Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO J ; 20(16): 4478-89, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11500375

RESUMEN

The Fanconi anemia (FA) complementation group C gene product (FANCC) functions to protect hematopoietic cells from cytotoxicity induced by interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha) and double-stranded RNA (dsRNA). Because apoptotic responses of mutant FA-C cells involve activation of interferon-inducible, dsRNA-dependent protein kinase PKR, we sought to identify FANCC-binding cofactors that may modulate PKR activation. We identified the molecular chaperone Hsp70 as an interacting partner of FANCC in lymphoblasts and HeLa cells using 'pull-down' and co-immunoprecipitation experiments. In vitro binding assays showed that the association of FANCC and Hsp70 involves the ATPase domain of Hsp70 and the central 320 residues of FANCC, and that both Hsp40 and ATP/ADP are required. In whole cells, Hsp70-FANCC binding and protection from IFN-gamma/TNF-alpha-induced cytotoxicity were blocked by alanine mutations located in a conserved motif within the Hsp70-interacting domain of FANCC. We therefore conclude that FANCC acts in concert with Hsp70 to prevent apoptosis in hematopoietic cells exposed to IFN-gamma and TNF-alpha.


Asunto(s)
Apoptosis , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Anemia de Fanconi/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas Nucleares , Proteínas/metabolismo , Adenosina Trifosfatasas/metabolismo , Sitios de Unión , Línea Celular , Citotoxicidad Inmunológica , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Proteínas del Choque Térmico HSP40 , Células HeLa , Proteínas de Choque Térmico/metabolismo , Humanos , Interferón gamma/farmacología , Proteínas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología , eIF-2 Quinasa
2.
Blood ; 98(5): 1392-401, 2001 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-11520787

RESUMEN

The Fanconi anemia (FA) group C gene product (FANCC) functions to protect cells from cytotoxic and genotoxic effects of cross-linking agents. FANCC is also required for optimal activation of STAT1 in response to cytokine and growth factors and for suppressing cytokine-induced apoptosis by modulating the activity of double-stranded RNA-dependent protein kinase. Because not all FANCC mutations affect STAT1 activation, the hypothesis was considered that cross-linker resistance function of FANCC depends on structural elements that differ from those required for the cytokine signaling functions of FANCC. Structure-function studies were designed to test this notion. Six separate alanine-substituted mutations were generated in 3 highly conserved motifs of FANCC. All mutants complemented mitomycin C (MMC) hypersensitive phenotype of FA-C cells and corrected aberrant posttranslational activation of FANCD2 in FA-C mutant cells. However, 2 of the mutants, S249A and E251A, failed to correct defective STAT1 activation. FA-C lymphoblasts carrying these 2 mutants demonstrated a defect in recruitment of STAT1 to the interferon gamma (IFN-gamma) receptor and GST-fusion proteins bearing S249A and E251A mutations were less efficient binding partners for STAT1 in stimulated lymphoblasts. These same mutations failed to complement the characteristic hypersensitive apoptotic responses of FA-C cells to tumor necrosis factor-alpha (TNF-alpha) and IFN-gamma. Cells bearing a naturally occurring FANCC mutation (322delG) that preserves this conserved region showed normal STAT1 activation but remained hypersensitive to MMC. The conclusion is that a central highly conserved domain of FANCC is required for functional interaction with STAT1 and that structural elements required for STAT1-related functions differ from those required for genotoxic responses to cross-linking agents. Preservation of signaling capacity of cells bearing the del322G mutation may account for the reduced severity and later onset of bone marrow failure associated with this mutation.


Asunto(s)
Proteínas de Ciclo Celular , Anemia de Fanconi/genética , Proteínas Nucleares , Proteínas/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Apoptosis/efectos de los fármacos , Línea Celular Transformada/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Prueba de Complementación Genética , Humanos , Interferón gamma/farmacología , Mitomicina/farmacología , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas/genética , Proteínas/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Factor de Transcripción STAT1 , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Relación Estructura-Actividad , Transactivadores/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
3.
Blood ; 97(10): 3017-24, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11342426

RESUMEN

Because hematopoietic cells derived from Fanconi anemia (FA) patients of the C-complementation group (FA-C) are hypersensitive to the inhibitory effects of interferon gamma (IFNgamma), the products of certain IFNgamma-inducible genes known to influence hematopoietic cell survival were quantified. High constitutive expression of the IFNgamma-inducible genes, IFN-stimulated gene factor 3 gamma subunit (ISGF3gamma), IFN regulatory factor-1 (IRF-1), and the cyclin-dependent kinase inhibitor p21(WAF1) was found in FANCC mutant B lymphoblasts, low-density bone marrow cells, and murine embryonic fibroblasts. Paradoxically, these cells do not activate signal transducer and activator of transcription (STAT) 1 properly. In an attempt to clarify mechanisms by which FA-C cells overexpress IFNgamma-inducible genes in the face of defective STAT1 phosphorylation, it was reasoned that decreased levels of activated STAT1 might result in reduced expression of a hematopoietic IFNgamma-responsive protein that normally modulates expression of other IFNgamma-responsive genes. Levels of the IFNgamma-inducible factor IFN consensus sequence binding protein (ICSBP), a negative trans-acting regulator of some IFNgamma-inducible genes, were quantified. ICSBP levels were reduced in FA-C B lymphoblasts and MEFs. However, enforced expression of ICSBP failed to down-regulate IRF-1, ISGF3gamma, and p21(WAF1). Thus, the FANCC protein functions to modulate expression of a family of genes that in normal cells are inducible only by specific environmental cues for apoptosis or mitogenic inhibition, but it does so independently of the classic IFN-STAT1 pathway and is not the direct result of reduced ICSBP expression.


Asunto(s)
Proteínas de Ciclo Celular , Anemia de Fanconi/genética , Regulación de la Expresión Génica , Interferón gamma/farmacología , Proteínas Nucleares , Proteínas/genética , Animales , Linfocitos B/metabolismo , Células de la Médula Ósea/metabolismo , Línea Celular Transformada , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Anemia de Fanconi/patología , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Fibroblastos/química , Células Madre Hematopoyéticas/patología , Herpesvirus Humano 4 , Humanos , Factor 1 Regulador del Interferón , Factores Reguladores del Interferón , Factor 3 de Genes Estimulados por el Interferón , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón , Ratones , Ratones Noqueados , Mutación , Fosfoproteínas/genética , Fosforilación , Proteínas/fisiología , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras/análisis , Factor de Transcripción STAT1 , Transactivadores/metabolismo , Factores de Transcripción/genética , Transfección
4.
Blood ; 97(6): 1644-52, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11238103

RESUMEN

Hematopoietic cells bearing inactivating mutations of Fanconi anemia group C (FANCC) are excessively apoptotic and demonstrate hypersensitivity not only to cross-linking agents but also to interferon gamma (IFN-gamma) and tumor necrosis factor-alpha. Seeking essential signaling pathways for this phenotype, this study quantified constitutive and induced RNA-dependent protein kinase (PKR) activation in Fanconi anemia cells of the C complementation group (FA-C). PKR was constitutively phosphorylated and exhibited an increased binding affinity for double-stranded RNA (dsRNA) in FANCC(-/-) cells. FANCC(-/-) cells were hypersensitive to both dsRNA and the combination of dsRNA and IFN-gamma in that these agents induced a higher fraction of apoptosis in FANCC(-/-) cells than in normal cells. Overexpression of wild-type PKR-sensitized FANCC(-/-) cells to apoptosis induced by IFN-gamma and dsRNA. Conversely, inhibition of PKR function by enforced expression of a dominant-negative inhibitory mutant of PKR (PKRDelta6) substantially reduced the IFN and dsRNA hypersensitivity of FANCC(-/-) cells. Two PKR target molecules, IkappaB-alpha and IRF-1, were not differentially activated in FANCC(-/-) cells, but enforced expression of a nonphosphorylatable form of eukaryotic translation initiation factor-2alpha reversed the PKR-mediated block of messenger RNA translation and partially abrogated the PKR-mediated apoptosis in FANCC(-/-) cells. Because no evidence was found of a PKR/FANCC complex in normal cells, it was concluded that an essential function of FANCC is to suppress, indirectly, the activity of PKR and that FANCC inactivation results in IFN hypersensitivity, at least in part, because this function of FANCC is abrogated.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Anemia de Fanconi/enzimología , Hipersensibilidad/enzimología , Interferón gamma/farmacología , Proteínas Nucleares , Proteínas/genética , ARN Bicatenario/farmacología , Factor de Necrosis Tumoral alfa/farmacología , eIF-2 Quinasa/fisiología , Animales , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 2 Eucariótico de Iniciación/fisiología , Anemia de Fanconi/etiología , Anemia de Fanconi/patología , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Fibroblastos , Hipersensibilidad/etiología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas/farmacología , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo
5.
Blood ; 96(13): 4204-11, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11110692

RESUMEN

Hematopoietic progenitor cells (HPC) from mice nullizygous at the Fanconi anemia (FA) group C locus and children with Fanconi anemia group C (FA-C) are hypersensitive to interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha. This hypersensitivity results, in part, from the capacity of these cytokines to prime the fas pathway. Because fas-mediated programmed cell death in many cells involves sequential activation of specific caspases, we tested the hypothesis that programmed cell death in FA HPC involves the ordered activation of specific caspase molecules. Lysates from lymphoblasts treated with both agonistic anti-fas antibody and IFN-gamma contained activated caspase 3 family members (caspases 3, 6, and 7), as well as caspase 8, whereas activation of caspases 1, 2, 4, 9, and 10 was not detected. The apoptotic effects of fas agonists in IFN-gamma-treated human and murine FA-C cells were blocked when pretreated with inhibitors (ac-DEVD-cho, CP-DEVD-cho, Z-DEVD-FMK) of the caspase 3 protease. Inhibitors (ac-YVAD-cho, CP-YVAD-cho, Z-YVAD-FMK) of caspase 1 did not block apoptosis or caspase 3 activation. Treatment of FA cells with the fluoromethyl ketone tetrapeptide caspase 8 inhibitor (ac-IETD-FMK) did suppress caspase 3 activation. A 4-fold greater fraction of IFN-induced FA-C cells expressed caspase 3 than FA-C cells complemented by retroviral-mediated transfer of FANCC. Therefore fas-induced apoptosis in Fanconi anemia cells of the C type involves the activation of caspase 8, which controls activation of caspase 3 family members and one direct or indirect function of the FANCC protein is to suppress apoptotic responses to IFN-gamma upstream of caspase 3 activation. (Blood. 2000;96:4204-4211)


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Caspasas/fisiología , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Anemia de Fanconi/patología , Células Madre Hematopoyéticas/efectos de los fármacos , Interferón gamma/farmacología , Isoenzimas/metabolismo , Proteínas Nucleares , Proteínas/fisiología , Animales , Médula Ósea/patología , Caspasa 3 , Caspasa 8 , Caspasa 9 , Inhibidores de Caspasas , Línea Celular Transformada , Células Cultivadas , Niño , Inhibidores de Cisteína Proteinasa/farmacología , Activación Enzimática/efectos de los fármacos , Anemia de Fanconi/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Heterogeneidad Genética , Células Madre Hematopoyéticas/enzimología , Humanos , Isoenzimas/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas/genética , Proteínas Recombinantes de Fusión/fisiología , Proteínas Recombinantes , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato , Transfección , Receptor fas/fisiología
6.
Mol Cell Biol ; 20(13): 4724-35, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10848598

RESUMEN

Hematopoietic progenitor cells from Fanconi anemia (FA) group C (FA-C) patients display hypersensitivity to the apoptotic effects of gamma interferon (IFN-gamma) and constitutively express a variety of IFN-dependent genes. Paradoxically, however, STAT1 activation is suppressed in IFN-stimulated FA cells, an abnormality corrected by transduction of normal FANCC cDNA. We therefore sought to define the specific role of FANCC protein in signal transduction through receptors that activate STAT1. Expression and phosphorylation of IFN-gamma receptor alpha chain (IFN-gammaRalpha) and JAK1 and JAK2 tyrosine kinases were equivalent in both normal and FA-C cells. However, in coimmunoprecipitation experiments STAT1 did not dock at the IFN-gammaR of FA-C cells, an abnormality corrected by transduction of the FANCC gene. In addition, glutathione S-transferase fusion genes encoding normal FANCC but not a mutant FANCC bearing an inactivating point mutation (L554P) bound to STAT1 in lysates of IFN-gamma-stimulated B cells and IFN-, granulocyte-macrophage colony-stimulating factor- and stem cell factor-stimulated MO7e cells. Kinetic studies revealed that the initial binding of FANCC was to nonphosphorylated STAT1 but that subsequently the complex moved to the receptor docking site, at which point STAT1 became phosphorylated. The STAT1 phosphorylation defect in FA-C cells was functionally significant in that IFN induction of IFN response factor 1 was suppressed and STAT1-DNA complexes were not detected in nuclear extracts of FA-C cells. We also determined that the IFN-gamma hypersensitivity of FA-C hematopoietic progenitor cells does not derive from STAT1 activation defects because granulocyte-macrophage CFU and erythroid burst-forming units from STAT1(-/-) mice were resistant to IFN-gamma. However, BFU-E responses to SCF and erythropoietin were suppressed in STAT(-/-) mice. Consequently, because the FANCC protein is involved in the activation of STAT1 through receptors for at least three hematopoietic growth and survival factor molecules, we reason that FA-C hematopoietic cells are excessively apoptotic because of an imbalance between survival cues (owing to a failure of STAT1 activation in FA-C cells) and apoptotic and mitogenic inhibitory cues (constitutively activated in FA-C cells in a STAT1-independent fashion).


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Sustancias de Crecimiento/metabolismo , Interferón gamma/metabolismo , Proteínas Nucleares , Proteínas/metabolismo , Proteínas Proto-Oncogénicas , Transactivadores/metabolismo , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Línea Celular/efectos de los fármacos , Proteínas de Unión al ADN/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Sustancias de Crecimiento/farmacología , Humanos , Factor 1 Regulador del Interferón , Interferón gamma/farmacología , Janus Quinasa 1 , Janus Quinasa 2 , Ratones , Ratones Noqueados , Mutación , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Pruebas de Precipitina , Proteínas Tirosina Quinasas/metabolismo , Proteínas/genética , Receptores de Interferón/efectos de los fármacos , Receptores de Interferón/metabolismo , Factor de Transcripción STAT1 , Transactivadores/genética , Receptor de Interferón gamma
8.
Cancer Res ; 58(22): 5123-9, 1998 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-9823322

RESUMEN

We identified an NH2-terminally truncated HER-2/neu product of M(r) 95,000 with in vitro kinase activity by Western blotting and immunoprecipitations using domain-specific antibodies. p95 levels correlated with the extracellular domain (ECD) shed from different cells under varied conditions. Both ECD and p95 were at approximately 20-fold lower levels in SKOV3 ovarian carcinoma cells, as compared to BT474 breast carcinoma cells. Both were stimulated by treatment of cells with the phorbol ester tumor promoter phorbol 12-myristate 13-acetate and the lysosomotrophic agent chloroquine. The hydroxamate inhibitor of metalloproteases, TAPI, suppressed both p95 and ECD in a dose-dependent fashion, with maximal inhibition at < or = 10 microM in BT474 cells. Cancer tissues were analyzed by Western blotting and scored for p95HER-2/neu and for p185HER-2/neu expression. Breast and ovarian cancer tissues were both found to express p95HER-2/neu in addition to p185HER-2/neu. Of 161 breast cancer tissues, 22.4% expressed p95, 21.7% overexpressed p185, and 14.3% were p95 positive and overexpressed p185. A higher proportion of node-positive patients (23 of 78) than node-negative patients (9 of 63) expressed p95 in all tumors combined (P = 0.032). In the group that overexpressed p185, those that contained p95 were associated with node-positive patients (15 of 21), whereas those that were p95 negative were associated with node-negative patients (8 of 11; P = 0.017). Neither p95- nor p185-rich patients significantly correlated with tumor size or with hormone receptor status in this study. Our findings show that breast cancers, which express the HER-2/neu oncogene, are heterogeneous with respect to HER-2/neu protein products. p95HER-2/neu appears to distinguish tumors that have metastasized to the lymph nodes from those in node-negative patients.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Neoplasias/química , Receptor ErbB-2/química , Células 3T3 , Animales , Neoplasias de la Mama/química , Neoplasias de la Mama/patología , Dipéptidos/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Humanos , Ácidos Hidroxámicos/farmacología , Metaloendopeptidasas/antagonistas & inhibidores , Ratones , Peso Molecular , Proteínas de Neoplasias/análisis , Fosforilación , Pronóstico , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor ErbB-2/análisis , Células Tumorales Cultivadas
9.
Blood ; 91(4): 1418-25, 1998 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9454773

RESUMEN

The Fanconi anemia (FA) complementation group C (FAC) protein gene encodes a cytoplasmic protein with a predicted Mr of 63,000. The protein's function is unknown, but it has been hypothesized that it either mediates resistance to DNA cross-linking agents or facilitates repair after exposure to such factors. The protein also plays a permissive role in the growth of colony-forming unit-granulocyte/macrophage (CFU-GM), burst-forming unit-erythroid (BFU-E), and CFU-erythroid (CFU-E). Attributing a specific function to this protein requires an understanding of its intracellular location. Recognizing that prior study has established the functional importance of its cytoplasmic location, we tested the hypothesis that FAC protein can also be found in the nucleus. Purified recombinant Escherichia coli-derived FAC antigens were used to create antisera able to specifically identify an Mr = 58,000 protein in lysates from human Epstein-Barr virus (EBV)-transformed cell lines by immunoblot analysis. Subcellular fractionation of the cell lysates followed by immunoblot analysis revealed that the majority of the FAC protein was cytoplasmic, as reported previously; however, approximately 10% of FAC protein was reproducibly detected in nuclear fractions. These results were reproducible by two different fractionation methods, and included markers to control for contamination of nuclear fractions by cytoplasmic proteins. Moreover, confocal image analysis of human 293 cells engineered to express FAC clearly demonstrated that FAC protein is located in both cytoplasmic and nuclear compartments, consistent with data obtained from fractionation of the FA cell lines. Finally, complementation of the FAC defect using retroviral-mediated gene transfer resulted in a substantial increase in nuclear FAC protein. Therefore, while cytoplasmic localization of this protein appears to be functionally important, it may also exert some essential nuclear function.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Anemia de Fanconi/genética , Proteínas Nucleares , Proteínas/genética , Línea Celular Transformada , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Humanos , Microscopía Confocal , Proteínas/metabolismo , Células Tumorales Cultivadas
10.
Exp Hematol ; 26(1): 19-26, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9430510

RESUMEN

The Fanconi anemia group C gene (FAC) encodes a 63-kDa protein that plays a role in the growth and differentiation of hematopoietic progenitor cells and in cellular resistance to bifunctional cross-linking agents. The function of the gene product is unknown, as are the factors that govern expression of the gene itself. Seeking to associate a function of this protein with a general metabolic pathway, we attempted to identify factors that induce or repress expression of the gene encoding it. Using two plasmids from which mutant FAC mRNA molecules were transcribed in vitro to serve as competitor mRNAs in quantitative-competitive reverse transcriptase-polymerase chain reaction analysis and novel rabbit antisera raised to recombinant FAC proteins, we quantified gene expression in human hematopoietic cells. We determined that FAC is expressed constitutively in unstimulated normal peripheral blood mononuclear leukocytes, in Epstein-Barr virus (EBV)-transformed B lymphocytes, and in the factor-dependent human myeloid leukemic cell line MO7e at levels of approximately 2000, 200, and 200 FAC mRNA molecules/cell, respectively, and in CD34+ cells from normal human bone marrow at approximately 2000 FAC mRNA molecules/cell. Neither mRNA nor protein increased in any of the cells studied after exposure to mitomycin C, diepoxybutane, hydrogen peroxide, gamma radiation, heat, transforming growth factor-beta, or interferon-gamma. Using these sensitive methods, we confirmed that the FAC gene is constitutively expressed, even in the face of extracellular factors for which the gene product is a known effector of resistance. We conclude that the protective functions of the FAC gene product do not depend upon stressor-induced FAC gene expression.


Asunto(s)
Proteínas de Ciclo Celular , Reactivos de Enlaces Cruzados/farmacología , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Hematopoyesis/genética , Monocitos/metabolismo , Proteínas Nucleares , Proteínas/genética , Animales , Células Cultivadas , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Trastornos de Estrés por Calor , Humanos , Mitosis/efectos de los fármacos , Monocitos/efectos de los fármacos , Monocitos/patología , Estrés Oxidativo , Conejos , Transfección
11.
Blood ; 90(3): 974-85, 1997 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9242526

RESUMEN

Hematopoietic progenitor cells (HPC) from mice nullizygous at the Fanconi anemia (FA) group C locus (FAC -/-) are hypersensitive to the mitotic inhibitory effects of interferon (IFN-gamma). We tested the hypothesis that HPC from the bone marrow of Fanconi group C children are similarly hypersensitive and that the fas pathway is involved in affecting programmed cell death in response to low doses of IFN-gamma. In normal human and murine HPC, IFN-gamma primed the fas pathway and induced both fas and interferon response factor-1 (IRF-1) gene expression. These IFN-gamma-induced apoptotic responses in HPC from the marrow of a child with FA of the C group (FA-C) and in FAC -/- mice occurred at significantly lower IFN doses (by an order of magnitude) than did the apoptotic responses of normal HPC. Treatment of FA-C CD34+ cells with low doses of recombinant IFN-gamma, inhibited growth of colony forming unit granulocyte-macrophage and burst-forming unit erythroid, while treatment with blocking antibodies to fas augmented clonal growth and abrogated the clonal inhibitory effect of IFN-gamma. Transfer of the normal FAC gene into FA-C B-cell lines prevented mitomycin C-induced apoptosis, but did not suppress fas expression or inhibit the primed fas pathway. However, the kinetics of Stat1-phosphate decay in IFN-gamma-treated cells was prolonged in mutant cells and was normalized by transduction of the normal FAC gene. Therefore, the normal FAC protein serves, in part, to modulate IFN-gamma signals. HPC bearing inactivating mutations of FAC fail to normally modulate IFN-gamma signals and, as a result, undergo apoptosis executed through the fas pathway.


Asunto(s)
Apoptosis/genética , Proteínas de Ciclo Celular , Anemia de Fanconi/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas Nucleares , Proteínas/fisiología , Anemia Aplásica/etiología , Anemia Aplásica/fisiopatología , Animales , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Células Cultivadas , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Anemia de Fanconi/complicaciones , Anemia de Fanconi/fisiopatología , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Proteína Ligando Fas , Prueba de Complementación Genética , Humanos , Factor 1 Regulador del Interferón , Interferón gamma/farmacología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Noqueados , Mitomicina/farmacología , Fosfoproteínas/genética , Fosfoproteínas/fisiología , Proteínas/genética , Proteínas Recombinantes , Transducción de Señal , Transfección , Receptor fas/genética , Receptor fas/fisiología
12.
J Cell Biochem ; 49(3): 290-5, 1992 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1379605

RESUMEN

Enhanced levels of disulfide-linked dimers of the neu oncogene product have been suggested to be associated with the transformed state [Weiner DB, Liu J, Cohen JA, Williams WV, Greene MI: Nature 338:230-231, (1989)]. We, therefore, investigated the properties of the dimeric forms of p185HER-2/neu from the human breast carcinoma cell line, SK-BR-3. We found disulfide-linked dimers as well as noncovalently associated dimers that were detected by cross-linking with bis(sulfosuccinimidyl) suberate (BS3). However, the disulfide-linked dimers did not exist in intact cells, since they were eliminated when the cells were lysed in the presence of the alkylating agent, sodium iodoacetate. Moreover, the disulfide-linked dimeric molecules were not the activated form of p185HER-2 since they incorporated about the same level of phosphate in an in vitro kinase reaction as the monomeric molecules. In contrast, the noncovalent dimers appeared to be present on the surface of intact cells and were phosphorylated at levels at least tenfold higher than monomers in an in vitro kinase reaction.


Asunto(s)
Disulfuros/química , Proteínas Oncogénicas Virales/química , Alquilantes/farmacología , Anticuerpos/inmunología , Neoplasias de la Mama , Humanos , Yodoacetatos/farmacología , Ácido Yodoacético , Proteínas Oncogénicas Virales/metabolismo , Fosforilación , Fosfotirosina , Pruebas de Precipitina , Receptor ErbB-2 , Células Tumorales Cultivadas , Tirosina/análogos & derivados , Tirosina/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...