Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Oncogene ; 27(14): 2035-44, 2008 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-17922027

RESUMO

ATP-dependent SWI/SNF chromatin remodeling complexes regulate cell-cycle and play critical roles in a variety of differentiation pathways. The core subunit SNF5/INI1 is a tumor suppressor that is inactivated in a highly aggressive childhood cancer of unknown cellular origin, termed malignant rhabdoid tumor (MRT). The highly undifferentiated phenotype of this tumor suggests that the loss-of-function of hSNF5/INI1 impairs specific differentiation programs of the MRT parental cell. Based on the hypothesis that these programs might be reinitialized upon hSNF5/INI1 re-expression in MRTs, we show that some MRT cell lines can differentiate toward the adipogenic lineage. We further show that the knock down of the SNF5/INI1 subunit abrogates adipocyte differentiation of murine 3T3-L1 preadipocytes and of human mesenchymal stem cells. Finally, we provide evidence that hSNF5/INI1 cooperates with C/EBPbeta and PPARgamma2 transcriptional regulators to activate the expression of adipocyte-specific genes. These data indicate that not only the ATPase subunit of the SWI/SNF complex, but also SNF5/INI1 is required for adipocyte differentiation. They further show that MRT cell lines harbor an adipogenic differentiation potential and that the tumor suppressor role of the SNF5/INI1 subunit may rely on its ability to regulate the balance between cell proliferation and differentiation.


Assuntos
Adipogenia/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Tumor Rabdoide/genética , Tumor Rabdoide/patologia , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Humanos , Camundongos , PPAR gama/metabolismo , Proteína SMARCB1 , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética
2.
Oncogene ; 26(2): 165-72, 2007 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-16832348

RESUMO

Following prolonged mitotic spindle disruption by microtubule poisons, mammalian cells delay their entry into anaphase, then progressively slip out of mitosis and become tetraploid. Normal cells then stop cycling before S-phase onset, but the mechanisms underlying this arrest are still unclear. Here we show that a double block prevents endo-reduplication. First, cells that exit mitosis without a functional microtubule network are driven toward G0. Reconstitution of the network unmasks a second block that relies on DNA double-strand breaks occurring early in the G1 phase that follows the mitotic block. We propose that a stress signal elicited upon mitotic impairment triggers breakage, which couples the leaky spindle checkpoint to the stringent DNA damage response. Consistent with this finding, cells defective for the damage response continue cycling and acquire, within a single cell cycle, both chromosome rearrangements and abnormal chromosome numbers that remarkably mimic the complex genetic hallmark of tumorigenesis.


Assuntos
Cromossomos , Dano ao DNA , Mitose/fisiologia , Ploidias , Adenocarcinoma/patologia , Anáfase , Animais , Antineoplásicos/farmacologia , Células Cultivadas , Neoplasias do Colo/patologia , Cricetinae , Cricetulus , Quebras de DNA de Cadeia Dupla , Fibroblastos/fisiologia , Fase G1/genética , Rearranjo Gênico , Humanos , Immunoblotting , Hibridização in Situ Fluorescente , Pulmão/fisiologia , Microtúbulos , Mitose/efeitos dos fármacos , Índice Mitótico , Nocodazol/farmacologia , Fase de Repouso do Ciclo Celular , Fuso Acromático
3.
Blood ; 96(8): 2849-55, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11023521

RESUMO

Human T-cell lymphotropic virus type I (HTLV-I)-associated adult T-cell leukemia/lymphoma (ATL) is a malignancy of mature activated T cells resistant to conventional chemotherapy. The viral transactivator protein Tax plays a critical role in HTLV-I-induced transformation and apoptosis resistance by inducing I kappa B-alpha degradation, resulting in the activation of the NF-kappa Bpathway. In these HTLV-I-transformed cells, arsenic trioxide (As) and interferon (IFN)-alpha synergize to induce cell cycle arrest and apoptosis. We demonstrate that cell death induction is only partly dependent upon caspase activation and is not associated with modulation of bcl-2, bax, or p53 expression. However, combined As and IFN induce the degradation of Tax, associated with an up-regulation of I kappa B-alpha resulting in a sharp decrease in RelA DNA binding nuclear factor (NF)-kappa B complexes because of the cytoplasmic retention of RelA. Taken the role of Tax in HTLV-I-induced transformation, its down-regulation probably accounts for cell death induction through inactivation of the NF-kappa B pathway. Such specific targeting of the viral oncoprotein by As-IFN treatment, reminiscent of As targeting of promyelocytic leukemia/retinoic acid receptor-alpha in acute promyelocytic leukemia, provides strong rational for combined As-IFN therapy in ATL patients. (Blood. 2000;96:2849-2855)


Assuntos
Apoptose/efeitos dos fármacos , Arsenicais/farmacologia , Transformação Celular Viral/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Produtos do Gene tax/fisiologia , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Proteínas I-kappa B , Interferon-alfa/farmacologia , Leucemia-Linfoma de Células T do Adulto/genética , NF-kappa B/metabolismo , Proteínas de Neoplasias/fisiologia , Óxidos/farmacologia , Ativação Transcricional/efeitos dos fármacos , Trióxido de Arsênio , Caspases/fisiologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Transformada , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Sinergismo Farmacológico , Ativação Enzimática , Humanos , Leucemia-Linfoma de Células T do Adulto/patologia , Ligases/metabolismo , Substâncias Macromoleculares , Inibidor de NF-kappaB alfa , Linfócitos T/metabolismo , Linfócitos T/virologia , Células Tumorais Cultivadas
4.
Proc Natl Acad Sci U S A ; 96(26): 14807-12, 1999 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-10611294

RESUMO

Analyzing the pathways by which retinoic acid (RA) induces promyelocytic leukemia/retinoic acid receptor alpha (PML/RARalpha) catabolism in acute promyelocytic leukemia (APL), we found that, in addition to caspase-mediated PML/RARalpha cleavage, RA triggers degradation of both PML/RARalpha and RARalpha. Similarly, in non-APL cells, RA directly targeted RARalpha and RARalpha fusions to the proteasome degradation pathway. Activation of either RARalpha or RXRalpha by specific agonists induced degradation of both proteins. Conversely, a mutation in RARalpha that abolishes heterodimer formation and DNA binding, blocked both RARalpha and RXRalpha degradation. Mutations in the RARalpha DNA-binding domain or AF-2 transcriptional activation region also impaired RARalpha catabolism. Hence, our results link transcriptional activation to receptor catabolism and suggest that transcriptional up-regulation of nuclear receptors by their ligands may be a feedback mechanism allowing sustained target-gene activation.


Assuntos
Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Receptores do Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Caspases/metabolismo , Análise Mutacional de DNA , Dimerização , Leucemia Promielocítica Aguda/metabolismo , Complexo de Endopeptidases do Proteassoma , Receptores do Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Receptores X de Retinoides , Fatores de Transcrição/metabolismo
5.
Oncogene ; 18(36): 5063-75, 1999 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-10490843

RESUMO

One of the most frequent genetic abnormalities associated with non Hodgkin lymphoma is the structural alteration of the 5' non coding/regulatory region of the BCL6 (LAZ3) protooncogene. BCL6 encodes a POZ/Zn finger protein, a structure similar to that of many Drosophila developmental regulators and to another protein involved in a human hematopoietic malignancy, PLZF. BCL6 is a sequence specific transcriptional repressor controlling germinal center formation and T cell dependent immune response. Although the expression of BCL6 negatively correlates with cellular proliferation in different cell types, the influence of BCL6 on cell growth and survival is currently unknown so that the way its deregulation may contribute to cancer remains elusive. To directly address this issue, we used a tetracycline-regulated system in human U2OS osteosarcoma cells and thus found that BCL6 mediates growth suppression associated with impaired S phase progression and apoptosis. Interestingly, overexpressed BCL6 can colocalize with sites of ongoing DNA synthesis, suggesting that it may directly interfere with S phase initiation and/or progression. In contrast, the isolated Zn finger region of BCL6, which binds BCL6 target sequence but lacks transcriptional repression activity, slows, but does not suppress, U2OS cell growth, is less efficient at delaying S phase progression, and does not trigger apoptosis. Thus, for a large part, the effects of BCL6 overexpression on cell growth and survival depend on its ability to engage protein/protein interactions with itself and/or its transcriptional corepressors. That BCL6 restricts cell growth suggests that its deregulation upon structural alterations may alleviate negative controls on the cell cycle and cell survival.


Assuntos
Apoptose/fisiologia , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fase S/fisiologia , Fatores de Transcrição/fisiologia , Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Humanos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-6 , Fatores de Transcrição/genética , Células Tumorais Cultivadas
6.
Blood ; 93(1): 278-83, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9864171

RESUMO

Human T-cell lymphotropic virus type I (HTLV-I) is the causative agent of adult T-cell leukemia/lymphoma (ATL). ATL is an aggressive proliferation of mature activated T cells associated with a poor prognosis. The combination of the antiviral agents, zidovudine (AZT) and interferon (IFN), is a potent treatment of ATL. Recently, arsenic trioxide (As) was shown to be an effective treatment of acute promyelocytic leukemia (APL). We have tested the effects of the combination of As and IFN on cell proliferation, cell cycle phases distribution, and apoptosis in ATL-derived or control T-cell lines. A high synergistic effect between IFN and As was observed in ATL-derived cell lines in comparison to the control cell lines, with a dramatic inhibition of cell proliferation, G1 arrest, and induction of apoptosis. Similar results were obtained with fresh leukemia cells derived from an ATL patient. Although the mechanisms involved are unclear, these results could provide a rational basis for combined As and IFN treatments in ATL.


Assuntos
Apoptose/efeitos dos fármacos , Arsenicais/farmacologia , Ciclo Celular/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Interferon-alfa/farmacologia , Óxidos/farmacologia , Antineoplásicos/farmacologia , Trióxido de Arsênio , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Células Cultivadas , Sinergismo Farmacológico , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Células Jurkat , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/virologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/virologia , Células Tumorais Cultivadas , Proteínas Virais/biossíntese
7.
Nat Genet ; 20(3): 259-65, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9806544

RESUMO

PML nuclear bodies (NBs) are nuclear matrix-associated structures altered by viruses and oncogenes. We show here that PML overexpression induces rapid cell death, independent of de novo transcription and cell cycling. PML death involves cytoplasmic features of apoptosis in the absence of caspase-3 activation, and caspase inhibitors such as zVAD accelerate PML death. zVAD also accelerates interferon (IFN)-induced death, suggesting that PML contributes to IFN-induced apoptosis. The death effector BAX and the cdk inhibitor p27KIP1 are novel NB-associated proteins recruited by PML to these nuclear domains, whereas the acute promyelocytic leukaemia (APL) PML/RAR alpha oncoprotein delocalizes them. Arsenic enhances targeting of PML, BAX and p27KIP1 to NBs and synergizes with PML and IFN to induce cell death. Thus, cell death susceptibility correlates with NB recruitment of NB proteins. These findings reveal a novel cell death pathway that neither requires nor induces caspase-3 activation, and suggest that NBs participate in the control of cell survival.


Assuntos
Apoptose/fisiologia , Proteínas de Ciclo Celular , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares , Proteínas Proto-Oncogênicas c-bcl-2 , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Arsênio/farmacologia , Caspase 3 , Caspases/fisiologia , Núcleo Celular/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27 , Inibidores de Cisteína Proteinase/farmacologia , Ativação Enzimática , Expressão Gênica , Humanos , Interferon Tipo I/farmacologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/genética , Proteína da Leucemia Promielocítica , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Proteínas Recombinantes , Fatores de Transcrição/genética , Proteína X Associada a bcl-2
8.
J Virol ; 72(2): 1043-51, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9444998

RESUMO

The interferon (IFN)-induced promyelocytic leukemia (PML) protein is specifically associated with nuclear bodies (NBs) whose functions are yet unknown. Two of the NB-associated proteins, PML and Sp100, are induced by IFN. Here we show that overexpression of PML and not Sp100 induces resistance to infections by vesicular stomatitis virus (VSV) (a rhabdovirus) and influenza A virus (an orthomyxovirus) but not by encephalomyocarditis virus (a picornavirus). Inhibition of viral multiplication was dependent on both the level of PML expression and the multiplicity of infection and reached 100-fold. PML was shown to interfere with VSV mRNA and protein synthesis. Compared to the IFN mediator MxA protein, PML had less powerful antiviral activity. While nuclear body localization of PML did not seem to be required for the antiviral effect, deletion of the PML coiled-coil domain completely abolished it. Taken together, these results suggest that PML can contribute to the antiviral state induced in IFN-treated cells.


Assuntos
Antígenos Nucleares , Vírus da Influenza A , Influenza Humana/metabolismo , Interferons/farmacologia , Proteínas de Neoplasias , Infecções por Rhabdoviridae/metabolismo , Fatores de Transcrição/metabolismo , Vírus da Estomatite Vesicular Indiana , Animais , Autoantígenos/metabolismo , Células CHO , Cricetinae , Suscetibilidade a Doenças , Humanos , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
9.
Proc Natl Acad Sci U S A ; 94(19): 10255-60, 1997 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-9294197

RESUMO

In acute promyelocytic leukemia (APL), the typical t(15;17) and the rare t(11;17) translocations express, respectively, the PML/RARalpha and PLZF/RARalpha fusion proteins (where RARalpha is retinoic acid receptor alpha). Herein, we demonstrate that the PLZF and PML proteins interact with each other and colocalize onto nuclear bodies (NBs). Furthermore, induction of PML expression by interferons leads to a recruitment of PLZF onto NBs without increase in the levels of the PLZF protein. PML/RARalpha and PLZF/RARalpha localize to the same microspeckled nuclear domains that appear to be common targets for the two fusion proteins in APL. Although PLZF/RARalpha does not affect the localization of PML, PML/RARalpha delocalizes the endogenous PLZF protein in t(15;17)-positive NB4 cells, pointing to a hierarchy in the nuclear targeting of these proteins. Thus, our results unify the molecular pathogenesis of APL with at least two different RARalpha gene translocations and stress the importance of alterations of PLZF and RARalpha nuclear localizations in this disease.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias , Proteínas Nucleares , Receptores do Ácido Retinoico/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células COS , Dimerização , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Receptor alfa de Ácido Retinoico , Proteínas Supressoras de Tumor
10.
Proc Natl Acad Sci U S A ; 94(8): 3978-83, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9108090

RESUMO

Acute promyelocytic leukemia (APL) is associated with the t(15;17) translocation, which generates a PML/RAR alpha fusion protein between PML, a growth suppressor localized on nuclear matrix-associated bodies, and RAR alpha, a nuclear receptor for retinoic acid (RA). PML/RAR alpha was proposed to block myeloid differentiation through inhibition of nuclear receptor response, as does a dominant negative RAR alpha mutant. In addition, in APL cells, PML/RAR alpha displaces PML and other nuclear body (NB) antigens onto nuclear microspeckles, likely resulting in the loss of PML and/or NB functions. RA leads to clinical remissions through induction of terminal differentiation, for which the respective contributions of RAR alpha (or PML/RAR alpha) activation, PML/RAR alpha degradation, and restoration of NB antigens localization are poorly determined. Arsenic trioxide also leads to remissions in APL patients, presumably through induction of apoptosis. We demonstrate that in non-APL cells, arsenic recruits the nucleoplasmic form of several NB antigens onto NB, but induces the degradation of PML only, identifying a powerful tool to approach NB function. In APL cells, arsenic targets PML and PML/RAR alpha onto NB and induces their degradation. Thus, RA and arsenic target RAR alpha and PML, respectively, but both induce the degradation of the PML/RAR alpha fusion protein, which should contribute to their therapeutic effects. The difference in the cellular events triggered by these two agents likely stems from RA-induced transcriptional activation and arsenic effects on NB proteins.


Assuntos
Arsênio/farmacologia , Núcleo Celular/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Apoptose/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Humanos , Leucemia Promielocítica Aguda/patologia , Leucemia Promielocítica Aguda/terapia
12.
Exp Cell Res ; 229(2): 253-60, 1996 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8986606

RESUMO

Acute promyelocytic leukemia (APL) is specifically associated to a t(15; 17) translocation which fuses a gene encoding a nuclear receptor for retinoic acid, RARalpha, to a previously unknown gene PML. The PML protein is localized in the nucleus on a specific domain of unknown function (PML nuclear bodies, NB) previously detected with autoimmune sera from patients with primary biliary cirrhosis (PBC). These bodies are nuclear matrix-associated and all of their identified components (PML, Sp100, and NDP52) are sharply upregulated by interferons. We show that autoantibodies against both PML and Sp100 are usually associated in sera with multiple nuclear dot anti-nuclear antibodies and demonstrate that PML is an autoantigen, not only in PBC, but also in other autoimmune diseases. In APL, the PML/RARalpha fusion interferes with both the retinoic acid (RA) response and PML localization on nuclear bodies, but the respective contribution of each defect to leukemogenesis is unclear. RA induces the terminal differentiation of APL blasts, yielding to complete remissions, and corrects the localization of NB antigens. Arsenic trioxide (As2O3) also induces remissions in APL, seemingly through induction of apoptosis. We show that in APL, As2O3 leads to the rapid reformation of PML bodies. Thus, both agents correct the defect in NB antigen localization, stressing the role of nuclear bodies in the pathogenesis of APL.


Assuntos
Doenças Autoimunes/genética , Leucemia Promielocítica Aguda/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares , Proteínas de Fusão Oncogênica/genética , Fatores de Transcrição/genética , Animais , Anticorpos Antinucleares , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Trióxido de Arsênio , Arsenicais/farmacologia , Autoantígenos , Doenças Autoimunes/tratamento farmacológico , Células CHO , Diferenciação Celular/efeitos dos fármacos , Cricetinae , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/etiologia , Proteínas de Neoplasias/imunologia , Proteínas de Fusão Oncogênica/imunologia , Óxidos/farmacologia , Proteína da Leucemia Promielocítica , Transfecção , Tretinoína/farmacologia , Proteínas Supressoras de Tumor
13.
Oncogene ; 12(9): 2011-7, 1996 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-8649862

RESUMO

Three hepatitis B viruses infecting humans, woodchucks and ground squirrels increase the risk of hepatocellular carcinoma in their respective hosts. The woodchuck hepatitis B virus (WHV), unlike the two other viruses, induces a rapid carcinogenic process characterized by direct activation of myc proto-oncogenes by insertion of viral DNA. The highly preferred target of insertional mutagenesis in woodchucks is N-myc2, an intronless N-myc gene. Strikingly, N-myc2 has no human homolog and the homologous N-myc2 locus previously detected in the ground squirrel genome, remains silent during hepatocarcinogenesis. Therefore, N-myc2 may represent a critical host determinant in the evolution of the disease associated with hepadnavirus infection. To address this question, we performed a structural and functional analysis of the ground squirrel N-myc2 locus. We show that ground squirrel N-myc2 is highly homologous to its woodchuck counterpart and is a functional proto-oncogene. Existence of a functional N-myc2 gene as a potential target for insertional activation by viral DNA is therefore not restricted to the woodchuck species. This suggests that viral rather than host factors determine the higher oncogenic phenotype of WHV as compared to the two other mammalian hepadnaviruses.


Assuntos
Genes myc , Hepadnaviridae/patogenicidade , Neoplasias Hepáticas Experimentais/virologia , Retroelementos , Sciuridae/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sequência Conservada , DNA Viral , Neoplasias Hepáticas Experimentais/genética , Dados de Sequência Molecular , Proto-Oncogene Mas , Ratos , Ratos Endogâmicos F344 , Homologia de Sequência de Aminoácidos , Transcrição Gênica
14.
Ann Endocrinol (Paris) ; 57(2): 91-100, 1996.
Artigo em Francês | MEDLINE | ID: mdl-8881287

RESUMO

PML is a protein involved in the t (15, 17) translocation of promyelocytic leukemia and is mainly localized in nuclear bodies. Here we show that PML exerts a very powerful enhancing activity (up to 20-fold) on the transactivating properties of the progesterone receptor (PR) and has a similar effect on several other steroid hormone receptors. There is probably a direct or indirect interaction between PR and PML since when the latter was expressed at high concentrations it shifted PR into the nuclear bodies. Use of deletion mutants showed that both activation functions (AF1 and AF2) of PR as well as the coiled coil and His-Cys rich domains of PML were required for transcriptional enhancement. The fusion protein PML-RAR, which is not localized in nuclear bodies, also enhanced the transactivating activity of PR but this effect was totally suppressed by the administration of retinoic acid. PML, which is ubiquitously expressed, may thus be involved in the transactivation properties of steroid hormone receptors. This mechanism may also play a role in the oncogenic properties of PML-RAR and in their suppression by the retinoic acid.


Assuntos
Leucemia Promielocítica Aguda/genética , Proteínas de Neoplasias/farmacologia , Proteínas Nucleares , Proteínas de Fusão Oncogênica/farmacologia , Receptores de Esteroides/genética , Fatores de Transcrição/farmacologia , Técnicas In Vitro , Receptores de Progesterona/análise , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Frações Subcelulares/metabolismo , Distribuição Tecidual , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Proteínas Supressoras de Tumor
15.
Leukemia ; 9(12): 2027-33, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8609713

RESUMO

PML has been identified through its fusion to the RAR alpha gene in acute promyelocytic leukemia (APL). The PML protein is specifically associated to nuclear bodies (NBs) whose alterations in APL were proposed to contribute to leukemogenesis. The role of this nuclear domain (which also harbors the Sp100 autoantigen and the NDP52 protein) is unknown. Here, we show that the PML protein, like Sp100 and NDP52, is induced by interferons (IFNs alpha, beta and gamma) in a large variety of human cells. Interestingly, the NBs that contain the three IFN-induced proteins appear to be associated to speckles labelled by the IFN-mediator Mx1. These observations link NBs to IFN response pathways, which may contribute to the elucidation of the biological role of these structures. In APL cells, IFNs induced both PML and PML/RAR alpha expression, resulting in an increased sequestration of PML and RXRs in the microspeckles induced by the fusion protein. As PML has growth suppressing properties, it may mediate some of the antiproliferative effects of IFN. In APL, inactivation of PML may result in disruption of growth control.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ligação ao GTP , Interferons/farmacologia , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias , Proteínas/metabolismo , Fatores de Transcrição/biossíntese , Western Blotting , Humanos , Microscopia Confocal , Proteínas de Resistência a Myxovirus , Proteínas Nucleares/biossíntese , Proteína da Leucemia Promielocítica , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
16.
Mol Endocrinol ; 9(12): 1791-803, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8614415

RESUMO

PML (promyelocytic leukemia) is a protein involved in the t (15;17) translocation of promyelocytic leukemia and is mainly localized in nuclear bodies. Here we show that PML exerts a very powerful enhancing activity (up to 20-fold) on the transactivating properties of the progesterone receptor (PR) and has a similar effect on several other steroid hormone receptors. There is probably a direct or indirect interaction between PR and PML, because when the latter was expressed at high concentrations it shifted PR into the nuclear bodies. The use of deletion mutants showed that both activation functions (AF1 and AF2) of PR as well as the coiled coil and His-Cys-rich domains of PML were required for transcriptional enhancement. The fusion protein PML-RAR which is not localized in nuclear bodies, also enhanced the transactivating activity of PR, but this effect was totally suppressed by the administration of retinoic acid. PML, which is ubiquitously expressed, may thus be involved in the transactivation properties of steroid hormone receptors. This mechanism may also play a role in the oncogenic properties of PML-RAR and in their suppression by retinoic acid.


Assuntos
Proteínas de Neoplasias , Proteínas Nucleares , Receptores do Ácido Retinoico/fisiologia , Fatores de Transcrição/farmacologia , Animais , Sequência de Bases , Células CHO , Linhagem Celular , Cricetinae , Células HeLa , Humanos , Dados de Sequência Molecular , Proteína da Leucemia Promielocítica , Receptores de Progesterona/genética , Receptores de Progesterona/fisiologia , Receptores do Ácido Retinoico/genética , Proteínas Recombinantes de Fusão , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Transfecção , Proteínas Supressoras de Tumor
17.
Oncogene ; 11(12): 2565-73, 1995 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-8545113

RESUMO

PML is a nuclear matrix protein with growth suppressing properties, whose expression is deregulated during oncogenesis. Moreover, in the t(15;17) translocation of acute promyelocytic leukaemia (APL), PML fusion to the retinoic acid receptor alpha (RAR alpha) is the likely molecular basis of leukaemogenesis. Here we show that interferons (IFNs) alpha, beta, and gamma upregulate PML mRNA expression. Analysis of 5' genomic sequences of the PML gene revealed an IFN-alpha/-beta stimulated response element (ISRE) and an IFN-gamma activation site (GAS) in the untranslated first exon. Binding of IFN signal transducers and activators of transcription (STATs) was demonstrated to be weak for the PML GAS, but strong for the PML ISRE which also seemed to contribute substantially to the IFN-gamma response. Thus, PML is a primary target gene of IFNs and would appear as a suitable candidate for mediating some of their antiproliferative effects. Abnormalities of PML structure, localisation or expression in human malignancy, constitute examples of how an IFN target gene may be altered in oncogenesis.


Assuntos
Interferons/farmacologia , Proteínas de Neoplasias , Proteínas Nucleares , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Cricetinae , Células HeLa , Humanos , Dados de Sequência Molecular , Proteína da Leucemia Promielocítica , Proteínas Supressoras de Tumor
18.
Exp Cell Res ; 218(1): 9-16, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7737384

RESUMO

We have studied at the ultrastructural level the localization of PML protein in response to adenovirus infection in HeLa cells. In nuclei of noninfected cells or at the early stage of infection, PML accumulated at the border of nuclear bodies as previously described [Koken et al. (1994) EMBO J. 13, 1073-1083]. Interestingly, we demonstrate herein that PML is also a component of the interchromatin granule-associated zones, recently described structures containing U1 snRNP [Visa et al. (1993) Eur. J. Cell Biol. 60, 308-321], suggesting that PML protein might be involved in some steps of splicing events. However, as the infection progressed these two cellular PML-containing structures disappeared but significant amounts of PML accumulated within two virus-induced structures, essentially the clear amorphous inclusions, but also the protein crystals. Relocalization of this protein to virus-induced structures may reflect an inactivation of PML functions.


Assuntos
Adenovírus Humanos/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Proteínas de Neoplasias , Proteínas Nucleares , Fatores de Transcrição/metabolismo , Western Blotting , Transformação Celular Viral , Imunofluorescência , Células HeLa , Humanos , Microscopia Eletrônica , Proteína da Leucemia Promielocítica , Fatores de Transcrição/análise , Proteínas Supressoras de Tumor
19.
Oncogene ; 10(7): 1315-24, 1995 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-7731682

RESUMO

Altered sub-nuclear localisation of the nuclear body-associated PML protein in acute promyelocytic leukaemia, has been proposed to contribute to leukaemogenesis. We have recently shown that PML is a primary target gene of interferons. Here, it is shown that PML has growth suppressive properties and displays an altered expression pattern during human oncogenesis. PML is widely expressed in cell-lines and is cell-cycle regulated. Overexpression of the protein induces a sharp reduction in growth rates in vitro and in vivo. In contrast with cell-lines, in normal tissues (including those that rapidly proliferate) only a few cells have detectable PML levels. However, these can be upregulated by soluble factors (e.g. IFN, estrogens). Human epithelial tumors show a gradual increase of PML levels as the lesion progresses from benign dysplasia to carcinoma. A similar induction is found in the surrounding stroma and vessels, which likely results from paracrine interactions. Strikingly, when malignant cells turn invasive, they loose PML expression, while expression is conserved in the stromal compartment. These observations point to the existence of a consistent deregulation in the expression of the PML growth-suppressor during human oncogenesis.


Assuntos
Ciclo Celular , Inibidores do Crescimento , Proteínas de Neoplasias , Neoplasias/metabolismo , Proteínas Nucleares , Fatores de Transcrição/metabolismo , Animais , Estrogênios/fisiologia , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteína da Leucemia Promielocítica , Pele/metabolismo , Transplante Heterólogo , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA