knockout , Mitomicina/farmacologia , Testes de Mutagenicidade , Fosforilação/efeitos dos fármacos , Ratos , Proteína do Retinoblastoma/genética , Proteína 1 de Ligação ao Retinoblastoma , Fase S/efeitos dos fármacos , Fator de Transcrição DP1 , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
8.
Mol Cell Biol ; 21(12): 4032-45, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11359910

RESUMO

The retinoblastoma tumor suppressor protein (RB) is a negative regulator of the cell cycle that inhibits both G(1) and S-phase progression. While RB-mediated G(1) inhibition has been extensively studied, the mechanism utilized for S-phase inhibition is unknown. To delineate the mechanism through which RB inhibits DNA replication, we generated cells which inducibly express a constitutively active allele of RB (PSM-RB). We show that RB-mediated S-phase inhibition does not inhibit the chromatin binding function of MCM2 or RPA, suggesting that RB does not regulate the prereplication complex or disrupt early initiation events. However, activation of RB in S-phase cells disrupts the chromatin tethering of PCNA, a requisite component of the DNA replication machinery. The action of RB was S phase specific and did not inhibit the DNA damage-mediated association of PCNA with chromatin. We also show that RB-mediated PCNA inhibition was dependent on downregulation of CDK2 activity, which was achieved through the downregulation of cyclin A. Importantly, restoration of cyclin-dependent kinase 2 (CDK2)-cyclin A and thus PCNA activity partially restored S-phase progression in the presence of active RB. Therefore, the data presented identify RB-mediated regulation of PCNA activity via CDK2 attenuation as a mechanism through which RB regulates S-phase progression. Together, these findings identify a novel pathway of RB-mediated replication inhibition.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteína do Retinoblastoma/metabolismo , Fase S/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Cromatina/metabolismo , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina , Primers do DNA/genética , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Ratos , Proteína de Replicação A , Proteína do Retinoblastoma/genética , Transdução de Sinais
9.
Cancer Res ; 58(9): 2042-9, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9581851

RESUMO

An important early event in the differentiation of skeletal muscle cells is exit from the cell cycle, after which full expression of the muscle phenotype occurs. Rhabdomyosarcoma (RMS), a tumor of skeletal muscle origin, expresses a number of muscle-specific proteins, including MyoD; however, these cells fail to arrest or differentiate when cultured in differentiation medium (DM). To determine the basis for the failure of RMS cells to differentiate or arrest, we studied the molecular response of the embryonal RMS cell line, RD, to culture in DM. Under these conditions, the retinoblastoma protein (RB) was primarily in the hyperphosphorylated state. This is in contrast to myoblasts cultured in DM, in which the hypophosphorylated form of RB is exclusively present. Measurements of the expression and activities of cyclin-dependent kinases (cdks) cdk2 and cdk4 indicated that RD cells maintained higher levels than do myoblasts, and the activity and abundance of these proteins did not significantly decrease upon culture in DM in RD cells, as they did in myoblasts. Similarly, elevated expression of cyclins D1, E, and A was observed in RD cells. Interestingly, cdk inhibitors are expressed in RD cells, with p16ink4 expression markedly elevated relative to myoblasts. Ectopic expression of p21cip1, p16ink4, or p27kip1 caused a growth arrest of RD cells but not detectable expression of a myogenic marker. Furthermore, a constitutively active RB protein could also inhibit the growth of RD cells without inducing myogenic differentiation. Taken together, these data suggest that the elevated levels of cdk2 and/or cdk4 observed in RD cells contribute to the inability of RD cells to growth arrest when cultured in DM but that these activities alone are not responsible for the failure of RD cells to differentiate.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Neoplasias Musculares/metabolismo , Rabdomiossarcoma/metabolismo , Ciclo Celular , Diferenciação Celular , Meios de Cultivo Condicionados , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Neoplasias Musculares/patologia , Proteína MyoD/metabolismo , Fosforilação , Proteína do Retinoblastoma/metabolismo , Rabdomiossarcoma/patologia , Células Tumorais Cultivadas
10.
Oncogene ; 16(13): 1655-63, 1998 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-9582013

RESUMO

The retinoblastoma tumor suppressor RB and its related proteins, p107 and p130, are targets of several viral oncoproteins, including SV40 large T-antigen (T-Ag) and adenovirus E1A. T-Ag and E1A each contains an LXCXE-peptide motif through which binding to the conserved 'A/B pocket' present in RB, p107 and p130 is achieved. The LXCXE-binding activity of RB, we have previously shown, is inhibited by phosphorylation at Thr 821 and 826 in the C-terminal region of RB. Thr 821 and its surrounding sequence is unique to RB and not found in p107 or p130. Interestingly, hyperphosphorylation of p107 does not disrupt its ability to bind T-Ag or to inhibit the transactivating function of E1A. Insertion of a fourteen amino acid sequence of RB containing Thr 821 into p107 prevents binding of T-Ag and E1A to phosphorylated p107. These results show that the RB sequence surrounding Thr 821 plays a critical role in the regulation of the 'A/B pocket' function. In addition, these data indicate that the protein binding functions of RB and p107 are not equivalently regulated by phosphorylation.


Assuntos
Antígenos Virais de Tumores/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas , Sequências Reguladoras de Ácido Nucleico , Proteína do Retinoblastoma/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Humanos , Dados de Sequência Molecular , Mutagênese Insercional , Proteínas Nucleares/genética , Fosforilação , Proteína do Retinoblastoma/genética , Proteína p107 Retinoblastoma-Like , Proteína p130 Retinoblastoma-Like , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas
11.
Oncogene ; 13(8): 1765-71, 1996 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-8895523

RESUMO

The immunosuppressant rapamycin has previously been shown to inhibit G1/S transition in several cell types. In Swiss-3T3 cells, rapamycin prolongs G1 through the inhibition of the S6-kinase. In T-lymphocytes, rapamycin blocks the mitogen-induced down regulation of p27Kip1, an inhibitor of the cdk/cyclin complexes. We show here that an NIH3T3 line (N-3T3) is also sensitive to the G1/S inhibitory effect of rapamycin. Unlike lymphocytes, rapamycin does not affect p27Kip1 in these immortalized fibroblasts, nor does rapamycin affect the activity of cyclin D- or cyclin E-dependent kinases. As a result, rapamycin does not inhibit the phosphorylation of the retinoblastoma protein (RB) or two RB-related proteins, p107 and p130. Despite the phosphorylation of RB/p107/ p130, the expression of cyclin A and its associated kinase activity is delayed in rapamycin-treated N-3T3 cells. Ectopic expression of cyclin A, but not cyclins D and E or E2F-1 and -4, can overcome the effect of rapamycin. Taken together, these results suggest that entry into S-phase is likely to involve rapamycin-sensitive pathways other than the phosphorylation of the pocket proteins.


Assuntos
Fase G1/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Polienos/farmacologia , Proteínas , Proteína do Retinoblastoma/metabolismo , Células 3T3 , Animais , Ciclinas/genética , Camundongos , Fosforilação , Proteína p107 Retinoblastoma-Like , Proteína p130 Retinoblastoma-Like , Fase S , Sirolimo
12.
Oncogene ; 19(15): 1857-67, 2000 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-10773875

RESUMO

Phosphorylation/inactivation of RB is typically required for cell cycle progression. However, we have identified a tumor cell line, C33A, which progresses through the cell cycle in the presence of an active allele of RB (PSM-RB). To determine how C33A cells evade RB-mediated arrest, we compared RB signaling to downstream effectors in this resistant cell line to that of the RB-sensitive SAOS-2 cell line. Although introduction of PSM-RB repressed E2F-mediated transcription in both C33A and SAOS-2 cells, PSM-RB failed to repress Cyclin A promoter activity in C33A. Ectopic expression of PSM-RB in SAOS-2 cells resulted in a decrease in both Cyclin A and Cdk2 protein levels without affecting Cyclin E or Cdk4. In contrast, over-expression of PSM-RB in C33A cells did not alter endogenous Cyclin A, Cyclin E, or Cdk2 protein levels or impact Cdk2 kinase activity, indicating that signaling from RB to down-stream targets is abrogated in this cell line. The importance of Cdk2 activity was demonstrated by p27Kip1, which attenuated Cdk2 activity and inhibited cell cycle progression in C33A cells. Since RB signaling to Cdk2 is disrupted in these tumor cells, we co-expressed two proteins that cooperate with RB in transcriptional repression, AHR and BRG-1, in an attempt to correct this signaling dysfunction. Co-expression of AHR/BRG-1 with PSM-RB attenuated Cyclin A and Cdk2 expression as well as Cdk2-associated kinase activity, resulting in cell cycle inhibition of C33A cells. Importantly, ectopic expression of Cyclin A was able to reverse the arrest mediated by co-expression of AHR/BRG-1 with PSM-RB. These results indicate that down-regulation of Cdk2 activity is requisite for RB-mediated cell cycle arrest. Thus, this study reveals a new mechanism through which tumor cells evade anti-proliferative signals, and provides insight into how RB-signaling is mediated.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Transporte , Proteínas de Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Proteínas de Ligação a DNA , Proteínas Serina-Treonina Quinases/metabolismo , Proteína do Retinoblastoma/metabolismo , Ciclo Celular , Ciclina A/genética , Quinase 2 Dependente de Ciclina , DNA Helicases , Regulação para Baixo , Fatores de Transcrição E2F , Humanos , Mutação , Proteínas Nucleares/farmacologia , Regiões Promotoras Genéticas , Receptores de Hidrocarboneto Arílico/metabolismo , Proteína 1 de Ligação ao Retinoblastoma , Transdução de Sinais , Fator de Transcrição DP1 , Fatores de Transcrição/metabolismo , Fatores de Transcrição/farmacologia , Células Tumorais Cultivadas
13.
Oncogene ; 18(37): 5239-45, 1999 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-10498874

RESUMO

Studies aimed at examining the precise function(s) of the retinoblastoma tumor suppressor protein, RB, have been hindered by the rapid phosphorylation and inactivation of ectopically expressed RB which occurs in the majority of cell types. Therefore, ectopically expressed RB is a poor inhibitor of cellular proliferation. We have designed constitutively active RB proteins, PSM-RB, that cannot be inactivated by phosphorylation. Using these proteins, we show that unlike wild-type RB, PSM-RB proteins inhibit cell cycle progression in a broad range of tumor cell types. Furthermore, unlike p16ink4a, PSM-RB is also a potent inhibitor of cell cycle progression in RB-deficient tumor cells. Surprisingly, we identified a tumor cell line that is resistant to the cell cycle inhibitory effects of PSM-RB. This finding challenges the hypothesis that RB must be inactivated in all cells for cell cycle progression to occur. Further characterization of this 'resistant' tumor line revealed that proliferation of these cells is still inhibited by PSM-RB. We show that this is due to PSM-RB-induced cell death. As such, these studies are the first to show that RB inhibits cellular proliferation through at least two distinct mechanisms - inhibition of cell cycle progression and induction of cell death.


Assuntos
Apoptose/fisiologia , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Proteína do Retinoblastoma/fisiologia , Apoptose/genética , Ciclo Celular/genética , Divisão Celular/genética , Genes do Retinoblastoma , Humanos , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/fisiologia , Neoplasias/genética , Neoplasias/patologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/fisiologia , Proteína do Retinoblastoma/deficiência , Transfecção , Células Tumorais Cultivadas
14.
Oncogene ; 14(5): 603-9, 1997 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-9053859

RESUMO

Deletions of chromosomal region 9p21 are among the most common genetic alterations observed during the clonal evolution of high grade malignant gliomas. Structural and functional evidence has suggested that homozygous deletion involving CDKN2A (the genetic locus encoding the cyclin-dependent kinase inhibitor p16(NK4a)) is a mechanism of inactivation of this gene and that it can be a growth suppressor in human gliomas. However, the presence of other potential suppressor genes in the 9p21 region and the relatively large sizes of the deletions has made it difficult to be certain that the CDKN2A gene is their actual target. Here, we tested this hypothesis by determining the growth suppressive effects, cell cycle inhibitions, and the activities of seven naturally occurring glioma-derived CDKN2A alleles carrying point mutations and found that two of them were functionally compromised. To resolve discrepancies among the different existing functional assays, we developed an assay for p16(INK4a) function that allowed us to demonstrate that the expression of wild-type CDKN2A, but not alleles with inactivating mutations, prevents pRB phosphorylation in vivo in human glioma cells. These data suggest that CDKN2A is a critical target for mutational inactivation in human malignant gliomas.


Assuntos
Proteínas de Transporte/metabolismo , Deleção Cromossômica , Cromossomos Humanos Par 9 , Glioma/enzimologia , Glioma/genética , Alelos , Proteínas de Transporte/genética , Ciclo Celular , Inibidor p16 de Quinase Dependente de Ciclina , Inibidores Enzimáticos/metabolismo , Genes Supressores de Tumor , Glioma/patologia , Humanos , Fosforilação , Biossíntese de Proteínas , Inibidores de Proteínas Quinases , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Proteína do Retinoblastoma/metabolismo , Transcrição Gênica , Transfecção
15.
Cell Prolif ; 30(3-4): 171-94, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9375029

RESUMO

A mathematical model integrating the roles of cyclin D, cdk4, cyclin E, cdk2, E2F and RB in control of the G1 phase of the cell cycle is described. Experimental results described with murine embryo fibroblasts (MEFs), either Rb+/+ or Rb-/-, and with the RB-deficient osteosarcoma cell line, Saos-2, served as the basis for the formulation of this mathematical model. A model employing the known interactions of these six proteins does not reproduce the experimental observations described in the MEFs. The appropriate modelling of G1 requires the inclusion of a sensing mechanism which adjusts the activity of cyclin E/cdk2 in response to both RB concentration and growth factors. Incorporation of this sensing mechanism into the model allows it to reproduce most of the experimental results observed in Saos-2 cells, Rb-/- MEFS, and Rb+/+ MEFs. The model also makes specific predictions which have not been tested experimentally.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Transporte , Proteínas de Ciclo Celular , Simulação por Computador , Proteínas de Ligação a DNA , Fase G1/fisiologia , Modelos Teóricos , Proteínas Proto-Oncogênicas , Proteína do Retinoblastoma/deficiência , Animais , Ciclina D , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Fatores de Transcrição E2F , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Camundongos , Camundongos Mutantes , Osteossarcoma , Proteínas Serina-Treonina Quinases/metabolismo , Proteína do Retinoblastoma/genética , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
16.
Oncogene ; 33(30): 3980-91, 2014 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-24121271

RESUMO

The processes that control the progression of ductal carcinoma in situ (DCIS) to invasive breast cancer remain poorly understood. Epidermal growth factor receptor 2 (ErbB2) overexpression is common in DCIS, as is disruption of the retinoblastoma tumor suppressor (RB) pathway. Here, we examined the cooperative impact of ErbB2 and RB deregulation on facets of disease progression. Our studies demonstrate that RB deficiency altered the expression of key molecules needed for proper cellular organization and epithelial cell-cell adhesion as part of a program related to the epithelial-to-mesenchymal transition (EMT). An increase in the invasive potential of ErbB2-overexpressing cells was observed upon RB depletion. Further, stable knockdown of RB resulted in invasive lesions in orthotopic xenograft assays, compared with DCIS-like lesions developing from RB-proficient cells. Conversely, the invasive phenotype observed in ErbB2-positive cancer models was inhibited through CDK4/6 inhibition in an RB-dependent manner. Finally, in a cohort of DCIS cases, we show that, although elevated levels of ErbB2 are associated with increased risk of a subsequent DCIS recurrence, it is not associated with progression to invasive disease. In contrast, RB loss in ErbB2-positive DCIS cases was associated with increased risk for invasive breast cancer. Taken together, these data demonstrate a key role for the RB pathway in invasion associated with breast tumor progression, and shed light on the key molecular events that promote the progression of DCIS to invasive disease.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma Intraductal não Infiltrante/metabolismo , Recidiva Local de Neoplasia/metabolismo , Receptor ErbB-2/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/patologia , Linhagem Celular Tumoral , Polaridade Celular , Proliferação de Células , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Transplante de Neoplasias , Proteína do Retinoblastoma/genética , Risco , Transcriptoma
18.
Oncogene ; 29(1): 68-80, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19802012

RESUMO

The retinoblastoma tumor suppressor, RB, is a key regulator of cellular proliferation that is functionally inactivated at high frequency in human cancer. Although RB has been extensively studied with regard to tumor etiology, loss of tumor-suppressor function often occurs relatively late in tumor progression. Therefore, inactivation of RB could have a profound impact on the behavior of tumors driven by discrete oncogenes. Here, collaboration between Ras or c-Myc deregulation and RB functional state was investigated in a model of conditional genetic deletion to decipher the effects related to disease progression. These studies showed that RB loss had a robust impact on mitogen dependence, anchorage dependence and overall survival, which was significantly modified by oncogene activation. Specifically, RB deficiency predisposed c-Myc-expressing cells to cell death and reduced overall tumorigenic proliferation. In contrast, RB deficiency exacerbated the tumorigenic behavior of Ras-transformed cells in both the model system and human tumor cell lines. As these tumors exhibited highly aggressive behavior, the possibility of exploiting the intrinsic sensitivity to cell death with RB loss was evaluated. Particularly, although Ras-transformed, RB-deficient cells bypassed the G1-checkpoint elicited by pharmacological activation of the p53 pathway, they were also highly sensitized to cell death. Altogether, these data suggest that the impact of RB deletion is dependent on the oncogene milieu, and can directly contribute to transformed phenotypes and response to therapeutic intervention.


Assuntos
Proliferação de Células , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína do Retinoblastoma/fisiologia , Proteínas ras/metabolismo , Células 3T3 , Animais , Apoptose , Linhagem Celular Tumoral , Células Cultivadas , Progressão da Doença , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Técnicas de Inativação de Genes , Humanos , Immunoblotting , Camundongos , Camundongos Knockout , Camundongos Nus , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Carga Tumoral , Proteínas ras/genética
19.
Oncogene ; 29(28): 4018-32, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20473330

RESUMO

A hallmark of cancer is the deregulation of cell-cycle machinery, ultimately facilitating aberrant proliferation that fuels tumorigenesis and disease progression. Particularly, in breast cancers, cyclin D1 has a crucial role in the development of disease. Recently, a highly specific inhibitor of CDK4/6 activity (PD-0332991) has been developed that may have efficacy in the treatment of breast cancer. To interrogate the utility of PD-0332991 in treating breast cancers, therapeutic response was evaluated on a panel of breast cancer cell lines. These analyses showed that the chronic loss of Rb is specifically associated with evolution to a CDK4/6-independent state and, ultimately, resistance to PD-0332991. However, to interrogate the functional consequence of Rb directly, knockdown experiments were performed in models that represent immortalized mammary epithelia and multiple subtypes of breast cancer. These studies showed a highly specific role for Rb in mediating the response to CDK4/6 inhibition that was dependent on transcriptional repression manifest through E2F, and the ability to attenuate CDK2 activity. Acquired resistance to PD-03322991 was specifically associated with attenuation of CDK2 inhibitors, indicating that redundancy in CDK functions represents a determinant of therapeutic failure. Despite these caveats, in specific models, PD-0332991 was a particularly effective therapy, which induced Rb-dependent cytostasis. Combined, these findings indicate the critical importance of fully understanding cell-cycle regulatory pathways in directing the utilization of CDK inhibitors in the clinic.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA