Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Virologie (Montrouge) ; 12(1): 39-52, 2008 Feb 01.
Artículo en Francés | MEDLINE | ID: mdl-36131433

RESUMEN

Discovered in 1957 for their antiviral properties, interferons (IFNs) are a growing cytokine family with diverse biological activities including antitumor and immunoregulatory activities. IFN are classified in three types I, II and III. They bind to different specific cell receptors and induce via the Jak/Stat pathway the expression of more than 300 genes, the products of which are believed to mediate their biological effects. Several proteins have been implicated in resistance to viral infection in IFN-treated cells, i.e. the dsRNAdependent protein kinase PKR, the 2'5' oligoadenylate synthetase/RNaseL and Mx proteins. However, it was demonstrated that cells from triple knockout mice lacking PKR, RNase L and Mx are still sensitive to the IFN-induced antiviral state, indicating that other pathways exist. One of these pathways implicates promyelocytic leukemia (PML) protein. This article reviews the potential antiviral activities of the different IFN-induced mediators focusing onPMLpathway and how viruses from different families overcome this defence.

2.
Cell Mol Biol (Noisy-le-grand) ; 52(1): 9-15, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16914093

RESUMEN

Interferons (IFNs) and arsenic trioxide (As2O3) are known inhibitors of cell proliferation and have been used in the treatment of certain forms of malignancy. IFNgamma treatment of cells leads to tyrosine phosphorylation of STAT1 followed by dimerization that accumulates in the nucleus. This is followed by DNA binding, activation of target gene transcription, dephosphorylation, and return to the cytoplasm. We have shown earlier that IFNgamma and As2O3 act synergistically in acute promyelocytic leukemia cells to upregulate IRF-1 expression and to induce apoptosis. Here, we show that in the human fibrosarcoma cell line 2fTGH, As2O3 prolongs IFNgamma-induced STAT1 phosphorylation resulting in persistent binding of STAT1 to GAS motif leading to an increase in IRF-1 expression which correlated with both higher anti-proliferative effect and increased apoptosis. These biological responses induced by IFNgamma alone or in combination with As2O3 were abolished when IRF-1 expression was down-regulated by RNA interference, thus demonstrating the key role of IRF-1.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Arsénico/farmacología , Factor 1 Regulador del Interferón/fisiología , Interferón gamma/farmacología , Secuencias de Aminoácidos , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proteínas Ligadas a GPI , Expresión Génica/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Humanos , Factor 1 Regulador del Interferón/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Células Tumorales Cultivadas
4.
Oncogene ; 20(49): 7274-86, 2001 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-11704856

RESUMEN

Interferons (IFNs) are a family of secreted proteins with antiviral, antiproliferative and immunomodulatory activities. The different biological actions of IFN are believed to be mediated by the products of specifically induced cellular genes in the target cells. The promyelocytic leukaemia (PML) protein localizes both in the nucleoplasm and in matrix-associated multi-protein complexes known as nuclear bodies (NBs). PML is essential for the proper formation and the integrity of the NBs. Modification of PML by the Small Ubiquitin MOdifier (SUMO) was shown to be required for its localization in NBs. The number and the intensity of PML NBs increase in response to interferon (IFN). Inactivation of the IFN-induced PML gene by its fusion to retinoic acid receptor alpha alters the normal localization of PML from the punctuate nuclear patterns of NBs to micro-dispersed tiny dots and results in uncontrolled growth in Acute Promyelocytic Leukaemia. The NBs-associated proteins, PML, Sp100, Sp140, Sp110, ISG20 and PA28 are induced by IFN suggesting that nuclear bodies could play a role in IFN response. Although the function of PML NBs is still unclear, some results indicate that they may represent preferential targets for viral infections and that PML could play a role in the mechanism of the antiviral action of IFNs. Viruses, which require the cellular machinery for their replication, have evolved different ways to counteract the action of IFN by inhibiting IFN signalling, by blocking the activities of specific antiviral mediators or by altering PML expression and/or localization on nuclear bodies.


Asunto(s)
Estructuras del Núcleo Celular/metabolismo , Infecciones por Virus ADN/metabolismo , Proteínas de Unión al GTP , Interferones/metabolismo , Proteínas de Neoplasias/metabolismo , Orgánulos/metabolismo , Infecciones por Virus ARN/metabolismo , Factores de Transcripción/metabolismo , Animales , Estructuras del Núcleo Celular/efectos de los fármacos , Enzimas/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Interferones/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Resistencia a Mixovirus , Proteínas Nucleares/metabolismo , Orgánulos/efectos de los fármacos , Proteína de la Leucemia Promielocítica , Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de Tumor
5.
EMBO J ; 20(13): 3495-505, 2001 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-11432836

RESUMEN

The promyelocytic leukaemia (PML) protein localizes in the nucleus both in the nucleoplasm and in matrix-associated multiprotein complexes known as nuclear bodies (NBs). The number and the intensity of PML NBs increase in response to interferon (IFN). Overexpression of PML affects the replication of vesicular stomatitis virus and influenza virus. However, PML has a less powerful antiviral activity against these viruses than the IFN mediator MxA. Here, we show that overexpression of PML, but not that of Mx1 or MxA, leads to a drastic decrease of a complex retrovirus, the human foamy virus (HFV), gene expression. PML represses HFV transcription by complexing the HFV transactivator, Tas, preventing its direct binding to viral DNA. This physical interaction requires the N-terminal region of Tas and the RING finger of PML, but does not necessitate PML localization in NBs. Finally, we show that IFN treatment inhibits HFV replication in wild-type but not in PML-/- cells. These findings point to a role for PML in transcriptional repression and suggest that PML could play a key role in mediating an IFN-induced antiviral state against a complex retrovirus.


Asunto(s)
Antivirales/metabolismo , Proteínas de Unión al GTP , Interferón-alfa/farmacología , Proteínas de Neoplasias/metabolismo , Proteínas/metabolismo , Spumavirus/fisiología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Astrocitoma , Células CHO , Cricetinae , ADN Viral/genética , Fibroblastos/fisiología , Fibroblastos/virología , Glioblastoma , Humanos , Células L , Ratones , Ratones Noqueados , Proteínas de Resistencia a Mixovirus , Proteínas de Neoplasias/genética , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , ARN Mensajero/genética , ARN Viral/genética , ADN Polimerasa Dirigida por ARN/metabolismo , Proteínas Recombinantes/metabolismo , Spumavirus/efectos de los fármacos , Spumavirus/genética , Factores de Transcripción/genética , Transfección , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Replicación Viral/efectos de los fármacos
6.
Oncogene ; 19(44): 5041-53, 2000 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-11042692

RESUMEN

Type I Interferon (IFN) and all-trans retinoic acid (RA) inhibit cell proliferation of squamous carcinoma cell lines (SCC). Examinations of growth-affected cell populations show that SCC lines ME-180 and SiHa treated with IFN-beta undergo a specific slower progression through the S phase that seems to trigger cellular death. In combination treatment RA potentiates IFN-beta effect in SCC ME-180 but not in SiHa cell line, partially resistant to RA antiproliferative action. RA added as single agent affects cell proliferation differently by inducing a slight G1 accumulation. The IFN-beta-induced S phase lengthening parallels the increased expression of PML, a nuclear phosphoprotein specifically up-regulated at transcriptional level by IFN, whose overexpression induces cell growth inhibition and tumor suppression. We report that PML up-regulation may account for the alteration of cell cycle progression induced by IFN-beta in SCC by infecting cells with PML-PINCO recombinant retrovirus carrying the PML-3 cDNA under the control of the 5' LTR. In fact PML overexpression reproduces the IFN-beta-induced S phase lengthening. These findings provide important insight into the mechanism of tumor suppressing function of PML and could allow PML to be included in the pathways responsible for IFN-induced cell growth suppression.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/patología , Interferón Tipo I/farmacología , Proteínas de Neoplasias/biosíntesis , Proteínas Nucleares , Fase S/efectos de los fármacos , Factores de Transcripción/biosíntesis , Neoplasias del Cuello Uterino/patología , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , División Celular/efectos de los fármacos , ADN de Neoplasias/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Crecimiento/administración & dosificación , Inhibidores de Crecimiento/farmacología , Humanos , Interferón Tipo I/administración & dosificación , Proteínas de Neoplasias/genética , Proteína de la Leucemia Promielocítica , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Proteínas Recombinantes , Factores de Transcripción/genética , Tretinoina/administración & dosificación , Tretinoina/farmacología , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Regulación hacia Arriba/efectos de los fármacos , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo
7.
Leukemia ; 14(4): 716-21, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10764160

RESUMEN

The combination of the anti-viral agents, zidovudine (AZT) and interferon-alpha (IFN), is a potent treatment of HTLV-I-associated adult T cell leukemia/lymphoma (ATL). In this study we investigate the possible mechanism of action of this combination by examining several cellular parameters including cell proliferation, cell cycle distribution and apoptosis. The ATL-derived T cell lines HuT-102 and MT-2 served as models. HTLV-I negative T cell lines (CEM and Jurkat) were used as controls. No significant modification of cell growth was observed except at suprapharmacological doses of AZT and IFN. Moreover, these effects were less pronounced in HTLV-I-infected cell lines compared to control cell lines. AZT and IFN treatment did not induce any significant modification of the expression of bcl-2 and p53. Interestingly no in vitro cytotoxic effect of AZT/IFN combination was observed on fresh leukemic cells derived from an acute ATL patient at diagnosis despite achievement of in vivo complete remission using the same therapy. These results suggest that the therapeutic effect of AZT and IFN is not through a direct cytotoxic effect of these drugs on the leukemic cells.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Factores Inmunológicos/farmacología , Interferón-alfa/farmacología , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Zidovudina/farmacología , Anciano , Antimetabolitos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/análisis , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , ADN de Neoplasias/análisis , Sinergismo Farmacológico , Femenino , Virus Linfotrópico T Tipo 1 Humano/aislamiento & purificación , Humanos , Factores Inmunológicos/uso terapéutico , Interferón-alfa/uso terapéutico , Células Jurkat/efectos de los fármacos , Leucemia-Linfoma de Células T del Adulto/patología , Inducción de Remisión , Células Tumorales Cultivadas/efectos de los fármacos , Zidovudina/uso terapéutico
8.
Leukemia ; 13(8): 1167-74, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10450744

RESUMEN

Retinoic acid (RA) and interferon (IFN) potentiate each other to induce biological responses. Their combination has shown synergistic differentiating, antiproliferative and antiviral activities in various cell lines including those derived from the acute promyelocytic leukemia (APL). IFNs have demonstrated broad applications in cancer, as well as in virologic diseases. RA has variable effectiveness in therapy. Its real success is in APL where it provides the first example of a differentiation therapy. However, complete clinical remission with RA alone is always transient as RA resistance develops in the treated patients as well as in vitro. In various cell lines, including those derived from APL, RA induces directly the expression of two transcription factors, Stat1 and IRF-1 which play central roles in the IFN signal transduction. In addition, RA induces IFN-alpha synthesis and enhances the IFN-induced Stat activation. Here, we review the molecular mechanisms by which RA and IFNs can cooperate in inducing differentiation, inhibition of cell growth or viral replication focusing on recent results derived from normal and RA-resistant APL cells.


Asunto(s)
Antineoplásicos/farmacología , Interferones/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Receptores de Interferón/metabolismo , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Tretinoina/farmacología , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Interferones/uso terapéutico , Receptor Cross-Talk , Tretinoina/uso terapéutico
9.
Oncogene ; 18(27): 3944-53, 1999 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-10435617

RESUMEN

In the t(15;17) acute promyelocytic leukaemia (APL), all trans-retinoic (RA) treatment induces maturation leading to clinically complete but not durable remission, as RA resistance develops in the treated patients as well as in vitro. RA and interferons (IFNs) are known inhibitors of proliferation in various cells including those from APL. In this report, we show that they can act cooperatively to inhibit growth and to induce differentiation of NB4 cells but not of two RA-resistant NB4 derived cell lines, NB4-R1 and NB4-R2. However, the resistant cell lines respond to IFN. In NB4 cells, RA increases the expression of Stat1, p48 and IRF-1, three transcription factors playing a central role in the IFN response and induces the synthesis and the secretion of IFN alpha. RA-induced IFN alpha seems to play a role in inhibition of NB4 cell growth but not in their differentiation. In the resistant cells, NB4-R1 and NB4-R2, both the induction of IFN and the increase of Statl and p48 expression by RA are completely blocked. In contrast, IRF-1 mRNA and protein expressions are induced in the three cell lines. This suggests that increase of IRF-1 expression is not sufficient for IFN induction. Our results identify some defects linked to RA-resistance in APL and support the hypothesis that RA-induced Stat1 expression and IFN secretion may be one of the mechanisms mediating growth inhibition by RA.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Interferón-alfa/biosíntesis , Leucemia Promielocítica Aguda/metabolismo , Transactivadores/biosíntesis , Factores de Transcripción/biosíntesis , Tretinoina/farmacología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Inhibidores de Crecimiento/fisiología , Humanos , Factor 1 Regulador del Interferón , Factor 3 Regulador del Interferón , Factor 3 de Genes Estimulados por el Interferón , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón , Interferón-alfa/farmacología , Interferón-alfa/fisiología , Leucemia Promielocítica Aguda/patología , Fosfoproteínas/biosíntesis , Fosfoproteínas/metabolismo , Fosforilación , Factor de Transcripción STAT1 , Transducción de Señal/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
10.
Oncogene ; 18(4): 935-41, 1999 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-10023669

RESUMEN

The PML protein is associated to nuclear bodies (NBs) whose functions are as yet unknown. PML and two other NBs-associated proteins, Sp100 And ISG20 are directly induced by interferons (IFN). PML and Sp100 proteins are covalently linked to SUMO-1, and ubiquitin-like peptide. PML NBs are disorganized in acute promyelocytic leukemia and during several DNA virus infections. In particular, the HSV-1 ICP0 protein is known to delocalize PML from NBs. Thus, NBs could play an important role in oncogenesis, IFN response and viral infections. Here, we show that HSV-1 induced PML protein degradation without altering its mRNA level. This degradation was time- and multiplicity of infection-dependent. Sp100 protein was also degraded, while another SUMO-1 conjugated protein, RanGAP1 and the IFN-induced protein kinase PKR were not. The proteasome inhibitor MG132 abrogated the HSV-1-induced PML and Sp100 degradation and partially restored their NB-localization. HSV-1 induced PML and Sp100 degradation constitutes a new example of viral inactivation of IFN target gene products.


Asunto(s)
Antígenos Nucleares , Autoantígenos/metabolismo , Proteínas Activadoras de GTPasa , Herpes Simple/metabolismo , Herpesvirus Humano 1 , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Autoantígenos/química , Proteínas Portadoras/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Interferones/farmacología , Leupeptinas/farmacología , Peso Molecular , Proteínas de Neoplasias/química , Proteínas Nucleares/química , Proteína de la Leucemia Promielocítica , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Proteína SUMO-1 , Factores de Tiempo , Factores de Transcripción/química , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Ubiquitinas/metabolismo
11.
Biol Cell ; 91(8): 617-28, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10629941

RESUMEN

The present study has documented changes in the in situ distribution of viral DNA and capsid proteins in 293 cells infected with fiber gene-deleted adenoviruses. It shows that infection results in the intense production of progeny viruses which appear morphologically intact although they are devoid of fiber-coding sequence in their genome and hence of fiber protein in their capsid. The data confirm, therefore, that fiber protein is not essential for the assembly of progeny viruses. The main contribution of our observations concerns specific intranuclear structures induced by infection with either wild-type or fiber gene-deleted viruses. These clear amorphous inclusions contain two cellular proteins, PML and Sp100, which in non-infected cells co-localize to a special type of nuclear bodies. PML and Sp100 nuclear bodies appear to directly modulate or to be altered in a wide variety of situations including viral infections, cell death and transformation. In cells infected with fiber gene-deleted viruses, the clear amorphous inclusions now accumulate non-used hexon and penton base proteins, whereas the absence of fiber protein prevents the assembly of capsid proteins in crystallin arrays. Taken together, the data suggest that the clear amorphous inclusions may correspond to storage sites of structural and regulatory proteins. Consequently, these virus-induced structures may promote the productive cycle of adenoviruses by regulating the amount of over-produced viral proteins and the shutoff of the host cell metabolism.


Asunto(s)
Infecciones por Adenoviridae/genética , Adenoviridae/genética , Antígenos Nucleares , Autoantígenos/análisis , Proteínas de la Cápside , Cápside/metabolismo , Eliminación de Gen , Proteínas de Neoplasias/análisis , Proteínas Nucleares/análisis , Factores de Transcripción/análisis , Adenoviridae/ultraestructura , Núcleo Celular/química , Núcleo Celular/ultraestructura , Núcleo Celular/virología , Células Cultivadas , ADN Viral/análisis , ADN Viral/genética , Inmunohistoquímica , Hibridación in Situ , Cuerpos de Inclusión/química , Cuerpos de Inclusión/ultraestructura , Microscopía Electrónica , Proteínas Supresoras de Tumor
12.
Bull Cancer ; 85(4): 313-8, 1998 Apr.
Artículo en Francés | MEDLINE | ID: mdl-9752294

RESUMEN

Retinoic acid (RA) and interferons (IFN) are negative regulators of cell proliferation. A number of clinical trials were thus carried out in cancer therapy with RA and/or IFN. In vitro and in vivo, their combination leads to a more potent cell growth inhibition. Moreover, RA and IFN act cooperatively to increase the expression of many IFN-stimulated genes, leading also to a higher cell differentiation and inhibition of viral replication. However, the molecular mechanisms by which RA and IFN potentiate each other are not fully understood. The cooperative effects by RA and IFN are mediated through multiple pathways. RA causes the induction and secretion of IFN alpha. RA also stimulates the IFN regulatory factor gene expression (IRF1 and p48). Additional mechanisms could be involved as RA increases the level of signal transducing activators of transcription (Stat) proteins, and thus enhances the IFN-induced Stat activation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Interferones/uso terapéutico , Tretinoina/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Interacciones Farmacológicas , Humanos , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Interferones/administración & dosificación , Interferones/clasificación , Interferones/metabolismo , Fosforilación , Transducción de Señal , Transactivadores/metabolismo , Tretinoina/administración & dosificación , Tretinoina/metabolismo
13.
Blood ; 91(11): 4300-10, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9596679

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Óxidos/farmacología , Tretinoina/farmacología , Trióxido de Arsénico , Diferenciación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Leucemia Promielocítica Aguda/patología , Microscopía Confocal , Células Tumorales Cultivadas
14.
J Virol ; 72(2): 1043-51, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9444998

RESUMEN

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.


Asunto(s)
Antígenos Nucleares , Virus de la Influenza A , Gripe Humana/metabolismo , Interferones/farmacología , Proteínas de Neoplasias , Infecciones por Rhabdoviridae/metabolismo , Factores de Transcripción/metabolismo , Virus de la Estomatitis Vesicular Indiana , Animales , Autoantígenos/metabolismo , Células CHO , Cricetinae , Susceptibilidad a Enfermedades , Humanos , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor
15.
Oncogene ; 15(19): 2349-59, 1997 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-9393879

RESUMEN

Retinoic acid (RA) and interferons (IFNs) are negative regulators of cell proliferation. In vitro and in vivo, their combination leads to a more potent growth inhibition. However, the molecular mechanisms by which RA and IFNs potentiate each other are not fully understood. As some IFN-induced gene products regulate cell growth and/or antiviral activity, we analysed the effects of RA on their expressions. RA increases the level of 2'5'oligoadenylate synthetase, p68 kinase, the promyelocytic leukemia protein (PML) and Sp100 in both HL-60 and WISH cells. Moreover, RA and IFN act cooperatively to increase the expression of these proteins. RA also inhibits vesicular stomatitis virus replication and induces a higher antiviral state and growth inhibition when combined with IFN. RA stimulates the IFN regulatory factor 1 (IRF-1) gene expression directly through the GAS motif and causes the induction and secretion of IFNalpha. Additional mechanisms could be involved as RA increases the level of signal transducing activators of transcription (STAT) proteins, and enhances the IFN-induced STAT activation, suggesting that cooperative effects by RA and IFN are mediated through multiple pathways.


Asunto(s)
Antígenos Nucleares , Interferón-alfa/farmacología , Tretinoina/farmacología , 2',5'-Oligoadenilato Sintetasa/metabolismo , Autoantígenos/metabolismo , División Celular/efectos de los fármacos , Línea Celular , Sinergismo Farmacológico , Células HL-60 , Células HeLa , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 94(19): 10255-60, 1997 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-9294197

RESUMEN

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.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias , Proteínas Nucleares , Receptores de Ácido Retinoico/metabolismo , Factores de Transcripción/metabolismo , Animales , Células COS , Dimerización , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo , Receptor alfa de Ácido Retinoico , Proteínas Supresoras de Tumor
17.
Proc Natl Acad Sci U S A ; 94(8): 3978-83, 1997 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9108090

RESUMEN

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.


Asunto(s)
Arsénico/farmacología , Núcleo Celular/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Apoptosis/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Humanos , Leucemia Promielocítica Aguda/patología , Leucemia Promielocítica Aguda/terapia
18.
Exp Cell Res ; 229(2): 253-60, 1996 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-8986606

RESUMEN

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.


Asunto(s)
Enfermedades Autoinmunes/genética , Leucemia Promielocítica Aguda/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares , Proteínas de Fusión Oncogénica/genética , Factores de Transcripción/genética , Animales , Anticuerpos Antinucleares , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales/farmacología , Autoantígenos , Enfermedades Autoinmunes/tratamiento farmacológico , Células CHO , Diferenciación Celular/efectos de los fármacos , Cricetinae , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/etiología , Proteínas de Neoplasias/inmunología , Proteínas de Fusión Oncogénica/inmunología , Óxidos/farmacología , Proteína de la Leucemia Promielocítica , Transfección , Tretinoina/farmacología , Proteínas Supresoras de Tumor
19.
Leukemia ; 9(12): 2027-33, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8609713

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al GTP , Interferones/farmacología , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias , Proteínas/metabolismo , Factores de Transcripción/biosíntesis , Western Blotting , Humanos , Microscopía Confocal , Proteínas de Resistencia a Mixovirus , Proteínas Nucleares/biosíntesis , Proteína de la Leucemia Promielocítica , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor
20.
Oncogene ; 11(12): 2565-73, 1995 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-8545113

RESUMEN

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.


Asunto(s)
Interferones/farmacología , Proteínas de Neoplasias , Proteínas Nucleares , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células CHO , Cricetinae , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteína de la Leucemia Promielocítica , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA