Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hum Genet ; 139(6-7): 707-721, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32232558

RESUMEN

The transcription factor IRF8 (ICSBP) is required for the development and maturation of myeloid cells (dendritic cells, monocytes, macrophages), and for expression of intrinsic anti-microbial function such as antigen capture, processing and presentation to lymphoid cells, and for activation of these cells in response to cytokines and pro-inflammatory stimuli (IFN-γ, IFN-ß, LPS). IRF8 deficiency in humans causes a severe primary immunodeficiency presenting as susceptibility to infections, complete or severe depletion of blood dendritic cells (DC) subsets, depletion of CD14+ and CD16+ monocytes and reduced numbers and impaired activity of NK cells. In genome-wide association studies (GWAS), sequence variants near IRF8 are significant risk factors for multiple chronic inflammatory diseases in humans including inflammatory bowel disease, lupus, rheumatoid arthritis, multiple sclerosis, and several others. Recent studies have cataloged all the genes bound by and transcriptionally activated by IRF8 in myeloid cells, either alone or in combination with other transcription factors (PU.1, IRF1, STAT1) at steady state and in response to pro-inflammatory stimuli. This IRF1/IRF8 regulome comprises immune pathways such as antigen processing and presentation pathways, expression of costimulatory molecules, cytokines and chemokines, response to stimuli such as cytokine receptors, pathogen-associated molecular pattern receptors, TLRs and nucleotide-binding oligomerization domain-like receptor signaling pathways, and small antiviral GTPases. Members of the IRF8/IRF1 regulome are over-represented amongst genes in which mutations cause primary immunodeficiencies, and are specifically enriched at GWAS loci associated with chronic inflammatory diseases in humans. These recent studies highlight a critical role of IRF8 in the activity of several immune cell types for protection against infections, but also in pathological inflammation associated with common human inflammatory conditions.


Asunto(s)
Síndromes de Inmunodeficiencia/etiología , Síndromes de Inmunodeficiencia/prevención & control , Inflamación/etiología , Inflamación/prevención & control , Factores Reguladores del Interferón/metabolismo , Linfocitos/inmunología , Células Mieloides/inmunología , Animales , Humanos , Síndromes de Inmunodeficiencia/metabolismo , Inflamación/metabolismo , Factores Reguladores del Interferón/genética , Linfocitos/metabolismo , Mutación , Células Mieloides/metabolismo
2.
Blood ; 124(12): 1894-904, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25122610

RESUMEN

We have previously reported on a unique patient in whom homozygosity for a mutation at IRF8 (IRF8(K108E)) causes a severe immunodeficiency. Laboratory evaluation revealed a highly unusual myeloid compartment, remarkable for the complete absence of CD141 and CD161 monocytes, absence of CD11c1 conventional dendritic cells (DCs) and CD11c1/CD1231 plasmacytoid DCs, and striking granulocytic hyperplasia. The patient initially presented with severe disseminated mycobacterial and mucocutaneous fungal infections and was ultimately cured by cord blood transplant. Sequencing RNA from the IRF8(K108E) patient's primary blood cells prior to transplant shows not only depletion of IRF8-bound and IRF8-regulated transcriptional targets, in keeping with the distorted composition of the myeloid compartment, but also a paucity of transcripts associated with activated CD41 and CD81 T lymphocytes. This suggests that T cells reared in the absence of a functional antigen-presenting compartment in IRF8(K108E) are anergic. Biochemical characterization of the IRF8(K108E) mutant in vitro shows that loss of the positively charged side chain at K108 causes loss of nuclear localization and loss of transcriptional activity, which is concomitant with decreased protein stability, increased ubiquitination, increased small ubiquitin-like modification, and enhanced proteasomal degradation. These findings provide functional insight into the molecular basis of immunodeficiency associated with loss of IRF8.


Asunto(s)
Células Dendríticas/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/genética , Mutación Missense , Sustitución de Aminoácidos , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Anergia Clonal/genética , Anergia Clonal/inmunología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Femenino , Células HEK293 , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/terapia , Lactante , Factores Reguladores del Interferón/metabolismo , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/inmunología , Linfohistiocitosis Hemofagocítica/terapia , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Proteínas Mutantes/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , ARN/genética , Subgrupos de Linfocitos T/inmunología
3.
J Cell Mol Med ; 18(8): 1588-98, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24954358

RESUMEN

Increased risk of bone fractures is observed in patients with chronic inflammatory conditions, such as inflammatory bowel disease and rheumatoid arthritis. Members of the Interferon Response Factor family of transcriptional regulators, IRF1 and IRF8, have been identified as genetic risk factors for several chronic inflammatory and autoimmune diseases. We have investigated a potential role for the Irf1 gene in bone metabolism. Here, we report that Irf1(-/-) mutant mice show altered bone morphology in association with altered trabecular bone architecture and increased cortical thickness and cellularity. Ex vivo studies on cells derived from bone marrow stimulated with Rank ligand revealed an increase in size and resorptive activity of tartrate-resistant acid-positive cells from Irf1(-/-) mutant mice compared with wild-type control mice. Irf1 deficiency was also associated with decreased proliferation of bone marrow-derived osteoblast precursors ex vivo, concomitant with increased mineralization activity compared with control cells. We show that Irf1 plays a role in bone metabolism and suggest that Irf1 regulates the maturation and activity of osteoclasts and osteoblasts. The altered bone phenotype of Irf1(-/-) mutants is strikingly similar to that of Stat1(-/-) mice, suggesting that the two interacting proteins play a critical enabling role in the common regulation of these two cell lineages.


Asunto(s)
Resorción Ósea/metabolismo , Huesos/metabolismo , Diferenciación Celular , Factor 1 Regulador del Interferón/fisiología , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Animales , Western Blotting , Resorción Ósea/patología , Huesos/citología , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoclastos/citología , Fenotipo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Science ; 337(6102): 1684-8, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22859821

RESUMEN

ISG15 is an interferon (IFN)-α/ß-inducible, ubiquitin-like intracellular protein. Its conjugation to various proteins (ISGylation) contributes to antiviral immunity in mice. Here, we describe human patients with inherited ISG15 deficiency and mycobacterial, but not viral, diseases. The lack of intracellular ISG15 production and protein ISGylation was not associated with cellular susceptibility to any viruses that we tested, consistent with the lack of viral diseases in these patients. By contrast, the lack of mycobacterium-induced ISG15 secretion by leukocytes-granulocyte, in particular-reduced the production of IFN-γ by lymphocytes, including natural killer cells, probably accounting for the enhanced susceptibility to mycobacterial disease. This experiment of nature shows that human ISGylation is largely redundant for antiviral immunity, but that ISG15 plays an essential role as an IFN-γ-inducing secreted molecule for optimal antimycobacterial immunity.


Asunto(s)
Citocinas/inmunología , Interferón gamma/inmunología , Infecciones por Mycobacterium/inmunología , Ubiquitinas/inmunología , Virosis/inmunología , Animales , Anticuerpos Antivirales/sangre , Citocinas/genética , Femenino , Granulocitos/inmunología , Humanos , Inmunidad , Interleucina-12/inmunología , Células Asesinas Naturales/inmunología , Masculino , Ratones , Infecciones por Mycobacterium/sangre , Infecciones por Mycobacterium/genética , Linaje , Linfocitos T/inmunología , Ubiquitinas/genética , Virosis/sangre
5.
N Engl J Med ; 365(2): 127-38, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21524210

RESUMEN

BACKGROUND: The genetic analysis of human primary immunodeficiencies has defined the contribution of specific cell populations and molecular pathways in the host defense against infection. Disseminated infection caused by bacille Calmette-Guérin (BCG) vaccines is an early manifestation of primary immunodeficiencies, such as severe combined immunodeficiency. In many affected persons, the cause of disseminated BCG disease is unexplained. METHODS: We evaluated an infant presenting with features of severe immunodeficiency, including early-onset disseminated BCG disease, who required hematopoietic stem-cell transplantation. We also studied two otherwise healthy subjects with a history of disseminated but curable BCG disease in childhood. We characterized the monocyte and dendritic-cell compartments in these three subjects and sequenced candidate genes in which mutations could plausibly confer susceptibility to BCG disease. RESULTS: We detected two distinct disease-causing mutations affecting interferon regulatory factor 8 (IRF8). Both K108E and T80A mutations impair IRF8 transcriptional activity by disrupting the interaction between IRF8 and DNA. The K108E variant was associated with an autosomal recessive severe immunodeficiency with a complete lack of circulating monocytes and dendritic cells. The T80A variant was associated with an autosomal dominant, milder immunodeficiency and a selective depletion of CD11c+CD1c+ circulating dendritic cells. CONCLUSIONS: These findings define a class of human primary immunodeficiencies that affect the differentiation of mononuclear phagocytes. They also show that human IRF8 is critical for the development of monocytes and dendritic cells and for antimycobacterial immunity. (Funded by the Medical Research Council and others.).


Asunto(s)
Células Dendríticas/inmunología , Síndromes de Inmunodeficiencia/genética , Factores Reguladores del Interferón/genética , Mutación , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos , Vacuna BCG/genética , Vacuna BCG/inmunología , Femenino , Genes Dominantes , Humanos , Lactante , Factores Reguladores del Interferón/deficiencia , Interleucina-12/biosíntesis , Leucocitos Mononucleares/inmunología , Masculino , Modelos Moleculares , Infecciones por Mycobacterium/genética , Infecciones por Mycobacterium/inmunología , Linaje , Conformación Proteica , Alineación de Secuencia
6.
Hum Mutat ; 30(7): E706-15, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19319979

RESUMEN

Neural tube defects (NTDs) are severe congenital malformations caused by failure of the neural tube to close during neurulation. Their etiology is complex involving both environmental and genetic factors. We have recently reported three mutations in the planar cell polarity gene VANGL1 associated with NTDs. The aim of the present study was to define the role of VANGL1 genetic variants in the development of NTDs in a large cohort of various ethnic origins. We identified five novel missense variants in VANGL1, p.Ser83Leu, p.Phe153Ser, p.Arg181Gln, p.Leu202Phe and p.Ala404Ser, occurring in sporadic and familial cases of spinal dysraphisms. All five variants affect evolutionary conserved residues and are absent from all controls analyzed. This study provides further evidence supporting the role of VANGL1 as a risk factor in the development of spinal NTDs.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Mutación Missense , Defectos del Tubo Neural/genética , Adolescente , Adulto , Estudios de Casos y Controles , Niño , Preescolar , Etnicidad , Humanos , Lactante , Recién Nacido , Italia/epidemiología , Defectos del Tubo Neural/epidemiología , Factores de Riesgo , Disrafia Espinal/genética , Estados Unidos/epidemiología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...