Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 354
Filtrar
1.
J Intern Med ; 278(2): 99-109, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25832550

RESUMEN

MicroRNAs (miRNAs) are one of a growing class of noncoding RNAs that are involved in the regulation of a wide range of metabolic processes including cellular differentiation, cell proliferation and apoptosis. The generation of miRNA is regulated in complex ways, for example by small interfering RNAs (small nucleolar and nuclear RNAs) and various other metabolites. This complexity of control is likely to explain how a relatively small part of the DNA that codes for proteins has enabled the evolution of such complex organisms as mammals. Non-protein-coding DNA is therefore thought to carry the memory of early evolutionary steps that led to progressively complex metabolic controls. Clinically, miRNAs are becoming increasingly important following the recognition that some congenital abnormalities can be traced to defects in miRNA processing. The potential for manipulating metabolism and affecting disease processes by the pharmaceutical or biological targeting of specific miRNA pathways is now being tested. miRNAs are also released into the extracellular milieu after packaging by cells into nano-sized extracellular vesicles. Such vesicles can be taken up by adjacent and possibly more distant cells, thereby allowing coordinated intercellular communication in specific tissues. Extracellular miRNAs found in the blood stream may also serve as novel biomarkers for both diagnosing specific forms of cancer and assessing the likelihood of metastasis, and as powerful prognostic indices for various cancers. Here, we discuss the role of intracellular and extracellular miRNAs in nutritional control of various (patho)physiological processes. In this review, we provide an update of the presentations from the 25th Marabou Symposium (Stockholm, 14-16 June 2013) entitled 'Role of miRNA in health and nutrition', attended by 50 international experts


Asunto(s)
MicroARNs/genética , Neoplasias/genética , Evaluación Nutricional , Animales , Comunicación Celular , Humanos , Pronóstico
2.
Cell Death Differ ; 18(4): 690-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21164518

RESUMEN

Thymocyte development is a tightly controlled multi-step process involving selective elimination of self-reactive and non-functional T cells by apoptosis. This developmental process depends on signaling by Notch, IL-7 and active glucose metabolism. In this study, we explored the requirement of glucose for thymocyte survival and found that in addition to metabolic regulation, glucose leads to the expression of anti-apoptotic genes. Under hyperglycemic conditions, both mouse and human thymocytes demonstrate enhanced survival. We show that glucose-induced anti-apoptotic genes are dependent on NF-κB p65 because high glucose is unable to attenuate normal ongoing apoptosis of thymocytes isolated from p65 knockout mice. Furthermore, we demonstrate that in vivo hyperglycemia decreases apoptosis of thymocytes allowing for survival of potentially self-reactive thymocytes. These results imply that hyperglycemic conditions could contribute to the development of autoimmunity through dysregulated thymic selection.


Asunto(s)
Apoptosis , Diabetes Mellitus Experimental/inmunología , Glucosa/farmacología , Linfocitos T/inmunología , Animales , Autoinmunidad , Supervivencia Celular , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-rel/genética , Proteínas Proto-Oncogénicas c-rel/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Glándula Tiroides/citología , Glándula Tiroides/inmunología , Glándula Tiroides/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Proteína X Asociada a bcl-2/metabolismo
3.
J Immunol ; 167(3): 1592-600, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11466381

RESUMEN

NF-kappaB binding sites are present in the promoter regions of many acute phase and inflammatory response genes, suggesting that NF-kappaB plays an important role in the initiation of innate immune responses. However, targeted mutations of the various NF-kappaB family members have yet to identify members responsible for this critical role. RelA-deficient mice die on embryonic day 15 from TNF-alpha-induced liver degeneration. To investigate the importance of RelA in innate immunity, we genetically suppressed this embryonic lethality by breeding the RelA deficiency onto a TNFR type 1 (TNFR1)-deficient background. TNFR1/RelA-deficient mice were born healthy, but were susceptible to bacterial infections and bacteremia and died within a few weeks after birth. Hemopoiesis was intact in TNFR1/RelA-deficient newborns, but neutrophil emigration to alveoli during LPS-induced pneumonia was severely reduced relative to that in wild-type or TNFR1-deficient mice. In contrast, radiation chimeras reconstituted with RelA or TNFR1/RelA-deficient hemopoietic cells were healthy and demonstrated no defect in neutrophil emigration during LPS-induced pneumonia. Analysis of RNA harvested from the lungs of mice 4 h after LPS insufflation revealed that the induction of several genes important for neutrophil recruitment to the lung was significantly reduced in TNFR1/RelA-deficient mice relative to that in wild-type or TNFR1-deficient mice. These results suggest that TNFR1-independent activation of RelA is essential in cells of nonhemopoietic origin during the initiation of an innate immune response.


Asunto(s)
Antígenos CD/genética , Eliminación de Gen , FN-kappa B/deficiencia , FN-kappa B/fisiología , Infiltración Neutrófila , Receptores del Factor de Necrosis Tumoral/genética , Animales , Antígenos CD/fisiología , Femenino , Muerte Fetal/genética , Muerte Fetal/inmunología , Muerte Fetal/patología , Muerte Fetal/prevención & control , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Marcación de Gen , Hematopoyesis/genética , Hematopoyesis/inmunología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/biosíntesis , FN-kappa B/genética , Infiltración Neutrófila/genética , Peritonitis/inducido químicamente , Peritonitis/patología , Neumonía Bacteriana/genética , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/patología , Quimera por Radiación/inmunología , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral , Análisis de Supervivencia , Tioglicolatos/toxicidad , Factor de Transcripción ReIA
4.
J Biol Chem ; 276(4): 2459-65, 2001 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-11042170

RESUMEN

p62Dok, the rasGAP-binding protein, is a common target of protein-tyrosine kinases. It is one of the major tyrosine-phosphorylated molecules in v-Src-transformed cells. Dok consists of an amino-terminal Pleckstrin homology domain, a putative phosphotyrosine binding domain, and a carboxyl-terminal tail containing multiple tyrosine phosphorylation sites. The importance and function of these sequences in Dok signaling remain largely unknown. We have demonstrated here that the expression of Dok can inhibit cellular transformation by the Src tyrosine kinase. Both the phosphotyrosine binding domain and the carboxyl-terminal tail of Dok (in particular residues 336-363) are necessary for such activity. Using a combinatorial peptide library approach, we have shown that the Dok phosphotyrosine binding domain binds phosphopeptides with the consensus motif of Y/MXXNXL-phosphotyrosine. Furthermore, Dok can homodimerize through its phosphotyrosine binding domain and Tyr(146) at the amino-terminal region. Mutations of this domain or Tyr(146) that block homodimerization significantly reduce the ability of Dok to inhibit Src transformation. Our results suggest that Dok oligomerization through its multiple domains plays a critical role in Dok signaling in response to tyrosine kinase activation.


Asunto(s)
Proteínas de Unión al ADN , Activadores de GTP Fosfohidrolasa/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN , Proteínas Activadoras de ras GTPasa/metabolismo , Células 3T3 , Animales , Transformación Celular Neoplásica , Ratones , Fosfopéptidos/metabolismo , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal
5.
Proc Natl Acad Sci U S A ; 97(23): 12705-10, 2000 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-11058167

RESUMEN

A major challenge in the study of gene regulation by NF-kappaB/Rel transcription factors is to understand, at the biological and mechanistic levels, the selective functions of individual Rel family members. To study selectivity, we have examined the NF-kappaB/Rel protein binding site (Rel site) within the IL-12 p40 promoter. IL-12 is a proinflammatory cytokine expressed by activated macrophages that serves as an essential inducer of T helper 1 cell development. In nuclear extracts from lipopolysaccharideactivated macrophages, the predominant Rel dimers capable of binding the IL-12 p40 Rel site were the p50/p65 and p50/c-Rel heterodimers and p50/p50 homodimer. The two heterodimers bound the site with comparable affinities and exhibited comparable transactivation activities. In striking contrast, p40 mRNA and protein concentrations were reduced dramatically in c-Rel(-/-) macrophages and only modestly in p65(-/-) macrophages. Other proinflammatory cytokine mRNAs and proteins were not significantly reduced in c-Rel(-/-) macrophages. These results reveal that a c-Rel-containing complex is an essential and selective activator of p40 transcription, which may reflect unique regulatory mechanisms or biological functions of IL-12. Furthermore, because selectivity was not observed in vitro or in transient transactivation experiments, these findings suggest that an understanding of the selectivity mechanism may require an analysis of the endogenous p40 locus.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Interleucina-12/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-rel/metabolismo , Activación Transcripcional , Animales , Línea Celular , Línea Celular Transformada , Citocinas/genética , Proteínas de Unión al ADN/genética , Dimerización , Humanos , Interleucina-12/biosíntesis , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Subunidad p50 de NF-kappa B , Proteínas Proto-Oncogénicas c-rel/genética , ARN Mensajero , Factor de Transcripción ReIA
6.
Mol Cell Biol ; 20(20): 7419-26, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11003639

RESUMEN

Achieving long-term retroviral expression in primary cells has been problematic. De novo DNA methylation of infecting proviruses has been proposed as a major cause of this transcriptional repression. Here we report the development of a mouse stem cell virus (MSCV) long terminal repeat-based retroviral vector that is expressed in both embryonic stem (ES) cells and hematopoietic stem (HS) cells. Infected HS cells and their differentiated descendants maintained long-term and stable retroviral expression after serial adoptive transfers. In addition, retrovirally infected ES cells showed detectable expression level of the green fluorescent protein (GFP). Moreover, GFP expression of integrated proviruses was maintained after in vitro differentiation of infected ES cells. Long-term passage of infected ES cells resulted in methylation-mediated silencing, while short-term expression was methylation independent. Tissues of transgenic animals, which we derived from ES cells carrying the MSCV-based provirus, did not express GFP. However, treatment with the demethylating agent 5-azadeoxycytidine reactivated the silent provirus, demonstrating that DNA methylation is involved in the maintenance of retroviral repression. Our results indicate that retroviral expression in ES cells is repressed by methylation-dependent as well as methylation-independent mechanisms.


Asunto(s)
Regulación Viral de la Expresión Génica , Silenciador del Gen , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Retroviridae/genética , Células Madre/metabolismo , Traslado Adoptivo , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Diferenciación Celular , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/efectos de los fármacos , Decitabina , Citometría de Flujo , Regulación Viral de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Provirus/efectos de los fármacos , Provirus/genética , Proteínas Recombinantes de Fusión/genética , Retroviridae/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/virología , Secuencias Repetidas Terminales/genética
8.
Genes Dev ; 14(4): 397-402, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10691732

RESUMEN

Although a small decrease in survival and increase in tumor incidence was observed in ATR(+/-) mice, ATR(-/-) embryos die early in development, subsequent to the blastocyst stage and prior to 7.5 days p.c. In culture, ATR(-/-) blastocysts cells continue to cycle into mitosis for 2 days but subsequently fail to expand and die of caspase-dependent apoptosis. Importantly, caspase-independent chromosome breaks are observed in ATR(-/-) cells prior to widespread apoptosis, implying that apoptosis is caused by a loss of genomic integrity. These data show that ATR is essential for early embryonic development and must function in processes other than regulation of p53.


Asunto(s)
Proteínas de Ciclo Celular , Aberraciones Cromosómicas , Muerte Fetal/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Apoptosis/genética , Proteínas de la Ataxia Telangiectasia Mutada , Proteína BRCA1/fisiología , Proteína BRCA2 , Blastocisto/patología , Caspasas/metabolismo , Transformación Celular Neoplásica/genética , Reparación del ADN/genética , Marcación de Gen , Genotipo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis , Proteínas de Neoplasias/fisiología , Neoplasias Experimentales/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/fisiología
9.
EMBO J ; 18(23): 6694-704, 1999 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-10581243

RESUMEN

The activation of NF-kappaB by receptors in the tumor necrosis factor (TNF) receptor and Toll/interleukin-1 (IL-1) receptor families requires the TRAF family of adaptor proteins. Receptor oligomerization causes the recruitment of TRAFs to the receptor complex, followed by the activation of a kinase cascade that results in the phosphorylation of IkappaB. TANK is a TRAF-binding protein that can inhibit the binding of TRAFs to receptor tails and can also inhibit NF-kappaB activation by these receptors. However, TANK also displays the ability to stimulate TRAF-mediated NF-kappaB activation. In this report, we investigate the mechanism of the stimulatory activity of TANK. We find that TANK interacts with TBK1 (TANK-binding kinase 1), a novel IKK-related kinase that can activate NF-kappaB in a kinase-dependent manner. TBK1, TANK and TRAF2 can form a ternary complex, and complex formation appears to be required for TBK1 activity. Kinase-inactive TBK1 inhibits TANK-mediated NF-kappaB activation but does not block the activation mediated by TNF-alpha, IL-1 or CD40. The TBK1-TANK-TRAF2 signaling complex functions upstream of NIK and the IKK complex and represents an alternative to the receptor signaling complex for TRAF-mediated activation of NF-kappaB.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Activación Enzimática , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Antígenos CD40/metabolismo , Ligando de CD40 , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Interleucina-1/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Glicoproteínas de Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Factor 2 Asociado a Receptor de TNF , Factor de Necrosis Tumoral alfa/metabolismo , Técnicas del Sistema de Dos Híbridos
10.
Immunity ; 11(3): 281-8, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10514006

RESUMEN

IL-2 is an important growth and survival factor for T lymphocytes but also sensitizes these cells to Fas-mediated activation-induced cell death (AICD). The molecular basis of these different effects of IL-2 was studied by introducing wild-type and mutant forms of the IL-2 receptor beta (IL-2Rbeta) chain that lacked specific signaling capacities into receptor-deficient T cells by retroviral gene transfer. Activation of Stat5 by IL-2 was found to be involved in T cell proliferation and promoted Fas ligand (FasL) expression and AICD. T cell survival was dependent on a receptor region that activated Akt and the expression of Bcl-2. Thus, distinct IL-2Rbeta chain signaling modules regulate T cell fate by stimulating growth and survival or by promoting apoptosis.


Asunto(s)
Apoptosis/inmunología , Interleucina-2/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Receptor fas/inmunología , Animales , División Celular , Supervivencia Celular , Células Cultivadas , Proteína Ligando Fas , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Interleucina-2/farmacología , Activación de Linfocitos , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis , Receptores de Interleucina-2/genética , Retroviridae , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
11.
Proc Natl Acad Sci U S A ; 96(21): 11848-53, 1999 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-10518539

RESUMEN

Individual Rel/NF-kappaB transcription factors, although dispensable for the development and maturation of most hemopoietic cells, are critical regulators of normal immune function. Redundancy among these proteins prompted us to examine the role of Rel and RelA in hemopoiesis by using mice that lack both subunits. Because of the death of double-mutant fetuses at day 13.5 of gestation (E13.5), E12 fetal liver hemopoietic progenitors were used for in vitro cultures and for repopulating stem cell studies in lethally irradiated normal recipient mice. Most striking, Rel/RelA-deficient hemopoietic precursors failed to promote the survival of myeloablated mice. This phenotype was associated with several defects including a reduction of spleen colony-forming unit progenitors, impaired erythropoiesis, and a deregulated expansion of granulocytes. In vitro progenitor assays also revealed that Rel or RelA serves an antiapoptotic role during monocyte differentiation. Despite the combined loss of Rel and RelA leading to these hemopoietic defects, c-rel(-/-)rela(-/-) stem cells contributed to the development of all lineages in mice engrafted with double-mutant fetal liver cells and normal bone marrow cells, albeit in a reduced fashion compared with controls. Collectively, these data indicate the loss of Rel and RelA does not appear to affect pluripotent stem cells; rather, Rel and RelA serve redundant functions in regulating differentiation and survival of committed progenitors in multiple hemopoietic lineages.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , FN-kappa B/genética , FN-kappa B/fisiología , Proteínas Proto-Oncogénicas c-rel/genética , Proteínas Proto-Oncogénicas c-rel/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Eritropoyesis , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Hígado/embriología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Células Madre , Factores de Tiempo , Regulación hacia Arriba
12.
Immunol Rev ; 168: 65-74, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10399065

RESUMEN

Despite a strong cytotoxic T-lymphocyte (CTL) response directed against viral antigens, untreated individuals infected with the human immunodeficiency virus (HIV-1) develop AIDS. We have found that primary T cells infected with HIV-1 downregulate surface MHC class I antigens and are resistant to lysis by HLA-A2-restricted CTL clones. In contrast, cells infected with an HIV-1 in which the nef gene is disrupted are sensitive to CTLs in an MHC and peptide-specific manner. In primary T cells HLA-A2 antigens are downmodulated more dramatically than total MHC class I antigens, suggesting that nef selectively downmodulates certain MHC class I antigens. In support of this, studies on cells expressing individual MHC class I alleles have revealed that nef does not downmodulate HLA-C and HLA-E antigens. This selective downmodulation allows infected cells to maintain resistance to certain natural killer cells that lyse infected cells expressing low levels of MHC class I antigens. Downmodulation of MHC class I HLA-A2 antigens occurs not only in primary T cells, but also in B and astrocytoma cell lines. No effect of other HIV-1 accessory proteins such as vpu and vpr was observed. Thus Nef is a protein that may promote escape of HIV-1 from immune surveillance.


Asunto(s)
VIH-1/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Productos del Gen gag/inmunología , Productos del Gen nef/inmunología , VIH-1/patogenicidad , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunidad Celular , Factores de Tiempo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana
13.
Immunity ; 10(6): 661-71, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10403641

RESUMEN

To avoid detection by CTL, HIV encodes mechanisms for removal of class I MHC proteins from the surface of infected cells. However, class I downregulation potentially exposes the virus-infected cell to attack by NK cells. Human lymphoid cells are protected from NK cell cytotoxicity primarily by HLA-C and HLA-E. We present evidence that HIV-1 selectively downregulates HLA-A and HLA-B but does not significantly affect HLA-C or HLA-E. We then identify the residues in HLA-C and HLA-E that protect them from HIV down-regulation. This selective downregulation allows HIV-infected cells to avoid NK cell-mediated lysis and may represent for HIV a balance between escape from CTL and maintenance of protection from NK cells. These results suggest that subpopulations of CTL and NK cells may be uniquely suited for combating HIV.


Asunto(s)
Regulación hacia Abajo/inmunología , VIH-1/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/inmunología , Células Cultivadas , Citoplasma/química , Citoplasma/inmunología , Citotoxicidad Inmunológica , Productos del Gen nef/fisiología , Antígenos HLA/fisiología , Antígeno HLA-A2/metabolismo , Antígenos HLA-C/fisiología , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/fisiología , Humanos , Células Asesinas Naturales/virología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Antígenos HLA-E
14.
J Biol Chem ; 274(18): 12748-52, 1999 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-10212258

RESUMEN

Cells from individuals with the recessive cancer-prone disorder ataxia telangiectasia (A-T) are hypersensitive to ionizing radiation (I-R). ATM (mutated in A-T) is a protein kinase whose activity is stimulated by I-R. c-Abl, a nonreceptor tyrosine kinase, interacts with ATM and is activated by ATM following I-R. Rad51 is a homologue of bacterial RecA protein required for DNA recombination and repair. Here we demonstrate that there is an I-R-induced Rad51 tyrosine phosphorylation, and this induction is dependent on both ATM and c-Abl. ATM, c-Abl, and Rad51 can be co-immunoprecipitated from cell extracts. Consistent with the physical interaction, c-Abl phosphorylates Rad51 in vitro and in vivo. In assays using purified components, phosphorylation of Rad51 by c-Abl enhances complex formation between Rad51 and Rad52, which cooperates with Rad51 in recombination and repair. After I-R, an increase in association between Rad51 and Rad52 occurs in wild-type cells but not in cells with mutations that compromise ATM or c-Abl. Our data suggest signaling mediated through ATM, and c-Abl is required for the correct post-translational modification of Rad51, which is critical for the assembly of Rad51 repair protein complex following I-R.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Recombinación Genética , Proteínas de Saccharomyces cerevisiae , Proteínas de la Ataxia Telangiectasia Mutada , Quinasa de Punto de Control 2 , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Recombinasa Rad51 , Proteína Recombinante y Reparadora de ADN Rad52 , Proteínas Supresoras de Tumor , Tirosina/metabolismo
15.
Proc Natl Acad Sci U S A ; 96(4): 1252-6, 1999 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-9990010

RESUMEN

Fas is a cell surface death receptor that regulates peripheral tolerance and lymphoid homeostasis. In many pathologic conditions, ectopic Fas activation mediates tissue destruction. Several proteins that can bind to the cytoplasmic death domain of Fas have been implicated in Fas signal transduction. Here we show that FADD, which couples Fas to pro-caspase-8, and, Daxx, which couples Fas to the Jun N-terminal kinase pathway, bind independently to the Fas death domain. We have isolated a death domain mutant, termed FasDelta, that selectively binds Daxx but not FADD. In tranfected tissue culture cells, FasDelta activated Jun N-terminal kinase normally but was impaired in cell death induction. These results suggest that FADD and Daxx activate two independent pathways downstream of Fas and confirm the essential role of FADD binding in apoptosis induction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas Quinasas Activadas por Mitógenos , Proteínas Nucleares , Transducción de Señal/fisiología , Receptor fas/fisiología , Proteínas Portadoras/genética , Línea Celular , Proteínas Co-Represoras , Activación Enzimática , Proteína de Dominio de Muerte Asociada a Fas , Células HeLa , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Cinética , Chaperonas Moleculares , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Receptor fas/genética
16.
Proc Natl Acad Sci U S A ; 96(4): 1421-6, 1999 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-9990039

RESUMEN

Several tumor necrosis factor receptor-associated factor (TRAF) family proteins including TRAF2, TRAF3, TRAF5, and TRAF6, as well as Jak3, have been implicated as potential mediators of CD40 signaling. An extensive in vitro binding study indicated that TRAF2 and TRAF3 bind to the CD40 cytoplasmic tail (CD40ct) with much higher affinity than TRAF5 and TRAF6 and that TRAF2 and TRAF3 bind to different residues of the CD40ct. Using CD40 mutants incapable of binding TRAF2, TRAF3, or Jak3, we found that the TRAF2-binding site of the CD40ct is critical for NF-kappaB and stress-activated protein kinase activation, as well as the up-regulation of the intercellular adhesion molecule-1 (ICAM-1) gene, whereas binding of TRAF3 and Jak3 is dispensable for all of these functions. Overexpression of a dominantly active IkappaBalpha strongly inhibited CD40-induced NF-kappaB activation, ICAM-1 promoter activity, and cell-surface ICAM-1 up-regulation. These studies suggest a potential signal transduction pathway from the CD40 receptor to the transcriptional activation of the ICAM-1 gene.


Asunto(s)
Antígenos CD40/fisiología , Regulación de la Expresión Génica , Molécula 1 de Adhesión Intercelular/genética , FN-kappa B/metabolismo , Proteínas/fisiología , Sustitución de Aminoácidos , Animales , Antígenos CD/genética , Antígenos CD/fisiología , Antígenos CD40/genética , Línea Celular , Genes Reporteros , Humanos , Janus Quinasa 3 , Linfoma de Células B , Ratones , Mutagénesis Sitio-Dirigida , Proteínas Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas/genética , Proteínas Recombinantes de Fusión/biosíntesis , Transducción de Señal , Factor 2 Asociado a Receptor de TNF , Transfección , Células Tumorales Cultivadas
17.
J Immunol ; 162(4): 1941-6, 1999 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9973462

RESUMEN

B cells lacking individual NF-kappa B/Rel family members exhibit defects in activation programs. We generated small resting B cells lacking p65 or p50 alone, or lacking both p50 and p65, then evaluated the ability of these cells to proliferate, secrete Ig, and undergo Ig class switching. B cells lacking p65 proliferated well in response to all stimuli tested. However, these cells demonstrated an isolated defect in switching to IgG3, which was associated with a decrease in gamma 3 germline CH gene expression. Whereas, previously reported, B cells lacking p50 alone had a severe proliferative defect in response to LPS, a moderate defect in response to CD40 ligand (CD40L), and normal proliferation to Ag receptor cross-linking using dextran-conjugated anti-IgD Abs (alpha delta-dex), B cells lacking both p50 and p65 exhibited severely impaired proliferation in response to LPS, alpha delta-dex, and CD40L. This defect could be overcome by simultaneous administration of alpha delta-dex and CD40L. In response to this latter combination of stimuli, B cells lacking both p50 and p65 secreted Ig and underwent isotype switching to IgG1 as efficiently as B cells lacking p50 alone. These data demonstrate a role for the p65 subunit of NF-kappa B in germline CH gene expression as well as functional redundancy between p50 and p65 during proliferative responses.


Asunto(s)
Linfocitos B/metabolismo , Cambio de Clase de Inmunoglobulina , Regiones Constantes de Inmunoglobulina/genética , Inmunoglobulina G/biosíntesis , Activación de Linfocitos , FN-kappa B/fisiología , Transcripción Genética/inmunología , Animales , Linfocitos B/inmunología , División Celular/genética , División Celular/inmunología , Células Cultivadas , Femenino , Cambio de Clase de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/genética , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , FN-kappa B/deficiencia , FN-kappa B/genética , Subunidad p50 de NF-kappa B , Factor de Transcripción ReIA
18.
Immunity ; 11(6): 763-70, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10626898

RESUMEN

The induction of apoptosis by death receptors serves to regulate immune responses by eliminating unwanted and harmful cells. Mature lymphocytes express FLICE inhibitory proteins (FLIPs) that block death receptor-induced cell death. Here, we show that both B and T cells downregulate c-FLIP upon activation in vitro. Retrovirus-mediated expression of c-FLIP blocks Fas-induced apoptosis of activated lymphocytes but does not affect cell death resulting from cytokine withdrawal. In vivo, c-FLIP expression results in defective superantigen-mediated elimination of T cells, the accumulation of activated B cells, the production of autoantibodies, and the development of autoimmune disease. No effect was seen on negative selection of thyomocytes. These results suggest that activation-dependent downregulation of c-FLIP renders mature lymphocytes sensitive to death receptor-mediated apoptosis and is required to maintain self-tolerance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Autoinmunidad/inmunología , Linfocitos B/inmunología , Proteínas Portadoras/inmunología , Péptidos y Proteínas de Señalización Intracelular , Linfocitos T/inmunología , Animales , Apoptosis , Enfermedades Autoinmunes/inmunología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Células Cultivadas , Regulación hacia Abajo , Enterotoxinas/inmunología , Proteína de Dominio de Muerte Asociada a Fas , Expresión Génica , Vectores Genéticos/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Retroviridae/genética , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Receptor fas/inmunología
19.
Blood ; 92(10): 3780-92, 1998 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-9808572

RESUMEN

Expression of the 210-kD bcr/abl fusion oncoprotein can cause a chronic myelogenous leukemia (CML)-like disease in mice receiving bone marrow cells transduced by bcr/abl-encoding retroviruses. However, previous methods failed to yield this disease at a frequency sufficient enough to allow for its use in the study of CML pathogenesis. To overcome this limitation, we have developed an efficient and reproducible method for inducing a CML-like disease in mice receiving P210 bcr/abl-transduced bone marrow cells. All mice receiving P210 bcr/abl-transduced bone marrow cells succumb to a myeloproliferative disease between 3 and 5 weeks after bone marrow transplantation. The myeloproliferative disease recapitulates many of the hallmarks of human CML and is characterized by high white blood cell counts and extensive extramedullary hematopoiesis in the spleen, liver, bone marrow, and lungs. Use of a retroviral vector coexpressing P210 bcr/abl and green fluorescent protein shows that the vast majority of bcr/abl-expressing cells are myeloid. Analysis of the proviral integration pattern shows that, in some mice, the myeloproliferative disease is clonal. In multiple mice, the CML-like disease has been transplantable, inducing a similar myeloproliferative syndrome within 1 month of transfer to sublethally irradiated syngeneic recipients. The disease in many of these mice has progressed to the development of acute lymphoma/leukemia resembling blast crisis. These results demonstrate that murine CML recapitulates important features of human CML. As such, it should be an excellent model for addressing specific issues relating to the pathogenesis and treatment of this disease.


Asunto(s)
Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Proteínas de Fusión bcr-abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Trastornos Mieloproliferativos/etiología , Oncogenes , Animales , Crisis Blástica/patología , Células de la Médula Ósea/virología , Células Cultivadas/trasplante , Células Clonales/patología , Proteínas de Fusión bcr-abl/análisis , Genes abl , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Hematopoyesis Extramedular , Humanos , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos BALB C , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Trasplante de Neoplasias , Provirus/genética , Quimera por Radiación , Proteínas Recombinantes de Fusión/análisis , Reproducibilidad de los Resultados , Retroviridae/genética , Esplenomegalia/patología , Transfección , Integración Viral
20.
Science ; 281(5384): 1860-3, 1998 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-9743501

RESUMEN

The Fas death receptor can activate the Jun NH2-terminal kinase (JNK) pathway through the receptor-associated protein Daxx. Daxx was found to activate the JNK kinase kinase ASK1, and overexpression of a kinase-deficient ASK1 mutant inhibited Fas- and Daxx-induced apoptosis and JNK activation. Fas activation induced Daxx to interact with ASK1, which consequently relieved an inhibitory intramolecular interaction between the amino- and carboxyl-termini of ASK1, activating its kinase activity. The Daxx-ASK1 connection completes a signaling pathway from a cell surface death receptor to kinase cascades that modulate nuclear transcription factors.


Asunto(s)
Apoptosis , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas Quinasas Activadas por Mitógenos , Proteínas Nucleares , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Alelos , Secuencia de Aminoácidos , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Proteínas Co-Represoras , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Quinasas Quinasa Quinasa PAM , Chaperonas Moleculares , Datos de Secuencia Molecular , Proteínas Serina-Treonina Quinasas/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Receptor fas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA