Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Cell Biol ; 137(4): 939-52, 1997 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-9151695

RESUMO

Carcinoembryonic antigen (CEA) is a tumor marker that is overexpressed in many human cancers and functions in vitro as a homotypic intercellular adhesion molecule. We have investigated the possibility of synergy between CEA, v-Myc, and Bcl-2 in the transformation of cells with differentiation capacity. We find that v-Myc increases the cell division rate and maximum density of rat L6 myoblasts but also markedly stimulates both apoptosis and surprisingly, differentiation, thus preventing transformation. The superposition of Bcl-2 blocks the apoptotic stimulation of v-Myc and independently promotes further cell division at confluence, but still allows differentiation. The further expression of CEA has a dominant effect in blocking differentiation, regardless of the presence of the other activated oncogenes, generating cells that enter a reversible quiescent G0-like state in medium promoting differentiation. Transfectants expressing CEA with or without v-myc and bcl-2 allow the emergence of cells with the property of heritable, efficient, anchorage-independent growth in soft agar and the ability to markedly reduce the latency for tumor formation in nude mice. We propose that by prolonging cell survival in the presence of differentiation signals, CEA represents a novel class of dominant differentiation-blocking oncogene.


Assuntos
Antígeno Carcinoembrionário/fisiologia , Transformação Celular Neoplásica , Proteína Oncogênica p55(v-myc)/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Animais , Apoptose , Diferenciação Celular , Divisão Celular , Genes bcl-2 , Genes myc , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Nus , Transplante de Neoplasias , Oncogenes , Ratos , Fase de Repouso do Ciclo Celular , Transfecção
2.
J Cell Biol ; 141(6): 1407-14, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9628896

RESUMO

OCI-5/GPC3 is a member of the glypican family. Glypicans are heparan sulfate proteoglycans that are bound to the cell surface through a glycosyl-phosphatidylinositol anchor. It has recently been shown that the OCI-5/GPC3 gene is mutated in patients with the Simpson-Golabi-Behmel Syndrome (SGBS), an X-linked disorder characterized by pre- and postnatal overgrowth and various visceral and skeletal dysmorphisms. Some of these dysmorphisms could be the result of deficient growth inhibition or apoptosis in certain cell types during development. Here we present evidence indicating that OCI-5/GPC3 induces apoptosis in cell lines derived from mesothelioma (II14) and breast cancer (MCF-7). This induction, however, is cell line specific since it is not observed in NIH 3T3 fibroblasts or HT-29 colorectal tumor cells. We also show that the apoptosis-inducing activity in II14 and MCF-7 cells requires the anchoring of OCI-5/GPC3 to the cell membrane. The glycosaminoglycan chains, on the other hand, are not required. MCF-7 cells can be rescued from OCI-5/GPC3-induced cell death by insulin-like growth factor 2. This factor has been implicated in Beckwith-Wiedemann, an overgrowth syndrome that has many similarities with SGBS. The discovery that OCI-5/GPC3 is able to induce apoptosis in a cell line- specific manner provides an insight into the mechanism that, at least in part, is responsible for the phenotype of SGBS patients.


Assuntos
Apoptose , Glipicanas/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Proteínas de Membrana/metabolismo , Células 3T3 , Animais , Células COS , Expressão Gênica , Glicosilfosfatidilinositóis/metabolismo , Glipicanas/genética , Transtornos do Crescimento , Células HT29 , Proteoglicanas de Heparan Sulfato/genética , Humanos , Fator de Crescimento Insulin-Like II/farmacologia , Proteínas de Membrana/genética , Mesotelioma , Camundongos , Ratos , Síndrome , Células Tumorais Cultivadas
3.
Mol Cell Biol ; 21(14): 4725-36, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11416148

RESUMO

The ability of the c-Myc oncoprotein to potentiate apoptosis has been well documented; however, the mechanism of action remains ill defined. We have previously identified spatially distinct apoptotic pathways within the same cell that are differentially inhibited by Bcl-2 targeted to either the mitochondria (Bcl-acta) or the endoplasmic reticulum (Bcl-cb5). We show here that in Rat1 cells expressing an exogenous c-myc allele, distinct apoptotic pathways can be inhibited by Bcl-2 or Bcl-acta yet be distinguished by their sensitivity to Bcl-cb5 as either susceptible (serum withdrawal, taxol, and ceramide) or refractory (etoposide and doxorubicin). Myc expression and apoptosis were universally associated with Bcl-acta and not Bcl-cb5, suggesting that Myc acts downstream at a point common to these distinct apoptotic signaling cascades. Analysis of Rat1 c-myc null cells shows these same death stimuli induce apoptosis with characteristic features of nuclear condensation, membrane blebbing, poly (ADP-ribose) polymerase cleavage, and DNA fragmentation; however, this Myc-independent apoptosis is not inhibited by Bcl-2. During apoptosis, Bax translocation to the mitochondria occurs in the presence or absence of Myc expression. Moreover, Bax mRNA and protein expression remain unchanged in the presence or absence of Myc. However, in the absence of Myc, Bax is not activated and cytochrome c is not released into the cytoplasm. Reintroduction of Myc into the c-myc null cells restores Bax activation, cytochrome c release, and inhibition of apoptosis by Bcl-2. These results demonstrate a role for Myc in the regulation of Bax activation during apoptosis. Moreover, apoptosis that can be triggered in the absence of Myc provides evidence that signaling pathways exist which circumvent Bax activation and cytochrome c release to trigger caspase activation. Thus, Myc increases the cellular competence to die by enhancing disparate apoptotic signals at a common mitochondrial amplification step involving Bax activation and cytochrome c release.


Assuntos
Apoptose , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Membrana Celular/metabolismo , Grupo dos Citocromos c/metabolismo , Ratos , Proteína X Associada a bcl-2
4.
Mol Cell Biol ; 17(1): 100-14, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8972190

RESUMO

Increasing evidence supports an important biological role for Myc in the downregulation of specific gene transcription. Recent studies suggest that c-Myc may suppress promoter activity through proteins of the basal transcription machinery. We have previously reported that Myc protein, in combination with additional cellular factors, suppresses transcription initiation from the c-myc promoter. To characterize the cis components of this Myc negative autoregulation pathway, fragments of the human c-myc promoter were inserted upstream of luciferase reporter genes and assayed for responsiveness to inducible MycER activation in Rat-1 fibroblasts. We found four- to fivefold suppression of a c-myc P2 minimal promoter fragment upon induction of wild-type MycER protein activity, while induction of a mutant MycER protein lacking amino acids 106 to 143 required for Myc autosuppression failed to elicit this response. This assay is physiologically significant, as it reflects Myc autosuppression of the endogenous c-myc gene with regard to kinetics, dose dependency, cell type specificity, and c-Myc functional domains. Analysis of mutations within the P2 minimal promoter indicated that the cis components of Myc autosuppression could not be ascribed to any known protein-binding motifs. In addition, to address the trans factors required for Myc negative autoregulation, we expressed MycEG and MaxEG leucine zipper dimerization mutants in Rat-1 cells and found that Myc-Max heterodimerization is obligatory for Myc autosuppression. Two models for the Myc autosuppression mechanism are discussed.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Genes myc/genética , Homeostase/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição , Células 3T3 , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fatores de Transcrição de Zíper de Leucina Básica , Linhagem Celular , Dimerização , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter/genética , Humanos , Luciferases/genética , Camundongos , Proteínas Proto-Oncogênicas c-myc/genética , Ratos , Receptores de Estrogênio/genética , Proteínas Recombinantes de Fusão , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Transcrição Gênica/fisiologia
5.
Mol Cell Biol ; 20(18): 6768-78, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10958674

RESUMO

Platelet-derived growth factor BB (PDGF BB) is a potent mitogen for fibroblasts as well as many other cell types. Interaction of PDGF BB with the PDGF beta receptor (PDGF-betaR) activates numerous signaling pathways and leads to a decrease in receptor expression on the cell surface. PDGF-betaR downregulation is effected at two levels, the immediate internalization of ligand-receptor complexes and the reduction in pdgf-betar mRNA expression. Our studies show that pdgf-betar mRNA suppression is regulated by the c-myc proto-oncogene. Both constitutive and inducible ectopic Myc protein can suppress pdgf-betar mRNA and protein. Suppression of pdgf-betar mRNA in response to Myc is specific, since expression of the related receptor pdgf-alphar is not affected. We further show that Myc suppresses pdgf-betar mRNA expression by a mechanism which is distinguishable from Myc autosuppression. Analysis of c-Myc-null fibroblasts demonstrates that Myc is required for the repression of pdgf-betar mRNA expression in quiescent fibroblasts following mitogen stimulation. In addition, it is evident that the Myc-mediated repression of pdgf-betar mRNA levels plays an important role in the regulation of basal pdgf-betar expression in proliferating cells. Thus, our studies suggest an essential role for Myc in a negative-feedback loop regulating the expression of the PDGF-betaR.


Assuntos
Regulação para Baixo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Repressoras/metabolismo , Células 3T3 , Animais , Becaplermina , Transformação Celular Neoplásica , Células Cultivadas , Cinética , Camundongos , Mitógenos/farmacologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-sis , RNA Mensageiro , Ratos , Transcrição Gênica
6.
Oncogene ; 36(49): 6815-6822, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-28806398

RESUMO

The oncogenic transcription factor c-MYC (MYC) is deregulated and often overexpressed in more than 50% of cancers. MYC deregulation is associated with poor prognosis and aggressive disease, suggesting that the development of therapeutic inhibitors targeting MYC would markedly impact patient outcome. MYC is highly regulated, with a protein and mRNA half-life of ~30 min. The most extensively studied pathway regulating MYC protein stability involves ubiquitylation and proteasomal degradation mediated by the E3-ligase, SCFFbxw7. Here we provide evidence for an SCFFbxw7-independent regulatory mechanism centred on the highly conserved lysine-52 (K52) within MYC Box I. This residue has been shown to be post-translationally modified by both ubiquitylation and SUMOylation, hinting at the interplay of post-translational modifications at this site and the importance of this residue. We demonstrate that mutation of K52 to arginine (R) renders the MYC protein more labile. Mechanistically, we show that the degradation pathway regulated by K52 is independent of the Cullin-RING ligase family of E3-ligases, which includes not only the canonical SCFFbxw7 but also other known MYC-targeting E3-ligases, such as SCFSkp2, SCFßTCRP, SCFFbxo28 and DCXTRUSS. Taken together, our data identify a novel regulatory pathway centred on K52 that may be exploited for the development of anti-MYC therapeutics.


Assuntos
Proteína 7 com Repetições F-Box-WD/metabolismo , Lisina/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ubiquitinação , Animais , Arginina/genética , Arginina/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proteína Duplacortina , Células HCT116 , Células HEK293 , Meia-Vida , Humanos , Immunoblotting , Lisina/genética , Mutação de Sentido Incorreto , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Ratos
7.
Oncogene ; 36(49): 6830-6837, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-28806396

RESUMO

Developing therapeutics to effectively inhibit the MYC oncoprotein would mark a key advance towards cancer patient care as MYC is deregulated in over 50% of human cancers. MYC deregulation is correlated with aggressive disease and poor patient outcome. Despite strong evidence in mouse models that inhibiting MYC would significantly impact tumour cell growth and patient survival, traditional approaches have not yet yielded the urgently needed therapeutic agents that directly target MYC. MYC functions through its interaction with MAX to regulate gene transcription by binding to E-box DNA response elements of MYC target genes. Here we used a structure-based strategy to design ME47, a small minimalist hybrid protein (MHP) able to disrupt the MAX:E-box interaction/binding and block transcriptional MYC activity. We show that inducing ME47 expression in established tumour xenografts inhibits tumour growth and decreases cellular proliferation. Mechanistically, we show by chromatin immunoprecipitation that ME47 binds to E-box binding sites of MYC target genes. Moreover, ME47 occupancy decreases MYC:DNA interaction at its cognate E-box binding sites. Taken together, ME47 is a prototypic MHP inhibitor that antagonizes tumour cell growth in vitro and in vivo and inhibits the interaction of MYC with DNA E-box elements. These results support ME47's role as a MYC inhibitor and suggest that MHPs provide an alternative therapeutic targeting system that can be used to target transcription factors important in human diseases, including cancer.


Assuntos
Elementos E-Box/genética , Motivos de Nucleotídeos/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Ligação Competitiva , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Células HEK293 , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Recombinantes de Fusão/genética , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Carga Tumoral/genética
8.
Cancer Res ; 55(9): 1982-8, 1995 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-7728769

RESUMO

Apoptosis can be regulated in a number of different systems by the actions of cytokines. Rapamycin has been shown to exert its effects on growth factor-induced cell proliferation, at least in part, by blocking the activation of the p70 S6 kinase and thus preventing the downstream signaling process, such as the activation of the members of the cdk family. To determine whether this pathway plays a role in the regulation of apoptosis, we assessed the effect of rapamycin on apoptosis induced by interleukin 2 deprivation in murine T-cell lines, by T-cell receptor ligation in a murine T-cell hybridoma, by enforced c-myc expression in murine fibroblasts, and by corticosteroids in murine T-lymphoma cell lines. Although rapamycin did not induce apoptosis on its own, rapamycin augmented apoptosis in each of the cell lines used as indicated by increased genomic DNA fragmentation, decreased cell viability, and characteristic apoptotic changes in morphology. These results suggest that a signal transduction pathway(s) inhibited by rapamycin plays an important role in the susceptibility of cells to apoptosis. Many chemotherapeutic agents kill cancer cells through the induction of apoptosis. Strikingly, rapamycin increased the ability of the alkylating agent, cisplatin, to induce apoptosis in the human promyelocytic leukemia cell line HL-60 and the human ovarian cancer cell line SKOV3. These data suggest that a signal transduction pathway, likely related to p70 S6 kinase, inhibited by rapamycin may be an important component of the pathway which prevents cell death in many cell lineages and also indicate that rapamycin has the potential to augment the efficacy of selected anticancer therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Polienos/farmacologia , Animais , Cisplatino/administração & dosagem , Cricetinae , Sinergismo Farmacológico , Feminino , Humanos , Imunossupressores/administração & dosagem , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/patologia , Ativação Linfocitária , Camundongos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Polienos/administração & dosagem , Sensibilidade e Especificidade , Sirolimo , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos
9.
Oncogene ; 17(17): 2149-54, 1998 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-9811446

RESUMO

The growth arrest and DNA damage inducible (gadd) genes are induced by various genotoxic and non-genotoxic stresses such as serum starvation, ultraviolet irradiation and treatment with alkylating agents. Their coordinate induction is a growth arrest signal which may play an important role in the response of cells to DNA damage. Conversely, c-myc is a strong proliferative signal, and overexpression of Myc is frequently observed in cancer cells. We have found that ectopic expression of v-myc in RAT-1 cells results in an attenuated induction of the three major gadd transcripts by methyl methanesulfonate (MMS), and almost completely blocks the response to ultraviolet (UV) radiation. Myc acts in part by reducing the stress-responsiveness of the gadd45 promoter, as a c-myc expression vector strongly suppressed activation of gadd45-reporter constructs. This activity of Myc localizes to a recently described GC-rich binding site within the gadd45 promoter. These results indicate that a coordinate down-regulation of the gadd gene response is one mechanism by which Myc can circumvent growth arrest and contribute to the neoplastic phenotype.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT , Proteínas de Ligação a DNA/biossíntese , Genes myc/fisiologia , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/biossíntese , Animais , Sequência de Bases , Dano ao DNA , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/efeitos da radiação , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular , Metanossulfonato de Metila/toxicidade , Dados de Sequência Molecular , Mutagênicos/toxicidade , Proteínas/efeitos dos fármacos , Proteínas/genética , Proteínas/efeitos da radiação , Ratos , Fator de Transcrição CHOP , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/efeitos da radiação , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos da radiação , Raios Ultravioleta , Regulação para Cima , Proteínas GADD45
10.
Oncogene ; 20(16): 1939-52, 2001 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-11360178

RESUMO

The disruption of mitochondrial function is a key component of apoptosis in most cell types. Localization of Bcl-2 to the outer mitochondrial and endoplasmic reticulum membranes is consistent with a role in the inhibition of many forms of apoptosis. In Rat-1 cells, a Bcl-2 mutant targeted exclusively to the endoplasmic reticulum (Bcl-cb5) was effective at inhibiting apoptosis induced by serum starvation/myc, or ceramide but not apoptosis induced by etoposide. The former conditions cause a decrease in mitochondrial transmembrane potential (Deltapsi(m)) as an early event that precedes the release of cytochrome c from mitochondria. By contrast, when cells are exposed to etoposide, a situation in which cytochrome c release and membrane localization of the pro-apoptotic protein Bax precede loss of Deltapsi(m), wild type Bcl-2 but not Bcl-cb5 prevents apoptosis. Therefore, Bcl-2 functions in spatially distinct pathways of apoptosis distinguished by the order of cytochrome c release and loss of Deltapsi(m).


Assuntos
Apoptose/fisiologia , Grupo dos Citocromos c/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Ceramidas/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Ativação Enzimática , Etoposídeo/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Genes myc/fisiologia , Membranas Intracelulares/metabolismo , Potenciais da Membrana/fisiologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Conformação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Estaurosporina/farmacologia , Proteína X Associada a bcl-2
11.
Oncogene ; 14(23): 2825-34, 1997 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-9190899

RESUMO

The c-Myc protein strongly stimulates cellular proliferation, inducing cells to exit G0/G1 and enter the cell cycle. At a molecular level, Myc prevents growth arrest and drives cell cycle progression through the transcriptional regulation of Myc-target genes. Expression of the growth arrest and DNA damage inducible gene 45 (gadd45) is elevated in response to DNA damaging agents, such as ionizing radiation via a p53-dependent mechanism, upon nutrient deprivation, or during differentiation. Gadd45 holds a vital role in growth arrest as ectopic expression confers a strong block to proliferation. Exposure of quiescent cells to mitogen stimulates a rapid increase in c-Myc expression which is followed by the subsequent reduction in gadd45 expression. The kinetics of these two regulatory events suggest that Myc suppresses the expression of gadd45, contributing to G0/G1 phase exit of the cell cycle. Indeed, ectopic Myc expression in primary and immortalized fibroblasts results in the suppression of gadd45 mRNA levels, by a mechanism which is independent of cell cycle progression. Using an inducible MycER system, rapid suppression of gadd45 mRNA is first evident approximately 0.5 h following Myc activation. The reduction in gadd45 mRNA expression occurs at the transcriptional level and is mediated by a p53-independent pathway. Moreover, Myc suppression and p53 induction of gadd45 following exposure to ionizing radiation are non-competitive co-regulatory events. Myc suppression of gadd45 defines a novel pathway through which Myc promotes cell cycle entry and prevents growth arrest of transformed cells.


Assuntos
Dano ao DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/farmacologia , Animais , Sítios de Ligação , Sangue , Células Cultivadas , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Camundongos , Modelos Moleculares , Regiões Promotoras Genéticas , Proteínas/genética , RNA Mensageiro/metabolismo , Ratos , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Proteínas GADD45
12.
Oncogene ; 12(1): 43-52, 1996 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-8552398

RESUMO

Cyclin D1 can bind and phosphorylate the product (pRb) of the retinoblastoma gene (RB-1) and recent evidence suggests pRb, in turn, may regulate cyclin D1 protein expression. In transformed cell lines, loss of pRb activity strongly correlates with a decrease in cyclin D1 protein expression, and conversely, introduction of pRb can induce cyclin D1 promoter activity. We show here that pRb does not regulate cyclin D1 directly as basal and serum-stimulated levels of cyclin D1 protein and kinase activity are similar in wildtype and pRb-deficient primary mouse embryonic fibroblasts (MEFs). These observations suggest that the suppression of cyclin D1 in pRb-minus tumour cell lines requires both loss of pRb and at least one additional genetic event. We have determined that constitutive, ectopic Myc expression in pRb-deficient, but not wildtype, MEFs suppresses cyclin D1 protein expression and kinase activity. Regulation is evident at either the level of RNA or protein expression. Phenotypically, pRb-deficient MEFs consistently exhibited a delayed growth response in comparison to wildtype MEFs. This growth delay is abrogated in pRb-deficient MEFs which are expressing ectopic Myc protein, coincident with the loss of cyclin D1 protein expression. Moreover, these cells exhibit an increased proliferative capacity, and they no longer show contact inhibition. Our results support a cross-regulatory mechanism between Myc, pRb and cyclin D1 and suggest a novel role for cyclin D1 in tumorigenesis.


Assuntos
Transformação Celular Neoplásica , Ciclinas/fisiologia , Proteínas Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Proteína do Retinoblastoma/fisiologia , Animais , Sequência de Bases , Células Cultivadas , Ciclina D1 , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/análise , Proteína do Retinoblastoma/deficiência
13.
Oncogene ; 18(23): 3520-8, 1999 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-10376530

RESUMO

Bcl-2 is a key inhibitor of a broad range of apoptotic pathways, yet neither the mechanism of action nor the role of Bcl-2 subcellular localization are well understood. The subcellular localization of Bcl-2 includes the mitochondrial membrane as well as the contiguous membrane of the endoplasmic reticulum and nuclear envelope. Most studies suggest that the ability of Bcl-2 to confer cell survival is dependent upon its localization to the mitochondria. In this manuscript, we show that Bcl-2 targeted to the endoplasmic reticulum can inhibit Myc-, but not etoposide-induced apoptosis in the Rat-1 fibroblast cell line. By contrast, wild type Bcl-2 can inhibit apoptosis triggered by either death agonist. We further show both Myc and etoposide trigger disruption of mitochondrial membrane potential (MMP) and induce poly-ADP ribose polymerase (PARP) cleavage, but release of calcium was not evident. Bcl-2 abrogates apoptosis at or upstream of MMP depletion showing that Bcl-2 does not have to reside at the mitochondria to prevent apoptosis. These results further elucidate the biochemical events associated with Myc- and etoposide-induced apoptosis and significantly advance our understanding of Bcl-2 function.


Assuntos
Apoptose/fisiologia , Retículo Endoplasmático/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Caspases/fisiologia , Linhagem Celular , Ativação Enzimática , Etoposídeo/farmacologia , Membranas Intracelulares/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mitocôndrias/fisiologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Ratos
14.
Cell Death Differ ; 7(8): 697-705, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10918443

RESUMO

c-myc nullizygous fibroblasts (KO cells) were used to compare the abilities of c-myc, N-myc and L-myc oncoproteins to accelerate growth, promote apoptosis, revert morphology, and regulate the expression of previously described c-myc target genes. All three myc oncoproteins were expressed following retroviral transduction of KO cells. The proteins all enhanced the growth rate of KO cells and significantly shortened the cell cycle transition time. They also accelerated apoptosis following serum deprivation, reverted the abnormal KO cell morphology, and modulated the expression of previously described c-myc target genes. In most cases, L-myc was equivalent to c-myc and N-myc in restoring all of the c-myc-dependent activities. These findings contrast with the previously reported weak transforming and transactivating properties of L-myc. Myc oncoproteins may thus impart both highly similar as well as dissimilar signals to the cells in which they are expressed.


Assuntos
Apoptose , Proteínas Proto-Oncogênicas c-myc/fisiologia , Animais , Divisão Celular , Linhagem Celular , Fibroblastos/citologia , Regulação da Expressão Gênica , Vetores Genéticos , Proteínas Proto-Oncogênicas c-myc/genética , Ratos , Retroviridae , Transformação Genética
15.
Leukemia ; 15(9): 1398-407, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11516100

RESUMO

Lovastatin is an inhibitor of the enzyme 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, the major regulatory enzyme of the mevalonate pathway. We have previously reported that lovastatin induces a significant apoptotic response in human acute myeloid leukemia (AML) cells. To identify the critical biochemical mechanism(s) essential for lovastatin-induced apoptosis, add-back experiments were conducted to determine which downstream product(s) of the mevalonate pathway could suppress this apoptotic response. Apoptosis induced by lovastatin was abrogated by mevalonate (MVA) and geranylgeranyl pyrophosphate (GGPP), and was partially inhibited by farnesyl pyrophosphate (FPP). Other products of the mevalonate pathway including cholesterol, squalene, lanosterol, desmosterol, dolichol, dolichol phosphate, ubiquinone, and isopentenyladenine did not affect lovastatin-induced apoptosis in AML cells. Our results suggest that inhibiting geranylgeranylation of target proteins is the predominant mechanism of lovastatin-induced apoptosis in AML cells. In support of this hypothesis, the geranylgeranyl transferase inhibitor (GGTI-298) mimicked the effect of lovastatin, whereas the farnesyl transferase inhibitor (FTI-277) was much less effective at triggering apoptosis in AML cells. Inhibition of geranylgeranylation was monitored and associated with the apoptotic response induced by lovastatin and GGTI-298 in the AML cells. We conclude that blockage of the mevalonate pathway, particularly inhibition of protein geranylgeranylation holds a critical role in the mechanism of lovastatin-induced apoptosis in AML cells.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Leucemia Mieloide/patologia , Lovastatina/farmacologia , Prenilação de Proteína/efeitos dos fármacos , Doença Aguda , Benzamidas/farmacologia , Nucleotídeos de Desoxiuracil/metabolismo , Fluoresceína-5-Isotiocianato , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Metionina/análogos & derivados , Metionina/farmacologia , Ácido Mevalônico/farmacologia , Fosfatos de Poli-Isoprenil/farmacologia , Sesquiterpenos , Células Tumorais Cultivadas
16.
Leukemia ; 16(4): 508-19, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11960327

RESUMO

The statin family of drugs target HMG-CoA reductase, the rate-limiting enzyme of the mevalonate pathway, and have been used successfully in the treatment of hypercholesterolemia for the past 15 years. Experimental evidence suggests this key biochemical pathway holds an important role in the carcinogenic process. Moreover, statin administration in vivo can provide an oncoprotective effect. Indeed, in vitro studies have shown the statins can trigger cells of certain tumor types, such as acute myelogenous leukemia, to undergo apoptosis in a sensitive and specific manner. Mechanistic studies show bcl-2 expression is down-regulated in transformed cells undergoing apoptosis in response to statin exposure. In addition, the apoptotic response is in part due to the depletion of the downstream product geranylgeranyl pyrophosphate, but not farnesyl pyrophosphate or other products of the mevalonate pathway including cholesterol. Clinically, preliminary phase I clinical trials have shown the achievable plasma concentration corresponds to the dose range that can trigger apoptosis of tumor types in vitro. Moreover, little toxicity was evident in vivo even at high concentrations. Clearly, additional clinical trials are warranted to further assess the safety and efficacy of statins as novel and immediately available anti-cancer agents. In this article, the experimental evidence supporting a role for the statin family of drugs to this new application will be reviewed.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Neoplasias/tratamento farmacológico , Divisão Celular/efeitos dos fármacos , Humanos , Neoplasias/patologia
17.
Clin Cancer Res ; 7(7): 2067-75, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11448925

RESUMO

The statin family of drugs inhibits 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, the rate-limiting enzyme of the mevalonate pathway, and is used clinically as a safe and effective approach in the control of hypercholesterolemia. We have shown previously (Dimitroulakos, J., Nohynek, D., Backway, K. L., Hedley, D. W., Yeger, H., Freedman, M. H., Minden, M D., and Penn, L. Z. Increased sensitivity of acute myelogenous leukemias to lovastatin-induced apoptosis: a potential therapeutic approach. Blood, 93: 1308-1318, 1999) that lovastatin, a prototypic member of the statin family, can induce apoptosis of human acute myeloid leukemia (AML) cells in a sensitive and specific manner. In the present study, we evaluated the relative potency and mechanism of action of the newer synthetic statins, fluvastatin, atorvastatin, and cerivastatin, to trigger tumor-specific apoptosis. Cerivastatin is at least 10 times more potent than the other statins at inducing apoptosis in AML cell lines. Cerivastatin-induced apoptosis is reversible with the addition of the immediate product of the HMG-CoA reductase reaction, mevalonate, or with a distal product of the pathway, geranylgeranyl pyrophosphate. This suggests protein geranylgeranylation is an essential downstream component of the mevalonate pathway for cerivastatin similar to lovastatin-induced apoptosis. The enhanced potency of cerivastatin expands the number of AML patient samples as well as the types of malignancies, which respond to statin-induced apoptosis with acute sensitivity. Cells derived from acute lymphocytic leukemia are only weakly sensitive to lovastatin cytotoxicity but show robust response to cerivastatin. Importantly, cerivastatin is not cytotoxic to nontransformed human bone marrow progenitors. These results strongly support the further testing of cerivastatin as a novel anticancer therapeutic alone and in combination with other agents in vivo.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Neoplasias/tratamento farmacológico , Piridinas/farmacologia , Doença Aguda , Atorvastatina , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ácidos Graxos Monoinsaturados/farmacologia , Fluvastatina , Ácidos Heptanoicos/farmacologia , Humanos , Indóis/farmacologia , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/patologia , Neoplasias/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Pirróis/farmacologia , Sensibilidade e Especificidade , Células Tumorais Cultivadas
18.
Clin Cancer Res ; 7(1): 158-67, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11205904

RESUMO

3-Hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase is the rate-limiting enzyme of the mevalonate pathway, the diverse array of end products of which are vital for a variety of cellular functions, including cholesterol synthesis and cell cycle progression. We showed previously that this enzyme holds a critical role in regulating tumor cell fate, including cell death, as its expression is down-regulated in response to retinoic acid, a potent anticancer therapeutic. Indeed, direct inhibition of HMG-CoA reductase with lovastatin, a competitive inhibitor of this enzyme, induced a pronounced apoptotic response in neuroblastoma and acute myeloid leukemic cells. We have now extended this work and evaluated a wide variety and large number of tumor-derived cell lines for their sensitivity to lovastatin-induced apoptosis. These cell lines were exposed to a wide range (0-100 microM) of lovastatin for 2 days and assayed for cell viability using the 3,4,5-dimethyl thiazlyl-2,2,5-diphenyltetrazolium bromide assay and the induction of apoptosis by flow cytometric and ultrastructural analyses. Lovastatin induced a pronounced apoptotic response in cells derived from juvenile monomyelocytic leukemia, pediatric solid malignancies (rhabdomyosarcoma and medulloblastoma), and squamous cell carcinoma of the cervix and of the head and neck. Interestingly, the subset of malignancies that are particularly sensitive to lovastatin-induced apoptosis correspond to those tumor subtypes that are sensitive to the biological and antiproliferative effects of retinoids in vitro. The nature of the biologically active form of lovastatin has been challenged recently as the growth-inhibitory effects of this drug were attributed to its prodrug lactone form that does not inhibit HMG-CoA reductase function. In this report, we demonstrate that the apoptotic properties of lovastatin are triggered by the open ring acid form that is a potent inhibitor of HMG-CoA reductase activity. Thus, we have identified a subset of tumors that are sensitive to lovastatin-induced apoptosis and show HMG-CoA reductase as a potential therapeutic target of these cancers.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Neoplasias/tratamento farmacológico , Adulto , Criança , Cromatografia Líquida de Alta Pressão , Feminino , Citometria de Fluxo , Formazans , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Espectrometria de Massas , Ácido Mevalônico/farmacologia , Microscopia Eletrônica , Sais de Tetrazólio , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos
19.
Curr Opin Investig Drugs ; 2(5): 684-92, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11569948

RESUMO

In the past ten years a wealth of fundamental knowledge delineating the molecular mechanism(s) of apoptosis has emerged, and can now be exploited to identify novel apoptotic modulators for the treatment of cancer. Two distinct yet complimentary classes of non-genotoxic agonists that can selectively kill tumor cells are discussed; agents that target 'classical' and 'atypical' apoptotic signaling pathways. The goal of agents targeting classical apoptosis and survival pathways is to directly modulate key apoptotic regulators such as Bcl-2, Akt/PKB, and p53. The aim of agents targeting atypical apoptotic pathways is to target signaling cascades whose inhibition remains non-lethal in normal cells, yet is suicidal in tumor cells. Such compounds presently under development include inhibitors of heat shock protein 90, histone deacetylases and HMG-CoA reductase. Both classes of apoptotic modulators have merit and identification of additional agonists of this nature will provide the many diverse cytotoxic agents that are required to combat the many diseases we call cancer.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Animais , Antineoplásicos/uso terapêutico , Humanos , Neoplasias/genética , Transdução de Sinais/efeitos dos fármacos
20.
Leuk Lymphoma ; 40(5-6): 659-62, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11426537

RESUMO

We recently reported that AML cells derived either from cell lines or from patients undergo apoptosis in response to lovastatin, an agent used extensively in the treatment of hypercholesterolemia. The concentration of lovastatin required to achieve this in culture varies from patient to patient, however, the in vitro concentrations required to kill AML cells, can be attained clinically. While in vitro studies assessing responsiveness of leukemic cells to lovastatin were being performed, a 72 year old female presented with relapsed AML. The patient did not desire any further induction therapy. As the patient's cells proved to be sensitive in culture to lovastatin, the patient was offered this drug. In this brief report we describe a case in which there was apparent control of the patient's leukemic blast cells by lovastatin at a dose double the usual recommended dose for hypercholesterolemia. This case illustrates the potential for lovastatin to provide a novel means of controlling leukemic cell growth in AML patients.


Assuntos
Anticolesterolemiantes/administração & dosagem , Antineoplásicos/administração & dosagem , Leucemia Mieloide Aguda/tratamento farmacológico , Lovastatina/administração & dosagem , Idoso , Morte Celular/efeitos dos fármacos , Feminino , Humanos , Leucemia Mieloide Aguda/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA