Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 6: e1634, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25675294

RESUMO

The p53 family and its cofactors are potent inducers of apoptosis and form a barrier to cancer. Here, we investigated the impact of the supposedly inhibitory member of the apoptosis-stimulating protein of p53, iASPP, on the activity of the p53 homolog TAp73, and its cofactors p300 and CBP. We found that iASPP interacted with and stabilized the histone acetyltransferase p300 and its homolog CBP upon cisplatin treatment. Vice versa, iASPP depletion by shRNA resulted in decreased amounts of p300 and CBP, impaired binding of p300 and TAp73 to target site promoters, reduced induction of pro-apoptotic TAp73 target genes, and impaired apoptosis. Mechanistically, we observed that the p300-regulatory E3 ubiquitin ligase BRMS1 could rescue the degradation of p300 and CBP in cisplatin-treated, iASPP-depleted cells. This argues that iASPP stabilizes p300 and CBP by interfering with their BRMS1-mediated ubiquitination, thereby contributing to apoptotic susceptibility. In line, iASPP overexpression partially abolished the interaction of BRMS1 and CBP upon DNA damage. Reduced levels of iASPP mRNA and protein as well as CBP protein were observed in human melanoma compared with normal skin tissue and benign melanocytic nevi. In line with our findings, iASPP overexpression or knockdown of BRMS1 each augmented p300/CBP levels in melanoma cell lines, thereby enhancing apoptosis upon DNA damage. Taken together, destabilization of p300/CBP by downregulation of iASPP expression levels appears to represent a molecular mechanism that contributes to chemoresistance in melanoma cells.


Assuntos
Proteína de Ligação a CREB/metabolismo , Proteína p300 Associada a E1A/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Repressoras/metabolismo , Apoptose , Proteína de Ligação a CREB/genética , Ciclo Celular , Imunoprecipitação da Cromatina , Proteína p300 Associada a E1A/genética , Células HEK293 , Humanos , Imunoprecipitação , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/genética , Melanoma/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Repressoras/genética
2.
Neuropathol Appl Neurobiol ; 40(2): 205-16, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24117486

RESUMO

AIMS: The Far Upstream Element [FUSE] Binding Protein 1 (FUBP1) regulates target genes, such as the cell cycle regulators MYC and p21. FUBP1 is up-regulated in many tumours and acts as an oncoprotein by stimulating proliferation and inhibiting apoptosis. Recently, FUBP1 mutations were identified in approximately 15% of oligodendrogliomas. To date, all reported FUBP1 mutations have been predicted to inactivate FUBP1, which suggests that in contrast to most other tumours FUBP1 may act as a tumour suppressor in oligodendrogliomas. METHODS: As no data are currently available concerning FUBP1 protein levels in gliomas, we examined the FUBP1 expression profiles of human glial tumours by immunohistochemistry and immunofluorescence. We analysed FUBP1 expression related to morphological differentiation, IDH1 and FUBP1 mutation status, 1p/19q loss of heterozygosity (LOH) as well as proliferation rate. RESULTS: Our findings demonstrate that FUBP1 expression levels are increased in all glioma subtypes as compared with normal central nervous system (CNS) control tissue and are associated with increased proliferation. In contrast, FUBP1 immunonegativity predicted FUBP1 mutation with a sensitivity of 100% and a specificity of 90% in our cohort and was associated with oligodendroglial differentiation, IDH1 mutation and 1p/19q loss of heterozygosity (LOH). Using this approach, we detected a to-date undescribed FUBP1 mutation in an oligodendroglioma. CONCLUSION: In summary, our data indicate an association between of FUBP1 expression and proliferation in gliomas. Furthermore, our findings present FUBP1 immunohistochemical analysis as a helpful additional tool for neuropathological glioma diagnostics predicting FUBP1 mutation.


Assuntos
DNA Helicases/genética , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Diferenciação Celular , Proliferação de Células , Deleção Cromossômica , Cromossomos Humanos Par 1 , Cromossomos Humanos Par 19 , Códon sem Sentido , Glioma/genética , Glioma/metabolismo , Humanos , Isocitrato Desidrogenase/genética , Perda de Heterozigosidade , Neurônios/metabolismo , Proteínas de Ligação a RNA
3.
Oncogene ; 32(20): 2586-91, 2013 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22751129

RESUMO

AVEN has been identified as an inhibitor of apoptosis, which binds to the adaptor protein, APAF-1, and thereby prevents apoptosome formation and mitochondrial apoptosis. Recent data have demonstrated high expression levels of AVEN messenger RNA in acute leukemias as well as a positive correlation between AVEN mRNA overexpression and poor prognosis in childhood acute lymphoblastic leukemia. On the basis of these data, we investigated the potential involvement of AVEN in tumorigenesis. First, we confirmed the overexpression of AVEN in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) patient samples. We then established a transgenic mouse model with T-cell-specific overexpression of AVEN, with which we demonstrated the oncogenic cooperation of AVEN with heterozygous loss of p53. Finally, we used a subcutaneous xenograft mouse model to show that AVEN knockdown in the T-ALL cell lines, MOLT-4 and CCRF-CEM, and in the acute myeloblastic leukemia cell line, Kasumi-1, leads to a halt in tumor growth owing to the increased apoptosis and decreased proliferation of tumor cells. Collectively, our data demonstrate that the anti-apoptotic molecule, AVEN, functions as an oncoprotein in hematopoietic neoplasms.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas de Membrana/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Regulação Leucêmica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes p53 , Humanos , Linfoma de Células T/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Timócitos/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cell Death Differ ; 19(9): 1435-45, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22388353

RESUMO

The anti-apoptotic molecule Aven was originally identified in a yeast two-hybrid screen for Bcl-x(L)-interacting proteins and has also been found to bind Apaf-1, thereby interfering with Apaf-1 self-association during apoptosome assembly. Aven is expressed in a wide variety of adult tissues and cell lines, and there is increasing evidence that its overexpression correlates with tumorigenesis, particularly in acute leukemias. The mechanism by which the anti-apoptotic activity of Aven is regulated remains poorly understood. Here we shed light on this issue by demonstrating that proteolytic removal of an inhibitory N-terminal Aven domain is necessary to activate the anti-apoptotic potential of the molecule. Furthermore, we identify Cathepsin D (CathD) as the protease responsible for Aven cleavage. On the basis of our results, we propose a model of Aven activation by which its N-terminal inhibitory domain is removed by CathD-mediated proteolysis, thereby unleashing its cytoprotective function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Catepsina D/metabolismo , Proteínas de Membrana/metabolismo , Proteólise , Doença Aguda , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Fator Apoptótico 1 Ativador de Proteases/genética , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Catepsina D/genética , Linhagem Celular Tumoral , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Proteínas de Membrana/genética , Estrutura Terciária de Proteína
5.
Cell Death Dis ; 1: e88, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21368861

RESUMO

Fas ligand (FasL) is a transmembrane protein that regulates cell death in Fas-bearing cells. FasL-mediated cell death is essential for immune system homeostasis and the elimination of viral or transformed cells. Because of its potent cytotoxic activity, FasL expression at the cell surface is tightly regulated, for example, via processing by ADAM10 and SPPL2a generating soluble FasL and the intracellular fragments APL (ADAM10-processed FasL form) and SPA (SPPL2a-processed APL). In this study, we report that FasL processing by ADAM10 counteracts Fas-mediated cell death and is strictly regulated by membrane localization, interactions and modifications of FasL. According to our observations, FasL processing occurs preferentially within cholesterol and sphingolipid-rich nanodomains (rafts) where efficient Fas-FasL contact occurs, Fas receptor and FasL interaction is also required for efficient FasL processing, and FasL palmitoylation, which occurs within its transmembrane domain, is critical for efficient FasL-mediated killing and FasL processing.


Assuntos
Apoptose , Proteína Ligante Fas/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Membrana Celular/metabolismo , Humanos , Lipoilação , Proteínas de Membrana/metabolismo , Ligação Proteica
6.
Cell Death Differ ; 14(9): 1678-87, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17557115

RESUMO

Fas ligand (FasL) is a type II transmembrane protein belonging to the tumor necrosis factor family. Its binding to the cognate Fas receptor triggers the apoptosis that plays a pivotal role in the maintenance of immune system homeostasis. The cell death-inducing property of FasL has been associated with its extracellular domain, which can be cleaved off by metalloprotease activity to produce soluble FasL. The fate of the remaining membrane-anchored N-terminal part of the FasL molecule has not been determined. Here we show that post-translational processing of overexpressed and endogenous FasL in T-cells by the disintegrin and metalloprotease ADAM10 generates a 17-kDa N-terminal fragment, which lacks the receptor-binding extracellular domain. This FasL remnant is membrane anchored and further processed by SPPL2a, a member of the signal peptide peptidase-like family of intramembrane-cleaving proteases. SPPL2a cleavage liberates a smaller and highly unstable fragment mainly containing the intracellular FasL domain (FasL ICD). We show that this fragment translocates to the nucleus and is capable of inhibiting gene transcription. With ADAM10 and SPPL2a we have identified two proteases implicated in FasL processing and release of the FasL ICD, which has been shown to be important for retrograde FasL signaling.


Assuntos
Proteínas ADAM/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Proteína Ligante Fas/metabolismo , Linfócitos T/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Proteína Ligante Fas/química , Humanos , Microscopia Confocal , Estrutura Terciária de Proteína , RNA Interferente Pequeno
7.
Cell Death Differ ; 13(2): 189-201, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16082389

RESUMO

The ecdysone-inducible mammalian expression system is frequently used for inducible transgene expression in vitro and in vivo. Here, we describe a strong antiapoptotic effect of ecdysone analogs in the human colon carcinoma cell line RKO, which is in contrast to published data that ecdysteroids do not influence mammalian cell physiology. Inhibition of Fas ligand- and TNF-related apoptosis-inducing ligand-induced apoptosis by muristerone A occurs at the level of caspase-8 activation and is neutralized by phosphatidylinositol-3-kinase/Akt, protein kinase C and mitogen-activated protein kinase inhibitors. Microarray, Northern and Western blot analysis revealed that incubation of RKO cells with muristerone A leads to changes in gene expression levels, including an upregulation of bcl-x(L) mRNA and protein levels. Our data imply that ecdysteroids and ecdysone mimics can induce and/or repress gene transcription in RKO and other mammalian cells, thereby influencing the apoptotic behavior. Therefore, the ecdysone-inducible mammalian expression system may not be suitable for the analysis of apoptosis-related genes.


Assuntos
Proteínas Reguladoras de Apoptose/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Colo/patologia , Ecdisterona/análogos & derivados , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicoproteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Fatores de Necrose Tumoral/farmacologia , Androstadienos/farmacologia , Apoptose/genética , Caspase 8 , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Cromonas/farmacologia , Neoplasias do Colo/química , Neoplasias do Colo/genética , Ecdisterona/farmacologia , Ativação Enzimática/efeitos dos fármacos , Proteína Ligante Fas , Flavonoides/farmacologia , Humanos , Morfolinas/farmacologia , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Antígeno Nuclear de Célula em Proliferação/análise , Antígeno Nuclear de Célula em Proliferação/genética , Proteína Quinase C/antagonistas & inibidores , RNA Mensageiro/análise , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF , Wortmanina , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
8.
Gut ; 55(2): 234-42, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16118352

RESUMO

BACKGROUND: High mobility group box 1 (HMGB1) is a non-histone chromosomal protein implicated in a variety of biologically important processes, including transcription, DNA repair, V(D)J recombination, differentiation, and development. Overexpression of HMGB1 inhibits apoptosis, arguing that the molecule may act as an antiapoptotic oncoprotein. Indeed, increased expression of HMGB1 has been reported for several different tumour types. In this study, we analysed human colon carcinoma for HMGB1 as well as for c-IAP2 expression levels. c-IAP2 is an antiapoptotic protein which may be upregulated as a consequence of nuclear factor kappaB (NFkappaB) activation via HMGB1. METHODS: A comparative genomic hybridisation (CGH) database comprising 1645 cases from different human tumour types was screened to detect cytogenetic changes at the HMGB1 locus. Immunohistochemical staining of human colon tissue microarrays and tumour biopsies, as well as western blot analysis of tumour lysates, were performed to detect elevated HMGB1 and c-IAP2 expression in colon carcinomas. The antiapoptotic potential of HMGB1 was analysed by measuring caspase activities, and luciferase reporter assays and quantitative polymerase chain reaction analysis were employed to confirm NFkappaB activation and c-IAP2 mRNA upregulation on HMGB1 overexpression. RESULTS: According to CGH analysis, the genomic locus containing the HMGB1 gene was overrepresented in one third (35/96) of colon cancers. Correspondingly, HMGB1 protein levels were significantly elevated in 90% of the 60 colon carcinomas tested compared with corresponding normal tissues evaluable from the same patients. HMGB1 increased NFkappaB activity and led to co-overexpression of the antiapoptotic NFkappaB target gene product c-IAP2 in vitro. Furthermore, increased HMGB1 levels correlated with enhanced amounts of c-IAP2 in colon tumours analysed by us. Finally, we demonstrated that HMGB1 overexpression suppressed caspase-9 and caspase-3 activity, suggesting that HMGB1 interferes with the apoptotic machinery at the level of apoptosomal caspase-9 activation. CONCLUSIONS: We identified in vitro a molecular pathway triggered by HMGB1 to inhibit apoptosis via c-IAP2 induction. Our data indicate a strong correlation between upregulation of the apoptosis repressing HMGB1 and c-IAP2 proteins in the pathogenesis of colon carcinoma.


Assuntos
Neoplasias do Colo/metabolismo , Proteína HMGB1/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Proteínas de Neoplasias/metabolismo , Apoptose , Western Blotting , Inibidores de Caspase , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , NF-kappa B/metabolismo , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA Mensageiro/genética , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transdução de Sinais , Regulação para Cima
9.
Cell Death Differ ; 13(3): 446-53, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16167071

RESUMO

The histone deacetylase (HDAC) inhibitor valproic acid (VPA) was recently shown to inhibit angiogenesis, but displays no toxicity in endothelial cells. Here, we demonstrate that VPA increases extracellular signal-regulated kinase 1/2 (ERK 1/2) phosphorylation in human umbilical vein endothelial cells (HUVEC). The investigation of structurally modified VPA derivatives revealed that the induction of ERK 1/2 phosphorylation is not correlated to HDAC inhibition. PD98059, a pharmacological inhibitor of the mitogen-activated protein kinase kinase 1/2, prevented the VPA-induced ERK 1/2 phosphorylation. In endothelial cells, ERK 1/2 phosphorylation is known to promote cell survival and angiogenesis. Our results showed that VPA-induced ERK 1/2 phosphorylation in turn causes phosphorylation of the antiapoptotic protein Bcl-2 and inhibits serum starvation-induced HUVEC apoptosis and cytochrome c release from the mitochondria. Moreover, the combination of VPA with PD98059 synergistically inhibited angiogenesis in vitro and in vivo.


Assuntos
Inibidores da Angiogênese/farmacologia , Apoptose/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Ácido Valproico/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Sinergismo Farmacológico , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Fosforilação
10.
Biochim Biophys Acta ; 1551(2): F1-37, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11591448

RESUMO

It has become clear that, together with deregulated growth, inhibition of programmed cell death (PCD) plays a pivotal role in tumorigenesis. In this review, we present an overview of the genes and mechanisms involved in PCD. We then summarize the evidence that impaired PCD is a prerequisite for tumorigenesis, as indicated by the fact that more and more neoplastic mutations appear to act by interfering with PCD. This has made the idea of restoration of corrupted 'death programs' an intriguing new area for potential cancer therapy.


Assuntos
Apoptose/genética , Neoplasias/etiologia , Animais , Caspases/genética , Evolução Molecular , Genes bcl-2 , Genes p53 , Humanos , Modelos Animais , Modelos Químicos , Mutação , Neoplasias/genética , Proteínas Oncogênicas/fisiologia , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais
11.
J Biol Chem ; 275(14): 10453-62, 2000 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-10744735

RESUMO

Death domain-containing members of the tumor necrosis factor (TNF) receptor family ("death receptors") can induce apoptosis upon stimulation by their natural ligands or by agonistic antibodies. Activated death receptors recruit death domain adapter proteins like Fas-associated death domain protein (FADD), and this ultimately leads to proteolytic activation of the caspase cascade and cell death. Recently, FADD has also been implicated in the regulation of proliferation; functional inhibition of FADD results in p53-dependent impairment of proliferation in activated T-cells. In this study we have further analyzed T-cells derived from transgenic mice expressing a dominant negative FADD mutant (FADD DN) under control of the lck promoter in vitro so as to identify the signaling pathways that become engaged upon T-cell receptor stimulation and that are regulated by death receptors. FADD DN expression inhibits T-cell proliferation, both at the G(0) --> S transition and in the G(1) phase of continuously proliferating cells. We observe a decrease in the release of calcium from intracellular stores after T-cell receptor stimulation, whereas influx of extracellular calcium seems to be unaffected. FADD DN-expressing fibroblasts show a similarly inhibited cell growth and impaired calcium mobilization indicating that the modulation of proliferation and calcium response by death receptors is not cell type-specific.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Cálcio/metabolismo , Proteínas de Transporte/fisiologia , Ciclo Celular/fisiologia , Linfócitos T/fisiologia , Células 3T3 , Animais , Apoptose , Bombesina/farmacologia , Proteínas de Transporte/química , Proteínas de Transporte/genética , Divisão Celular/efeitos dos fármacos , Proteína de Domínio de Morte Associada a Fas , Fibroblastos/citologia , Fibroblastos/fisiologia , Regulação da Expressão Gênica/imunologia , Interleucina-2/genética , Interleucina-6/genética , Ionomicina/farmacologia , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Transdução de Sinais , Baço/imunologia , Linfócitos T/citologia , Transcrição Gênica
12.
Curr Biol ; 8(8): 467-70, 1998 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-9550704

RESUMO

Members of the tumour necrosis factor (TNF) receptor family exert pleiotropic effects and can trigger both apoptosis and proliferation [1]. In their cytoplasmic region, some of these receptors share a conserved sequence motif - the 'death domain' - which is required for transduction of the apoptotic signal by recruiting other death-domain-containing adaptor molecules like the Fas-associated protein FADD/MORT1 or the TNF receptor-associated protein TRADD [2-4]. FADD links the receptor signal to the activation of the caspase family of cysteine proteases [5,6]. Functional inactivation of individual receptor family members often fails to exhibit a distinctive phenotype, probably because of redundancy [7-9]. To circumvent this problem, we used a dominant-negative mutant of FADD (FADD-DN) which should block all TNF receptor family members that use FADD as an adaptor. We established transgenic mice expressing FADD-DN under the influence of the lck promoter and investigated the consequences of its expression in T cells. As expected, FADD-DN thymocytes were protected from death induced by CD95 (Fas/Apo1), whereas apoptosis induced by ultraviolet (UV) irradiation, anti-CD3 antibody treatment or dexamethasone was unaffected, as was spontaneous cell death. Surprisingly, however, we also observed profound inhibition of thymocyte proliferation in vivo and of activation-induced proliferation of thymocytes and mature T cells in vitro. This inhibition of proliferation was not due to increased cell death and appeared to be p53 dependent.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/imunologia , Linfócitos T/citologia , Timo/imunologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose , Proteínas de Transporte/genética , Divisão Celular , Células Cultivadas , Proteína de Domínio de Morte Associada a Fas , Genes Dominantes , Ativação Linfocitária , Camundongos , Camundongos Transgênicos , Mitógenos/farmacologia , Mutação , Receptores do Fator de Necrose Tumoral/imunologia , Proteína Supressora de Tumor p53/genética , Receptor fas/farmacologia
14.
Science ; 278(5341): 1305-9, 1997 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-9360929

RESUMO

Induction of apoptosis by oncogenes like c-myc may be important in restraining the emergence of neoplasia. However, the mechanism by which c-myc induces apoptosis is unknown. CD95 (also termed Fas or APO-1) is a cell surface transmembrane receptor of the tumor necrosis factor receptor family that activates an intrinsic apoptotic suicide program in cells upon binding either its ligand CD95L or antibody. c-myc-induced apoptosis was shown to require interaction on the cell surface between CD95 and its ligand. c-Myc acts downstream of the CD95 receptor by sensitizing cells to the CD95 death signal. Moreover, IGF-I signaling and Bcl-2 suppress c-myc-induced apoptosis by also acting downstream of CD95. These findings link two apoptotic pathways previously thought to be independent and establish the dependency of Myc on CD95 signaling for its killing activity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose , Glicoproteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptor fas/metabolismo , Células 3T3 , Animais , Comunicação Autócrina , Proteínas de Transporte/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Proteína Ligante Fas , Proteína de Domínio de Morte Associada a Fas , Regulação da Expressão Gênica , Genes myc , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like I/fisiologia , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Ratos
15.
Mol Cell Biol ; 17(5): 2468-74, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9111315

RESUMO

Apoptosis as a form of programmed cell death (PCD) in multicellular organisms is a well-established genetically controlled process that leads to elimination of unnecessary or damaged cells. Recently, PCD has also been described for unicellular organisms as a process for the socially advantageous regulation of cell survival. The human Bcl-2 family member Bak induces apoptosis in mammalian cells which is counteracted by the Bcl-x(L) protein. We show that Bak also kills the unicellular fission yeast Schizosaccharomyces pombe and that this is inhibited by coexpression of human Bcl-x(L). Moreover, the same critical BH3 domain of Bak that is required for induction of apoptosis in mammalian cells is also required for inducing death in yeast. This suggests that Bak kills mammalian and yeast cells by similar mechanisms. The phenotype of the Bak-induced death in yeast involves condensation and fragmentation of the chromatin as well as dissolution of the nuclear envelope, all of which are features of mammalian apoptosis. These data suggest that the evolutionarily conserved metazoan PCD pathway is also present in unicellular yeast.


Assuntos
Apoptose , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces/citologia , Cromatina/ultraestrutura , Clonagem Molecular , Fragmentação do DNA , DNA Complementar/metabolismo , DNA Fúngico/metabolismo , Eletroforese em Gel de Poliacrilamida , Proteínas Fúngicas/genética , Humanos , Microscopia Eletrônica , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína bcl-X
16.
Curr Biol ; 6(12): 1553-6, 1996 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8994810

RESUMO

Recent evidence indicates that the c-Myc proto-oncogene activates transcription of cdc25A. The Cdc25A protein phosphatase is required both for progression through mitosis and for Myc-induced apoptosis, making cdc25A the most attractive Myc target gene identified so far.


Assuntos
Proteínas de Ciclo Celular/genética , Regulação da Expressão Gênica , Fosfoproteínas Fosfatases/genética , Proteínas Proto-Oncogênicas c-myc/genética , Transativadores/genética , Animais , Apoptose , Ciclo Celular , Sequências Hélice-Alça-Hélice , Humanos , Zíper de Leucina , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transativadores/metabolismo , Fosfatases cdc25
17.
Nucleic Acids Res ; 24(13): 2528-34, 1996 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8692692

RESUMO

Infecting mice with a mutant Moloney murine leukemia virus which contains the bacterial suppressor tRNA supF in its LTR allows rapid cloning of proviral integration sites from genomic tumour DNA. In a previous study Emu pim-1/Emu L-myc bitransgenic mice had been inoculated neonatally with MoMuLV supF virus. The retroviral infection led to acceleration of lymphomagenesis indicating the proviral activation of further oncogenes cooperating with myc and pim-1 in tumour development. Using a functional supF screen for analysis of genomic mouse tumour DNA libraries which had been constructed in the phage vector EMBL3A, a common proviral integration site on mouse chromosome 5 was cloned and found to be identical to the proviral integration site evi-5 which has recently been identified in an AKXD T-cell lymphoma and which is located 18 kb upstream of the gfi-1 gene. Tumours bearing evi-5 integrations showed an enhanced gfi-1 expression level suggesting that gfi-1 is the target gene for insertions at the evi-5 locus. Together with three other previously described Moloney integration clusters all responsible for enhanced gfi-1 expression the number of tumours from infected double transgenic Emu L-myc/Emu pim-1 transgenic mice with retrovirally activated gfi-1 added up to 53% underscoring the role of GFI-1 as an effective collaborator for MYC and PIM-1 in the process of lymphomagenesis.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Linfoma/genética , Vírus da Leucemia Murina de Moloney/genética , Proteínas Serina-Treonina Quinases , Fatores de Transcrição , Integração Viral/genética , Animais , Clonagem Molecular , DNA de Neoplasias/genética , Genes Virais , Genes myc , Linfoma/etiologia , Linfoma de Células B/etiologia , Linfoma de Células B/genética , Linfoma de Células T/etiologia , Linfoma de Células T/genética , Camundongos , Camundongos Transgênicos , Oncogenes , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-pim-1 , RNA Mensageiro/análise , RNA de Transferência/genética , Seleção Genética
18.
Oncogene ; 12(8): 1789-801, 1996 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-8622900

RESUMO

The clonality of lymphomas that originate in myc/pim-1 bitransgenic mice due to synergistic action of both oncogenes indicates the requirement of additional events for progression to full malignancy. To isolate genes that cooperate with both myc and pim-1, we have used provirus tagging with E mu L-myc/pim-1 double transgenic mice. We find accelerated tumour formation in infected animals and show that the gfi-1 gene and neighbouring loci on mouse chromosome 5 are occupied by proviruses in about 53% of the tumours leading in all cases to high level gfi-1 expression. In agreement with data from Gilks et al. (1993) we find that forced expression of the gfi-1 encoded zinc finger protein in IL-2 dependent T-cells provokes increased survival upon IL-2 depletion and we present evidence that this occurs at least in part through stimulation of proliferation. Our data suggest that gfi-1 is a proto-oncogene cooperation with both myc and pim-1 genes in T-cell lymphomagenesis.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas de Ligação a DNA/fisiologia , Genes myc , Proteínas de Homeodomínio , Interleucina-2/metabolismo , Linfoma de Células T/genética , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Transativadores , Fatores de Transcrição , Sequência de Aminoácidos , Animais , Sequência de Bases , Ciclo Celular/genética , Mapeamento Cromossômico , Sequência Conservada , Elementos de DNA Transponíveis , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Helminto/genética , Interleucina-2/genética , Linfoma de Células T/patologia , Linfoma de Células T/virologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Vírus da Leucemia Murina de Moloney/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-pim-1 , Transfecção , Células Tumorais Cultivadas
19.
Oncogene ; 11(10): 2165-74, 1995 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-7478538

RESUMO

Coexpression of the proto-oncogenes c-myc and bcl-2 under the control of the immunoglobulin enhancer E mu provokes the rapid development of primitive lymphoid tumors in transgenic mice. In the present study we show that the myc family members N-myc and L-myc also cooperate with bcl-2 in oncogenesis and can provoke the development of more mature pre-B, B and T cell type lymphomas. The analysis of prelymphomatous B-cells from single E mu N-myc and bcl-2-Ig transgenic animals and from young, tumor free, double transgenic E mu N-myc/bcl-2-Ig mice revealed that E mu directed expression of N-myc leads to very rapid apoptosis after explantation and culturing compared to B-cells from normal mice. As expected, B-cells from bcl-2-Ig transgenics were protected to a certain degree from apoptosis. Strikingly however, B-cells from E mu N-myc/bcl-2-Ig double transgenic animals were found to be almost completely resistant towards a number of different apoptotic stimuli. Furthermore, after treatment with H2O2, which can trigger apoptosis, B-cells from E mu N-myc animals reach levels of intracellular free Ca2+ concentrations that are comparable to B-cells from normal mice, whereas B-cells from bcl-2-Ig or E mu N-myc/bcl-2-Ig double transgenic mice show no increase in intracellular Ca2+ concentrations after stimulation with H2O2. These findings suggest that the prevention of apoptosis conferred by bcl-2 correlates with the inhibition of intracellular Ca2+ fluxes whereas induction of apoptosis mediated by N-myc requires normal Ca2+ levels. We hypothesize therefore that the regulation of intracellular Ca2+ concentrations represent one important parameter in the oncogenic cooperation between bcl-2 and N-myc.


Assuntos
Apoptose/fisiologia , Linfócitos B/metabolismo , Cálcio/metabolismo , Genes myc/fisiologia , Linfoma de Células B/genética , Lesões Pré-Cancerosas/genética , Proteínas Proto-Oncogênicas/genética , Animais , Linfócitos B/citologia , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Feminino , Expressão Gênica , Peróxido de Hidrogênio/farmacologia , Líquido Intracelular/metabolismo , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas c-bcl-2
20.
Oncogene ; 10(12): 2397-401, 1995 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-7784089

RESUMO

The Fas/Apo-1 receptor is an integral membrane protein that transduces apoptotic signals upon binding to its natural ligand or to specific antibodies. Loss of Fas/Apo-1 receptor leads in (lpr,lpr) mice to a nonmalignant accumulation of abnormal T-cells very probably due to the lack of induction of apoptosis in peripheral T-cells. It has been reported that soluble forms of Fas/Apo-1 receptor that may interfere with apoptotic signaling occur in patients suffering from various forms of lymphoid neoplasms. Therefore, we wished to investigate whether the loss of proper homeostatic regulation through Fas/Apo-1 receptor mediated apoptosis could influence the process of lymphomagenesis. To this end, we performed two experiments (i) we infected (lpr,lpr) animals with Moloney Murine Leukemia Virus (MoMuLV) that causes T-cell lymphoma in mice and (ii) we crossed (lpr,lpr) animals with E mu L-myc transgenic mice that are prone to develop T- and B-cell lymphoma due to deregulated expression of the L-myc transgene by the immunoglobulin enhancer E mu. We find that infection with MoMuLV did not accelerate the formation of lymphoid neoplasms in (lpr,lpr) mice when compared to infected normal animals. However, E mu L-myc/(lpr,lpr) animals that constitutively express the L-myc transgene in the lymphoid lineage clearly show accelerated formation of T- and B-cell lymphoma when compared to normal E mu L-myc transgenics. These data demonstrate that in cooperation with particular oncogenes impairment of Fas/Apo-1 receptor function can indeed affect and modulate the process of tumor formation.


Assuntos
Apoptose , Genes myc , Linfoma de Células B/química , Linfoma de Células B/etiologia , Linfoma de Células T/química , Linfoma de Células T/etiologia , Vírus da Leucemia Murina de Moloney , Receptores de Superfície Celular/fisiologia , Infecções por Retroviridae , Infecções Tumorais por Vírus , Animais , Rearranjo Gênico de Cadeia Pesada de Linfócito B , Rearranjo Gênico de Cadeia Leve de Linfócito B , Linfoma de Células B/genética , Linfoma de Células B/virologia , Linfoma de Células T/genética , Linfoma de Células T/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...