Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 19(54): 6240-50, 2000 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-11175338

RESUMO

Most acute promyelocytic leukemia (APL) cases are associated with recurrent translocations between the gene of retinoic receptor alpha and that of PML (t(15;17)) or PLZF (t(11;17)). PML localizes onto discrete intranuclear domains, the PML-nuclear bodies, and displays anti-oncogenic and pro-apoptotic properties. PLZF encodes a transcription factor belonging to the POZ/domain and Krüppel zinc finger (POK) family which interacts directly with PML. PLZF is related to another POK protein, LAZ3(BCL6), which is structurally altered, and presumably misexpressed, in many non-Hodgkin lymphoma (NHL) cases. PLZF and LAZ3 share many functional properties: both inhibit cell growth, concentrate into punctated nuclear subdomains and are sequence-specific transcriptional repressors recruiting a histone deacetylase-repressing complex. Given these similarities, we tested whether both proteins could be targeted by each other. Here, LAZ3 and PLZF are shown to colocalize onto nuclear dots. Moreover, truncated derivatives of one protein, which display a diffuse nuclear localization, are recruited onto nuclear dots by the full-length other. The colocalization and the reciprocal 'rescue' is the result of a direct interaction between LAZ3 and PLZF, as indicated by yeast two hybrid assays, in vitro immunoprecipitations, and GST pull down experiments. In contrast to LAZ3 homomerization, LAZ3/PLZF heteromerization in yeast does not solely depend on POZ/POZ contacts but rather also relies on interactions between the two zinc finger regions and 'cross' contacts between the zinc finger region and the POZ domain of each partner. Likewise, LAZ3 shows some colocalization with the PLZF partner PML upon stable overexpression of both proteins in CHO cells and interacts with PML in yeast. Finally, endogenous LAZ3 and PLZF are co-induced and partially colocalized in myeloid MDS cells. These data indicate that a physical interaction between LAZ3 and PLZF underlies their simultaneous recruitment onto multiproteic nuclear complexes, presumably involved in transcriptional silencing and whose integrity (for APL) and/or function (for APL and NHL) may be altered in oncogenesis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Linfoma não Hodgkin/metabolismo , Fatores de Transcrição/metabolismo , Dedos de Zinco , Animais , Células CHO , Células COS , Cricetinae , Proteínas de Ligação a DNA/genética , Imunofluorescência , Humanos , Fatores de Transcrição Kruppel-Like , Leucemia Promielocítica Aguda/genética , Linfoma não Hodgkin/genética , Substâncias Macromoleculares , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Proteína da Leucemia Promielocítica , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteínas Proto-Oncogênicas c-bcl-6 , Deleção de Sequência , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor , Técnicas do Sistema de Duplo-Híbrido
2.
Oncogene ; 18(5): 1209-17, 1999 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-10022127

RESUMO

We report the cloning and characterization of a novel member of the Transcriptional Intermediary Factor 1 (TIF1) gene family, human TIF1gamma. Similar to TIF1alpha and TIF1beta, the structure of TIF1beta is characterized by multiple domains: RING finger, B boxes, Coiled coil, PHD/TTC, and bromodomain. Although structurally related to TIF1alpha and TIF1beta, TIF1gamma presents several functional differences. In contrast to TIF1alpha, but like TIF1beta, TIF1 does not interact with nuclear receptors in yeast two-hybrid or GST pull-down assays and does not interfere with retinoic acid response in transfected mammalian cells. Whereas TIF1alpha and TIF1beta were previously found to interact with the KRAB silencing domain of KOX1 and with the HP1alpha, MODI (HP1beta) and MOD2 (HP1gamma) heterochromatinic proteins, suggesting that they may participate in a complex involved in heterochromatin-induced gene repression, TIF1gamma does not interact with either the KRAB domain of KOX1 or the HP1 proteins. Nevertheless, TIF1gamma, like TIF1alpha and TIF1beta, exhibits a strong silencing activity when tethered to a promoter. Since deletion of a novel motif unique to the three TIF1 proteins, called TIF1 signature sequence (TSS), abrogates transcriptional repression by TIF1gamma, this motif likely participates in TIF1 dependent repression.


Assuntos
Família Multigênica , Sequência de Aminoácidos , Sequência de Bases , Homólogo 5 da Proteína Cromobox , Bandeamento Cromossômico , Mapeamento Cromossômico , Clonagem Molecular , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Ligação Proteica , RNA Mensageiro/isolamento & purificação , Proteínas Recombinantes/biossíntese , Proteínas Repressoras/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Fatores de Transcrição/genética , Transcrição Gênica , Transfecção , Proteína 28 com Motivo Tripartido
3.
Oncogene ; 18(4): 1113-8, 1999 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-10023688

RESUMO

Primary blasts of a t(11;17)(q23;q21) acute promyelocytic leukaemia (APL) patient were analysed with respect to retinoic acid (RA) and arsenic trioxide (As2O3) sensitivity as well as PLZF/RARalpha status. Although RA induced partial monocytic differentiation ex vivo, but not in vivo, As203 failed to induce apoptosis in culture, contrasting with t(15;17) APL and arguing against the clinical use of As203 in t(11;17)(q23;q21) APL. Prior to cell culture, PLZF/RARalpha was found to exactly co-localize with PML onto PML nuclear bodies. However upon cell culture, it quickly shifted towards microspeckles, its localization found in transfection experiments. Arsenic trioxide, known to induce aggregation of PML nuclear bodies, left the microspeckled PLZF/RARalpha localization completely unaffected. RA treatment led to PLZF/RARalpha degradation. However, this complete PLZF/RARalpha degradation was not accompanied by differentiation or apoptosis, which could suggest a contribution of the reciprocal RARalpha/PLZF fusion product in leukaemogenesis or the existence of irreversible changes induced by the chimera.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/farmacologia , Proteínas de Ligação a DNA/efeitos dos fármacos , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Fusão Oncogênica/efeitos dos fármacos , Óxidos/farmacologia , Receptores do Ácido Retinoico/efeitos dos fármacos , Fatores de Transcrição/efeitos dos fármacos , Tretinoína/farmacologia , Apoptose , Trióxido de Arsênio , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Cromossomos Humanos Par 11 , Cromossomos Humanos Par 17 , Proteínas de Ligação a DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos , Imunofluorescência , Humanos , Fatores de Transcrição Kruppel-Like , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patologia , Proteínas de Fusão Oncogênica/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Fatores de Transcrição/metabolismo , Translocação Genética , Células Tumorais Cultivadas/efeitos dos fármacos
4.
Blood ; 91(11): 4300-10, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9596679

RESUMO

In the acute promyelocytic leukemia (APL) cell line NB4, as well as in APL patients' cells, arsenic trioxide (As2O3) leads to incomplete cell maturation, induction of apoptosis, as well as to the degradation of the oncogenic PML/RARalpha fusion protein. We have isolated an arsenic-resistant NB4 subline (NB4-AsR), which fails to undergo apoptosis, but maintains the partial differentiation response to this drug. When grown in the presence of As2O3, NB4-AsR cells degrade PML/RARalpha, slightly differentiate, and become more sensitive to serum deprivation-induced apoptosis. Similarly, in RA-resistant NB4-R1 cells, RA induced a significant PML/RARalpha degradation and yet failed to induce cell maturation. Thus, As2O3- or retinoic acid (RA)-induced PML/RARalpha degradation may be a prerequisite, but is not sufficient for the full differentiative/apoptotic response to these drugs. Strikingly, RA-triggered differentiation and apoptosis were greatly accelerated in As2O3-treated NB4-AsR cells. The synergism between these two agents in this setting could provide an experimental basis for combined or sequential RA/As2O3 therapies.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Arsenicais/farmacologia , Óxidos/farmacologia , Tretinoína/farmacologia , Trióxido de Arsênio , Diferenciação Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Humanos , Leucemia Promielocítica Aguda/patologia , Microscopia Confocal , Células Tumorais Cultivadas
5.
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
6.
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
7.
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
8.
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
9.
Cell ; 86(5): 799-810, 1996 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-8797826

RESUMO

The ubiquitin-conjugating yeast enzyme RAD6 and its human homologs hHR6A and hHR6B are implicated in postreplication repair and damage-induced mutagenesis. The yeast protein is also required for sporulation and may modulate chromatin structure via histone ubiquitination. We report the phenotype of the first animal mutant in the ubiquitin pathway: inactivation of the hHR6B-homologous gene in mice causes male infertility. Derailment of spermatogenesis becomes overt during the postmeiotic condensation of chromatin in spermatids. These findings provide a parallel between yeast sporulation and mammalian spermatogenesis and strongly implicate hHR6-dependent ubiquitination in chromatin remodeling. Since heterozygous male mice and even knockout female mice are completely normal and fertile and thus able to transmit the defect, similar hHR6B mutations may cause male infertility in man.


Assuntos
Cromatina/metabolismo , Infertilidade Masculina/genética , Ligases/genética , Espermatogênese/genética , Sequência de Aminoácidos , Animais , Apoptose , Peso Corporal , Proteínas Cromossômicas não Histona/análise , Reparo do DNA , Feminino , Histonas/análise , Humanos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Tamanho do Órgão , Fenótipo , Contagem de Espermatozoides , Espermátides/citologia , Espermatozoides/anormalidades , Espermatozoides/citologia , Testículo/química , Enzimas de Conjugação de Ubiquitina , Ubiquitinas/metabolismo
10.
Blood ; 88(4): 1423-6, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8695863

RESUMO

The PML gene is fused to the retinoic acid receptor alpha (RAR alpha) gene in t(15;17) acute promyelocytic leukemia (APL), creating a PML-RAR alpha fusion oncoprotein. The PML gene product has been localized to subnuclear dot-like structures variously termed PODs, ND10s, Kr bodies, or PML nuclear bodies (PML NBs). The present study describes the cloning of a lymphoid-restricted gene, LYSP100, that is homologous to another protein that localizes to PML NBs, SP100. In addition to SP100 homology regions, one LYSP100 cDNA isoform contains a bromodomain and a PHD/TTC domain, which are present in a variety of transcriptional regulatory proteins. By immunofluorescence, LYSP100 was localized to nuclear dots that were surprisingly largely nonoverlapping with PML NBs. However, a minority of LYSP100 nuclear dots exactly colocalized with PML and SP100. We term the LYSP100 structures "LANDs," for LYSP100-associated nuclear domains. Although LYSP100 is expressed only in lymphoid cells, LANDs could be visualized in HeLa cells by transfection of a LYSP100 cDNA. Immunoelectron microscopy revealed LANDs to be globular, electron-dense structures morphologically distinct from the annular structures characteristic of PML NBs. LANDs were most often found in the nucleoplasm, but were also found at the nuclear membrane and in the cytoplasm, suggesting that these structures may traffic between the cytoplasm and the nucleus. By double-immunogold labeling of PML and LYSP100, some LANDs were shown to contain both PML and LYSP100. Thus, PML is localized to a second subnuclear domain that is morphologically and biochemically distinct from PML NBs.


Assuntos
Antígenos Nucleares , Núcleo Celular/ultraestrutura , Linfócitos/ultraestrutura , Proteínas de Neoplasias , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Autoantígenos/química , Sequência de Bases , Compartimento Celular , Clonagem Molecular , Primers do DNA/química , Técnica Indireta de Fluorescência para Anticorpo , Células HeLa , Humanos , Substâncias Macromoleculares , Microscopia Confocal , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transfecção , Proteínas Supressoras de Tumor
11.
Dev Biol ; 173(1): 119-32, 1996 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-8575614

RESUMO

RAD6, a member of the expanding family of ubiquitin-conjugating (E2) enzymes, functions in the so-called "N-rule" protein breakdown pathway of Saccharomyces cerevisiae. In vitro, the protein can attach one or multiple ubiquitin (Ub) moieties to histones H2A and B and trigger their E3-dependent degradation. Rad6 mutants display a remarkably pleiotropic phenotype, implicating the protein in DNA damage-induced mutagenesis, postreplication repair, repression of retrotransposition, and sporulation. RAD6 transcription is strongly induced upon UV exposure and in meiosis, suggesting that it is part of a damage-induced response pathway and that it is involved in meiotic recombination. It is postulated that the protein exerts its functions by modulating chromatin structure. Previously, we have cloned two human homologs of this gene (designated HHR6A and HHR6B) and demonstrated that they partially complement the yeast defect. Here we present a detailed characterisation of their expression at the transcript and protein levels. Both HHR6 proteins, resolved by 2-dimensional immunoblot analysis, are expressed in all mammalian tissues and cell types examined, indicating that both genes are functional and constitutively expressed. Although the proteins are highly conserved, the UV induction present in yeast is not preserved, pointing to important differences in damage response between yeast and mammals. Absence of alterations in HHR6 transcripts or protein upon heat shock and during the cell cycle suggests that the proteins are not involved in stress response or cell cycle regulation. Elevated levels of HHR6 transcripts and proteins were found in testis. Enhanced HHR6 expression did not coincide with meiotic recombination but with the replacement of histones by transition proteins. Immunohistochemistry demonstrated that the HHR6 proteins are located in the nucleus, consistent with a functional link with chromatin. Electron microscopy combined with immunogold labeling revealed a preferential localisation of HHR6 in euchromatin areas, suggesting that the protein is associated with transcriptionally active regions. Our findings support the idea that both HHR6 genes have overlapping, constitutive functions related to chromatin conformation and that they have a specific role in spermatogenesis, involving Ub-mediated histone degradation.


Assuntos
Cromatina/metabolismo , Reparo do DNA , Regulação da Expressão Gênica , Ligases/biossíntese , Proteínas de Saccharomyces cerevisiae , Espermatogênese/fisiologia , Animais , Compartimento Celular , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Resposta ao Choque Térmico , Histonas/metabolismo , Humanos , Imuno-Histoquímica , Células Intersticiais do Testículo/química , Ligases/genética , Masculino , Camundongos , Mitose , RNA Mensageiro/análise , Ratos , Testículo/anatomia & histologia , Testículo/fisiologia , Distribuição Tecidual , Enzimas de Conjugação de Ubiquitina , Raios Ultravioleta
12.
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
13.
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
14.
Oncogene ; 11(12): 2689-97, 1995 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-8545127

RESUMO

The LAZ3/BCL6 gene was identified by its disruption in 3q27 translocations associated with diffuse large cell lymphomas. It is predicted to be a transcription factor as it contains six Krüppel-like Zinc finger motifs and a N-terminal BTB/POZ domain, a protein/protein interaction interface that is widely conserved in Metazoans. Using two antisera raised against non overlapping regions of the predicted ORF, we demonstrate that the LAZ3/BCL6 protein appears as a close ca. 79 kDa doublet in B lymphoid cell lines with either a rearranged or a non rearranged LAZ3/BCL6 locus. By immunofluorescence experiments on transiently transfected COS-1 or NIH3T3 cells, we show that the LAZ3/BCL6 protein displays a punctuated nuclear localisation. This appears to rely on LAZ3/BCL6 proper folding and/or activities as it is impaired in a hormone reversible-fashion through fusion of LAZ3/BCL6 to the ligand-binding domain of the oestrogen receptor. Moreover, deletion of its BTB/POZ domain leads to the disappearance of the nuclear dots although the protein remains nuclear. In addition, by using the yeast two-hybrid system, we show that the LAZ3/BCL6 BTB/POZ domain homomerises in vivo. Thus, the LAZ3/BCL6 BTB/POZ domain has the capability to self-interact and target the protein to discrete nuclear substructures.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fatores de Transcrição/fisiologia , Dedos de Zinco , Células 3T3 , Animais , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-bcl-6
15.
J Virol ; 69(9): 5261-8, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7636968

RESUMO

Human foamy retrovirus (HFV) is found as two proviruses (HFV and delta HFV) which differ by a splice-induced deletion within the bel1 transactivator gene. The defective delta HFV (which lacks a functional Bel1 but harbors an intronless bet gene) is predominantly found in nonlytic infections in vitro as well as in vivo. Here, we show that infection of cell lines stably transduced by delta HFV DNA with the highly lytic HFV leads to chronic infections characterized by an absence of lysis, a balanced ratio of HFV to delta HFV, and a persistent Bet expression accompanied by a shutoff of structural genes. While this system only partially reflects the natural situation, in which target cells are infected by HFV and delta HFV simultaneously, it strongly suggests that delta HFV is a defective interfering retrovirus. Accordingly, previous or concomitant exposure to delta HFV viruses greatly enhances the formation of lysis-resistant clones in culture after HFV infection. The inability of delta HFV proviruses encoding a mutated bet gene to induce chronic infection suggests a role for Bet in this process. Through a specific, splice-induced, genomic deletion, resulting in a switch from Bel1 to Bet expression, the lytic properties of HFV are progressively lost. Such programmed inactivation of a key gene represents a new regulatory mechanism of gene expression in retroviruses.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Vírus Defeituosos/genética , Genes Virais , Proteínas dos Retroviridae/biossíntese , Deleção de Sequência , Spumavirus/genética , Transativadores/biossíntese , Proteínas Estruturais Virais/genética , Sequência de Aminoácidos , Âmnio , Linhagem Celular , Mapeamento Cromossômico , Células Clonais , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Vírus Defeituosos/patogenicidade , Humanos , Íntrons , Dados de Sequência Molecular , Infecções por Retroviridae/fisiopatologia , Infecções por Retroviridae/virologia , Proteínas dos Retroviridae/química , Proteínas dos Retroviridae/genética , Homologia de Sequência de Aminoácidos , Spumavirus/patogenicidade , Fatores de Tempo , Transativadores/química , Transativadores/genética , Transfecção
16.
Dev Biol ; 170(2): 730-3, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7649399

RESUMO

During the meiotic prophase of spermatogenesis, the X and Y chromosomes form the heterochromatic sex body, showing little transcriptional activity. It has been suggested that transcription of the Xist gene is involved in this inactivation. After completion of the meiotic divisions, at least two Y chromosomal genes, Zfy and Sry, are transcribed in haploid spermatids. In contrast, postmeiotic transcription of X chromosomal genes has not been demonstrated. Using highly purified preparations of mouse pachytene spermatocytes, round spermatids, and cytoplasmic fragments from elongated spermatids, the present experiments show differential postmeiotic expression of the Y chromosomal genes Ubely and Sry, with highest mRNA levels in round spermatids and cytoplasmic fragments, respectively. Postmeiotic transcription of the X chromosomal gene Ube1x is indicated by an increased level of Ube1x mRNA in round spermatids and cytoplasmic fragments. The X chromosomal gene MHR6A shows a marked temporary postmeiotic expression in round spermatids. This postmeiotic activity of the X chromosome is a novel finding, which may have implications for our understanding of X chromosome inactivation during spermatogenesis and paternal genome imprinting.


Assuntos
Espermatogênese/genética , Cromossomo X , Cromossomo Y , Animais , Sequência de Bases , Primers do DNA/genética , Expressão Gênica , Masculino , Meiose/genética , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espermátides/metabolismo , Espermatócitos/metabolismo , Transcrição Gênica
17.
C R Acad Sci III ; 318(7): 733-9, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7583761

RESUMO

Metal-binding cysteine and histidine residues are often used to stabilise a protein fold through coordination of zinc ions. These zinc fingers are either involved in nucleic acid binding (TFIIIA, GAL4, nuclear receptors, retroviral gag...) or in yet unidentified biochemical functions (LIM and RING domains). The latter characterized by a unique histidine residue in the zinc binding motif (C2HC5 and C3HC4 for the LIM and RING respectively) may constitute protein/protein interaction interfaces. We have identified a new C4HC3 motif in a variety of proteins including the Drosophila trithorax and its human homologue ALL-1 involved in oncogenic translocations in acute leukaemias. This domain, for which we propose the name TTC (for trithorax consensus) is found in many transcriptional regulators or DNA-binding proteins. Interestingly, TTC was found in several bromodomain containing transcriptional adaptors including the E1A-binding p300 and the CREB-binding CBP proteins. In CBP, this domain does not appear to be involved in DNA, CREB or TFIIB binding. In the chromosomal translocations that involve the 11q23 locus, the C-terminal end of ALL-1 (which contains 4 TTC fingers) is constantly lost. The absence of these motifs in the fusion genes may relate to their leukemogenic potential.


Assuntos
Dedos de Zinco/genética , Sequência de Aminoácidos , Animais , Drosophila/química , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , Translocação Genética
18.
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
20.
EMBO J ; 13(5): 1073-83, 1994 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8131741

RESUMO

Nuclear bodies (NBs) are ultrastructurally defined granules predominantly found in dividing cells. Here we show that PML, a protein involved in the t(15;17) translocation of acute promyelocytic leukaemia (APL), is specifically bound to a NB. PML and several NB-associated proteins, found as auto-antigens in primary biliary cirrhosis (PBC), are co-localized and co-regulated. The APL-derived PML-RAR alpha fusion protein is shown to be predominantly localized in the cytoplasm, whereas a fraction is nuclear and delocalizes the NB antigens to multiple smaller nuclear clusters devoid of ultrastructural organization. RA administration (which in APL patients induces blast differentiation and consequently complete remissions) causes the re-aggregation of PML and PBC auto-antigens onto the NB, while PML-RAR alpha remains mainly cytoplasmic. Thus, PML-RAR alpha expression leads to a RA-reversible alteration of a nuclear domain. These results shed a new light on the pathogenesis of APL and provide a molecular link between NBs and oncogenesis.


Assuntos
Cromossomos Humanos Par 15 , Cromossomos Humanos Par 17 , Grânulos Citoplasmáticos/fisiologia , Leucemia Promielocítica Aguda/genética , Proteínas de Neoplasias , Proteínas Nucleares , Fatores de Transcrição/metabolismo , Translocação Genética , Tretinoína/farmacologia , Animais , Células CHO , Cricetinae , Grânulos Citoplasmáticos/efeitos dos fármacos , Humanos , Microscopia Imunoeletrônica , Proteína da Leucemia Promielocítica , Receptores do Ácido Retinoico/análise , Receptores do Ácido Retinoico/biossíntese , Receptores do Ácido Retinoico/metabolismo , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/análise , Fatores de Transcrição/biossíntese , Transfecção , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...