Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 48(5): 979-991.e8, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29752066

RESUMEN

The triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial innate immune receptor associated with a lethal form of early, progressive dementia, Nasu-Hakola disease, and with an increased risk of Alzheimer's disease. Microglial defects in phagocytosis of toxic aggregates or apoptotic membranes were proposed to be at the origin of the pathological processes in the presence of Trem2 inactivating mutations. Here, we show that TREM2 is essential for microglia-mediated synaptic refinement during the early stages of brain development. The absence of Trem2 resulted in impaired synapse elimination, accompanied by enhanced excitatory neurotransmission and reduced long-range functional connectivity. Trem2-/- mice displayed repetitive behavior and altered sociability. TREM2 protein levels were also negatively correlated with the severity of symptoms in humans affected by autism. These data unveil the role of TREM2 in neuronal circuit sculpting and provide the evidence for the receptor's involvement in neurodevelopmental diseases.


Asunto(s)
Encéfalo/inmunología , Glicoproteínas de Membrana/inmunología , Microglía/inmunología , Neuronas/inmunología , Receptores Inmunológicos/inmunología , Sinapsis/inmunología , Animales , Trastorno Autístico/genética , Trastorno Autístico/inmunología , Trastorno Autístico/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Microglía/metabolismo , Neuronas/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/genética , Transmisión Sináptica/inmunología
2.
Nat Immunol ; 10(7): 734-43, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19503107

RESUMEN

Macrophage colony-stimulating factor (M-CSF) influences the proliferation and survival of mononuclear phagocytes through the receptor CSF-1R. The adaptor protein DAP12 is critical for the function of mononuclear phagocytes. DAP12-mutant mice and humans have defects in osteoclasts and microglia, as well as brain and bone abnormalities. Here we show DAP12 deficiency impaired the M-CSF-induced proliferation and survival of macrophages in vitro. DAP12-deficient mice had fewer microglia in defined central nervous system areas, and DAP12-deficient progenitors regenerated myeloid cells inefficiently after bone marrow transplantation. Signaling by M-CSF through CSF-1R induced the stabilization and nuclear translocation of beta-catenin, which activated genes involved in the cell cycle. DAP12 was essential for phosphorylation and nuclear accumulation of beta-catenin. Our results provide a mechanistic explanation for the many defects of DAP12-deficient mononuclear phagocytes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proliferación Celular/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , beta Catenina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citometría de Flujo , Quinasa 2 de Adhesión Focal/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Immunoblotting , Inmunohistoquímica , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos , Fosforilación
3.
Blood ; 121(17): 3473-83, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23426944

RESUMEN

Efferocytosis of apoptotic neutrophils by macrophages following tissue injury is fundamental to the resolution of inflammation and initiation of tissue repair. Using a sterile peritonitis model in mice, we identified interleukin (IL)-4-producing efferocytosing macrophages in the peritoneum that activate invariant natural killer T (iNKT) cells to produce cytokines including IL-4, IL-13, and interferon-γ. Importantly, IL-4 from macrophages contributes to alternative activation of peritoneal exudate macrophages and augments type 2 cytokine production from NKT cells to suppress inflammation. The increased peritonitis in mice deficient in IL-4, NKT cells, or IL-4Rα expression on myeloid cells suggested that each is a key component for resolution of sterile inflammation. The reduced NAD phosphate oxidase is also critical for this model, because in mice with X-linked chronic granulomatous disease (X-CGD) that lack oxidase subunits, activation of iNKT cells by X-CGD peritoneal exudate macrophages was impaired during sterile peritonitis, resulting in enhanced and prolonged inflammation in these mice. Therefore, efferocytosis-induced IL-4 production and activation of IL-4-producing iNKT cells by macrophages are immunomodulatory events in an innate immune circuit required to resolve sterile inflammation and promote tissue repair.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Enfermedad Granulomatosa Crónica/inmunología , Inflamación/prevención & control , Interleucina-4/fisiología , Macrófagos/inmunología , Células T Asesinas Naturales/inmunología , Fagocitosis/fisiología , Animales , Citocinas/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Enfermedad Granulomatosa Crónica/metabolismo , Enfermedad Granulomatosa Crónica/patología , Inflamación/inmunología , Inflamación/patología , Interferón gamma/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/patología , Enfermedades Peritoneales/inmunología , Enfermedades Peritoneales/patología , Enfermedades Peritoneales/prevención & control , Receptores de Superficie Celular/fisiología
4.
Acta Neuropathol ; 129(3): 429-47, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25631124

RESUMEN

Microglia are phagocytic cells that survey the brain and perform neuroprotective functions in response to tissue damage, but their activating receptors are largely unknown. Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial immunoreceptor whose loss-of-function mutations in humans cause presenile dementia, while genetic variants are associated with increased risk of neurodegenerative diseases. In myeloid cells, TREM2 has been involved in the regulation of phagocytosis, cell proliferation and inflammatory responses in vitro. However, it is unknown how TREM2 contributes to microglia function in vivo. Here, we identify a critical role for TREM2 in the activation and function of microglia during cuprizone (CPZ)-induced demyelination. TREM2-deficient (TREM2(-/-)) mice had defective clearance of myelin debris and more axonal pathology, resulting in impaired clinical performances compared to wild-type (WT) mice. TREM2(-/-) microglia proliferated less in areas of demyelination and were less activated, displaying a more resting morphology and decreased expression of the activation markers MHC II and inducible nitric oxide synthase as compared to WT. Mechanistically, gene expression and ultrastructural analysis of microglia suggested a defect in myelin degradation and phagosome processing during CPZ intoxication in TREM2(-/-) microglia. These findings place TREM2 as a key regulator of microglia activation in vivo in response to tissue damage.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proliferación Celular , Quelantes/toxicidad , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Inmunoelectrónica , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
J Immunol ; 191(12): 5933-40, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24218450

RESUMEN

Plasmacytoid dendritic cells (pDCs) are a dendritic cell subset that secrete type I IFNs in response to microbial stimuli. The scaffold protein, CD2-associated protein (CD2AP), is a marker of human pDCs as it is highly expressed in this cell type. Recently, in human pDCs, decreased CD2AP expression appeared to enhance the production of type I IFNs via an inhibitory receptor-induced signaling cascade. In this study, we sought to determine the role of CD2AP in murine pDCs using CD2AP knockout (KO) mice. CD2AP was dispensable for the development of pDCs and for the upregulation of activation markers following stimulation. Loss of CD2AP expression did not affect the production of type I IFNs stimulated by TLR ligation, and only slightly impaired type I IFN production when inhibitory pathways were engaged in vitro. This was also confirmed by showing that CD2AP deficiency did not influence type I IFN production by pDCs in vivo. Because CD2AP plays a role in regulating actin dynamics, we examined the actin cytoskeleton in pDCs and found that activated CD2AP KO pDCs had significantly higher levels of actin polymerization than wild-type pDCs. Using two different inflammation models, we found that CD2AP KO pDCs have a defect in lymph node migration, correlating with the defects in actin dynamics. Our work excludes a role for CD2AP in the regulation of type I IFNs in pDCs, and suggests that the major function of CD2AP is on the actin cytoskeleton, affecting migration to local lymph nodes under conditions of inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Citocinas/biosíntesis , Proteínas del Citoesqueleto/fisiología , Células Dendríticas/citología , Inflamación/patología , Ganglios Linfáticos/patología , Actinas/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Movimiento Celular/fisiología , Citocinas/genética , Proteínas del Citoesqueleto/deficiencia , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Células Dendríticas/metabolismo , Inflamación/inmunología , Interferón-alfa/biosíntesis , Interferón-alfa/genética , Ganglios Linfáticos/inmunología , Tejido Linfoide/citología , Ratones , Ratones Congénicos , Ratones Noqueados , Mycobacterium tuberculosis/inmunología , Quimera por Radiación , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Vesiculovirus/inmunología
6.
Nature ; 457(7230): 722-5, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-18978771

RESUMEN

Natural killer (NK) cells are classically viewed as lymphocytes that provide innate surveillance against virally infected cells and tumour cells through the release of cytolytic mediators and interferon (IFN)-gamma. In humans, blood CD56(dim) NK cells specialize in the lysis of cell targets. In the lymph nodes, CD56(bright) NK cells secrete IFN-gamma cooperating with dendritic cells and T cells in the generation of adaptive responses. Here we report the characterization of a human NK cell subset located in mucosa-associated lymphoid tissues, such as tonsils and Peyer's patches, which is hard-wired to secrete interleukin (IL)-22, IL-26 and leukaemia inhibitory factor. These NK cells, which we refer to as NK-22 cells, are triggered by acute exposure to IL-23. In vitro, NK-22-secreted cytokines stimulate epithelial cells to secrete IL-10, proliferate and express a variety of mitogenic and anti-apoptotic molecules. NK-22 cells are also found in mouse mucosa-associated lymphoid tissues and appear in the small intestine lamina propria during bacterial infection, suggesting that NK-22 cells provide an innate source of IL-22 that may help constrain inflammation and protect mucosal sites.


Asunto(s)
Inmunidad Mucosa/inmunología , Interleucinas/biosíntesis , Interleucinas/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Animales , Antígeno CD56/análisis , Adhesión Celular , Quimiocina CCL20/biosíntesis , Quimiocina CCL20/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-23/inmunología , Interleucinas/metabolismo , Células Asesinas Naturales/metabolismo , Factor Inhibidor de Leucemia/biosíntesis , Ratones , Monocitos/inmunología , Tonsila Palatina/citología , Tonsila Palatina/inmunología , Receptores CCR6/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Receptores Toll-Like/inmunología , Interleucina-22
7.
J Immunol ; 188(6): 2612-21, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22312126

RESUMEN

TREM2 is an immunoreceptor expressed on osteoclasts (OC) and microglia that transmits intracellular signals through the adaptor DAP12. Individuals with genetic mutations inactivating TREM2 or DAP12 develop the Nasu-Hakola disease (NHD) with cystic-like lesions of the bone and brain demyelination that lead to fractures and presenile dementia. The mechanisms of this disease are poorly understood. In this study, we report that TREM2-deficient mice have an osteopenic phenotype reminiscent of NHD. In vitro, lack of TREM2 impairs proliferation and ß-catenin activation in osteoclast precursors (OcP) in response to M-CSF. This defect results in accelerated differentiation of OcP into mature OC. Corroborating the importance of a balanced proliferation and differentiation of OcP for bone homeostasis, we show that conditional deletion of ß-catenin in OcP also results in reduced OcP proliferation and accelerated osteoclastogenesis in vitro as well as osteopenia in vivo. These results reveal that TREM2 regulates the rate of osteoclastogenesis and provide a mechanism for the bone pathology in NHD.


Asunto(s)
Huesos/metabolismo , Diferenciación Celular/fisiología , Homeostasis/fisiología , Glicoproteínas de Membrana/metabolismo , Osteoclastos/citología , Receptores Inmunológicos/metabolismo , beta Catenina/metabolismo , Animales , Western Blotting , Huesos/citología , Proliferación Celular , Femenino , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología
8.
Proc Natl Acad Sci U S A ; 107(24): 10961-6, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20534450

RESUMEN

Natural killer-22 (NK-22) cells are a human NK cell subset situated in mucosal-associated lymphoid tissues that specialize in IL-22 secretion in response to IL-23. Here we investigated the cytokine requirements for NK-22 cell expansion. IL-7 maintained the survival of NK-22 cells and IL-22 production in response to IL-23 but was insufficient to induce robust expansion. Proliferation of NK-22 cells was increased markedly by adding either IL-1beta or IL-2 to IL-7 and was even stronger in the presence of IL-1beta plus IL-2. In contrast to IL-7, continuous culture in IL-1beta and IL-2 modified NK-22 cytokine profiles. IL-1beta promoted constitutive IL-22 secretion rather than acute IL-22 production in response to IL-23 and induced IL-17 in some cells. IL-2 reduced secretion of IL-22 and IL-17, increasing production of IFN-gamma and leukemia inhibitory factor. Functional deviation toward IFN-gamma production also was induced by continuous culture in IL-23. These results demonstrate the functional plasticity of NK-22 cells, which may allow flexible responses to different pathogens. Finally, we found that NK-22 cells released the B-cell survival factor, B-cell activating factor belonging to the TNF family (BAFF), suggesting a potential role of NK-22 cells in promoting B-cell-mediated mucosal immunity.


Asunto(s)
Interleucina-1beta/farmacología , Interleucina-2/farmacología , Interleucina-7/farmacología , Interleucinas/biosíntesis , Células Asesinas Naturales/clasificación , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/clasificación , Subgrupos Linfocitarios/inmunología , Factor Activador de Células B/biosíntesis , Secuencia de Bases , Proliferación Celular/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/genética , Cartilla de ADN/genética , Humanos , Técnicas In Vitro , Interferón gamma/biosíntesis , Subunidad p19 de la Interleucina-23/farmacología , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Factor Inhibidor de Leucemia/biosíntesis , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/efectos de los fármacos , Tonsila Palatina/citología , Tonsila Palatina/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Interleucina-22
9.
Blood ; 116(16): 2942-9, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20606167

RESUMEN

Chemokine CC motif receptor-like 2 (CCRL2) is a heptahelic transmembrane receptor that shows the highest degree of homology with CCR1, an inflammatory chemokine receptor. CCRL2 mRNA was rapidly (30 minutes) and transiently (2-4 hours) regulated during dendritic cell (DC) maturation. Protein expression paralleled RNA regulation. In vivo, CCRL2 was expressed by activated DC and macrophages, but not by eosinophils and T cells. CCRL2(-/-) mice showed normal recruitment of circulating DC into the lung, but a defective trafficking of antigen-loaded lung DC to mediastinal lymph nodes. This defect was associated to a reduction in lymph node cellularity and reduced priming of T helper cell 2 response. CCRL2(-/-) mice were protected in a model of ovalbumin-induced airway inflammation, with reduced leukocyte recruitment in the BAL (eosinophils and mononuclear cells) and reduced production of the T helper cell 2 cytokines, interleukin-4 and -5, and chemokines CCL11 and CCL17. The central role of CCRL2 deficiency in DC was supported by the fact that adoptive transfer of CCRL2(-/-) antigen-loaded DC in wild-type animals recapitulated the phenotype observed in knockout mice. These data show a nonredundant role of CCRL2 in lung DC trafficking and propose a role for this receptor in the control of excessive airway inflammatory responses.


Asunto(s)
Movimiento Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Pulmón/citología , Receptores de Quimiocina/inmunología , Alérgenos/inmunología , Animales , Citocinas/inmunología , Células Dendríticas/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Inflamación/inducido químicamente , Ganglios Linfáticos/citología , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores CCR , Receptores de Quimiocina/genética
10.
Acta Neuropathol Commun ; 5(1): 23, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28302159

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that lacks a predictive and broadly applicable biomarker. Continued focus on mutation-specific upstream mechanisms has yet to predict disease progression in the clinic. Utilising cellular pathology common to the majority of ALS patients, we implemented an objective transcriptome-driven approach to develop noninvasive prognostic biomarkers for disease progression. Genes expressed in laser captured motor neurons in direct correlation (Spearman rank correlation, p < 0.01) with counts of neuropathology were developed into co-expression network modules. Screening modules using three gene sets representing rate of disease progression and upstream genetic association with ALS led to the prioritisation of a single module enriched for immune response to motor neuron degeneration. Genes in the network module are important for microglial activation and predict disease progression in genetically heterogeneous ALS cohorts: Expression of three genes in peripheral lymphocytes - LILRA2, ITGB2 and CEBPD - differentiate patients with rapid and slowly progressive disease, suggesting promise as a blood-derived biomarker. TREM2 is a member of the network module and the level of soluble TREM2 protein in cerebrospinal fluid is shown to predict survival when measured in late stage disease (Spearman rank correlation, p = 0.01). Our data-driven systems approach has, for the first time, directly linked microglia to the development of motor neuron pathology. LILRA2, ITGB2 and CEBPD represent peripherally accessible candidate biomarkers and TREM2 provides a broadly applicable therapeutic target for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Microglía/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/genética , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Linfocitos/metabolismo , Linfocitos/patología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microglía/patología , Persona de Mediana Edad , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Pronóstico , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología
11.
Thromb Haemost ; 95(1): 22-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16543957

RESUMEN

Dendritic cells (DC) are professional antigen presenting cells which play a pivotal role in the activation of adaptive immunity. Tissue invasion by pathogens induces the recruitment of blood DC to the site of infection and contributes to their subsequent migration to secondary lymphoid organs. This complex process relies on the expression and regulation of receptors for chemotactic factors on the surface of migrating DC and on the activation of adhesion molecules which allow DC to properly interact with both blood and lymphatic vessels. In the absence of correct tissue localization, DC fail to promote proper immune responses. Therefore, the interaction of DC with endothelial cells represents a fundamental step for DC biology.


Asunto(s)
Movimiento Celular , Células Dendríticas/metabolismo , Endotelio Linfático/metabolismo , Endotelio Vascular/metabolismo , Animales , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Quimiocinas/metabolismo , Factores Quimiotácticos/metabolismo , Humanos , Receptores de Quimiocina/metabolismo , Transducción de Señal
12.
EMBO Mol Med ; 7(4): 464-76, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25762615

RESUMEN

Type I interferons (IFN-I) are implicated in the pathogenesis of systemic lupus erythematosus (SLE). In SLE, immune complexes bind to the CD32a (FcγRIIa) receptor on the surface of plasmacytoid dendritic cells (pDCs) and stimulate the secretion of IFN-I from pDCs. BDCA2 is a pDC-specific receptor that, when engaged, inhibits the production of IFN-I in human pDCs. BDCA2 engagement, therefore, represents an attractive therapeutic target for inhibiting pDC-derived IFN-I and may be an effective therapy for the treatment of SLE. In this study, we show that 24F4A, a humanized monoclonal antibody (mAb) against BDCA2, engages BDCA2 and leads to its internalization and the consequent inhibition of TLR-induced IFN-I by pDCs in vitro using blood from both healthy and SLE donors. These effects were confirmed in vivo using a single injection of 24F4A in cynomolgus monkeys. 24F4A also inhibited pDC activation by SLE-associated immune complexes (IC). In addition to the inhibitory effect of 24F4A through engagement of BDCA2, the Fc region of 24F4A was critical for potent inhibition of IC-induced IFN-I production through internalization of CD32a. This study highlights the novel therapeutic potential of an effector-competent anti-BDCA2 mAb that demonstrates a dual mechanism to dampen pDC responses for enhanced clinical efficacy in SLE.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/inmunología , Células Dendríticas/inmunología , Lectinas Tipo C/antagonistas & inhibidores , Glicoproteínas de Membrana/antagonistas & inhibidores , Células Plasmáticas/inmunología , Receptores de IgG/inmunología , Receptores Inmunológicos/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales de Origen Murino/farmacología , Células Dendríticas/citología , Femenino , Humanos , Lectinas Tipo C/inmunología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Masculino , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Células Plasmáticas/citología , Receptores Inmunológicos/inmunología
13.
Mol Cell Biol ; 33(4): 830-44, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23230271

RESUMEN

To determine talin1's role in osteoclasts, we mated TLN1(fl/fl) mice with those expressing cathepsin K-Cre (CtsK-TLN1) to delete the gene in mature osteoclasts or with lysozyme M-Cre (LysM-TLN1) mice to delete TLN1 in all osteoclast lineage cells. Absence of TLN1 impairs macrophage colony-stimulating factor (M-CSF)-stimulated inside-out integrin activation and cytoskeleton organization in mature osteoclasts. Talin1-deficient precursors normally express osteoclast differentiation markers when exposed to M-CSF and receptor activator of nuclear factor κB (RANK) ligand but attach to substrate and migrate poorly, arresting their development into mature resorptive cells. In keeping with inhibited resorption, CtsK-TLN1 mice exhibit an ∼5-fold increase in bone mass. Osteoclast-specific deletion of Rap1 (CtsK-Rap1), which promotes talin/ß integrin recognition, yields similar osteopetrotic mice. The fact that the osteopetrosis of CtsK-TLN1 and CtsK-Rap1 mice is substantially more severe than that of those lacking αvß3 is likely due to added failed activation of ß1 integrins. In keeping with osteoclast dysfunction, mice in whom talin is deleted late in the course of osteoclastogenesis are substantially protected from ovariectomy-induced osteoporosis and the periarticular osteolysis attending inflammatory arthritis. Thus, talin1 and Rap1 are critical for resorptive function, and their selective inhibition in mature osteoclasts retards pathological bone loss.


Asunto(s)
Osteoclastos/citología , Osteoclastos/patología , Talina/metabolismo , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Femenino , Eliminación de Gen , Integrina alfaVbeta3/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Osteoclastos/metabolismo , Osteopetrosis/genética , Osteopetrosis/metabolismo , Osteopetrosis/patología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Talina/genética , Proteínas de Unión al GTP rap/genética , Proteínas de Unión al GTP rap1/genética
14.
J Clin Invest ; 123(1): 138-49, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23241959

RESUMEN

Acute respiratory infections are responsible for more than 4 million deaths each year. Neutrophils play an essential role in the innate immune response to lung infection. These cells have an armamentarium of pattern recognition molecules and antimicrobial agents that identify and eliminate pathogens. In the setting of infection, neutrophil triggering receptor expressed on myeloid cells 1 (TREM-1) amplifies inflammatory signaling. Here we demonstrate for the first time that TREM-1 also plays an important role in transepithelial migration of neutrophils into the airspace. We developed a TREM-1/3-deficient mouse model of pneumonia and found that absence of TREM-1/3 markedly increased mortality following Pseudomonas aeruginosa challenge. Unexpectedly, TREM-1/3 deficiency resulted in increased local and systemic cytokine production. TREM-1/3-deficient neutrophils demonstrated intact bacterial killing, phagocytosis, and chemotaxis; however, histologic examination of TREM-1/3-deficient lungs revealed decreased neutrophil infiltration of the airways. TREM-1/3-deficient neutrophils effectively migrated across primary endothelial cell monolayers but failed to migrate across primary airway epithelia grown at the air-liquid interface. These data define a new function for TREM-1 in neutrophil migration across airway epithelial cells and suggest that it amplifies inflammation through targeted neutrophil migration into the lung.


Asunto(s)
Pulmón/metabolismo , Glicoproteínas de Membrana/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Neumonía Bacteriana/metabolismo , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Receptores Inmunológicos/metabolismo , Migración Transendotelial y Transepitelial , Animales , Pulmón/microbiología , Pulmón/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Neutrófilos/patología , Neumonía Bacteriana/genética , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/patología , Receptores Inmunológicos/genética , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología , Receptor Activador Expresado en Células Mieloides 1
15.
J Clin Invest ; 121(4): 1497-507, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21403398

RESUMEN

Viral infections have been linked to the onset of type I diabetes (T1D), with viruses postulated to induce disease directly by causing ß cell injury and subsequent release of autoantigens and indirectly via the host type I interferon (IFN-I) response triggered by the virus. Consistent with this, resistance to T1D is associated with polymorphisms that impair the function of melanoma differentiation associated gene-5 (MDA5), a sensor of viral RNA that elicits IFN-I responses. In animal models, triggering of another viral sensor, TLR3, has been implicated in diabetes. Here, we found that MDA5 and TLR3 are both required to prevent diabetes in mice infected with encephalomyocarditis virus strain D (EMCV-D), which has tropism for the insulin-producing ß cells of the pancreas. Infection of Tlr3-/- mice caused diabetes due to impaired IFN-I responses and virus-induced ß cell damage rather than T cell-mediated autoimmunity. Mice lacking just 1 copy of Mda5 developed transient hyperglycemia when infected with EMCV-D, whereas homozygous Mda5-/- mice developed severe cardiac pathology. TLR3 and MDA5 controlled EMCV-D infection and diabetes by acting in hematopoietic and stromal cells, respectively, inducing IFN-I responses at kinetically distinct time points. We therefore conclude that optimal functioning of viral sensors and prompt IFN-I responses are required to prevent diabetes when caused by a virus that infects and damages the ß cells of the pancreas.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Virus de la Encefalomiocarditis/patogenicidad , Interferón Tipo I/biosíntesis , ARN Viral/inmunología , Animales , Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/virología , Helicasa Inducida por Interferón IFIH1 , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología
16.
J Immunol ; 174(5): 3032-40, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15728517

RESUMEN

Although blood monocytes possess significant cytotoxic activity against tumor cells, tumor-infiltrating monocytes are commonly deactivated in cancer patients. Monocytes pre-exposed to tumor cells show significantly decreased expression levels of TNF-alpha, IL-12p40, and IL-1R-associated kinase (IRAK)-1. Activation of the Ser/Thr kinase IRAK-1 is an important event in several inflammatory processes. By contrast, another IRAK family member, IRAK-M, negatively regulates this pathway, and is up-regulated in cultures of endotoxin-tolerant monocytes and in monocytes from septic patients within the timeframe of tolerance. In this study, we show that IRAK-M expression is enhanced at the mRNA and protein level in human monocytes cultured in the presence of tumor cells. IRAK-M was induced in monocytes upon coculturing with different tumor cells, as well as by fixed tumor cells and medium supplemented with the supernatant from tumor cell cultures. Moreover, blood monocytes from patients with chronic myeloid leukemia and patients with metastasis also overexpressed IRAK-M. Low concentrations of hyaluronan, a cell surface glycosaminoglycan released by tumor cells, also up-regulated IRAK-M. The induction of IRAK-M by hyaluronan and tumor cells was abolished by incubation with anti-CD44 or anti-TLR4 blocking Abs. Furthermore, down-regulation of IRAK-M expression by small interfering RNAs specific for IRAK-M reinstates both TNF-alpha mRNA expression and protein production in human monocytes re-exposed to a tumor cell line. Altogether, our findings indicate that deactivation of human monocytes in the presence of tumor cells involves IRAK-M up-regulation, and this effect appears to be mediated by hyaluronan through the engagement of CD44 and TLR4.


Asunto(s)
Receptores de Hialuranos/fisiología , Glicoproteínas de Membrana/fisiología , Monocitos/inmunología , Monocitos/metabolismo , Proteínas Quinasas/biosíntesis , Receptores de Superficie Celular/fisiología , Receptores de Interleucina-1/biosíntesis , Regulación hacia Arriba/inmunología , Adulto , Anticuerpos Bloqueadores/farmacología , Comunicación Celular/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Regulación hacia Abajo/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Células HL-60 , Células HeLa , Humanos , Receptores de Hialuranos/inmunología , Ácido Hialurónico/farmacología , Quinasas Asociadas a Receptores de Interleucina-1 , Células Jurkat , Activación de Macrófagos/inmunología , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/inmunología , Receptor Toll-Like 4 , Receptores Toll-Like , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
17.
J Immunol ; 170(5): 2279-82, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12594248

RESUMEN

In an effort to define the actual function of the promiscuous putatively silent chemokine receptor D6, transfectants were generated in different cell types. Engagement of D6 by inflammatory CC chemokines elicited no calcium response nor chemotaxis, but resulted in efficient agonist internalization and degradation. Also in lymphatic endothelium, where this receptor is expressed in vivo, D6 did not elicit cellular responses other than ligand internalization and degradation. In particular, no evidence was obtained for D6-mediated transcytosis of chemokines in the apical-to-basal or basal-to-apical directions. These results indicate that D6 acts as an inflammatory chemokine scavenging nonactivatory decoy receptors and suggest that in lymphatic vessels D6 may function as a gatekeeper for inflammatory CC chemokines, by clearing them and preventing excessive diffusion via afferent lymphatics to lymph nodes.


Asunto(s)
Quimiocinas CC/metabolismo , Receptores de Quimiocina/fisiología , Animales , Células CHO , Células Cultivadas , Cricetinae , Endotelio Linfático/citología , Endotelio Linfático/inmunología , Endotelio Linfático/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Radioisótopos de Yodo/metabolismo , Ligandos , Ratones , Ratones Endogámicos DBA , Unión Proteica/inmunología , Ensayo de Unión Radioligante , Receptores CCR10 , Receptores de Quimiocina/biosíntesis , Transducción de Señal/inmunología , Transfección , Células Tumorales Cultivadas , Receptor de Quimiocina D6
18.
Blood ; 104(2): 444-52, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15026312

RESUMEN

The chemokine receptor CXCR4 and its functional ligand, CXCL12, are essential regulators of development and homeostasis of hematopoietic and lymphoid organs. Heterozygous truncating mutations in the CXCR4 intracellular tail cause a rare genetic disease known as WHIM syndrome (warts, hypogammaglobulinemia, infections, myelokathexis), whose pathophysiology remains unclear. We report CXCR4 function in 3 patients with WHIM syndrome carrying heterozygous truncating mutations of CXCR4. We show that CXCR4 gene mutations in WHIM patients do not affect cell surface expression of the chemokine receptor and its internalization upon stimulation with CXCL12. Moreover, no significant differences in calcium mobilization in response to CXCL12 are found. However, the chemotactic response of both polymorphonuclear cells and T lymphocytes in response to CXCL12 is increased. Furthermore, immunophenotypic analysis of circulating T and B lymphocytes reveals a decreased number of memory B cells and of naive T cells and an accumulation of effector memory T cells associated with a restricted T-cell repertoire. Based on our results, we suggest that the altered leukocyte response to CXCL12 may account for the pathologic retention of mature polymorphonuclear cells in the bone marrow (myelokathexis) and for an altered lymphocyte trafficking, which may cause the immunophenotyping abnormalities observed in WHIM patients.


Asunto(s)
Agammaglobulinemia/inmunología , Infecciones Bacterianas/inmunología , Quimiocinas CXC/genética , Neutropenia/inmunología , Verrugas/inmunología , Adolescente , Adulto , Agammaglobulinemia/genética , Linfocitos B/citología , Linfocitos B/inmunología , Infecciones Bacterianas/genética , Médula Ósea/inmunología , Calcio/metabolismo , Quimiocina CXCL12 , Niño , Análisis Mutacional de ADN , Femenino , Citometría de Flujo , Expresión Génica/inmunología , Humanos , Inmunofenotipificación , Neutropenia/genética , Neutrófilos/citología , Neutrófilos/inmunología , Síndrome , Linfocitos T/citología , Linfocitos T/inmunología , Verrugas/genética
19.
EMBO J ; 23(17): 3505-15, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15318168

RESUMEN

Gene-targeted mice were used to evaluate the role of the gamma isoform of phosphoinositide 3-kinase (PI3Kgamma) in dendritic cell (DC) migration and induction of specific T-cell-mediated immune responses. DC obtained from PI3Kgamma-/- mice showed a reduced ability to respond to chemokines in vitro and ex vivo and to travel to draining lymph nodes under inflammatory conditions. PI3Kgamma-/- mice had a selective defect in the number of skin Langerhans cells and in lymph node CD8alpha- DC. Furthermore, PI3Kgamma-/- mice showed a defective capacity to mount contact hypersensitivity and delayed-type hypersensitivity reactions. This defect was directly related to the reduced ability of antigen-loaded DC to migrate from the periphery to draining lymph nodes. Thus, PI3Kgamma plays a nonredundant role in DC trafficking and in the activation of specific immunity. Therefore, PI3Kgamma may be considered a new target to control exaggerated immune reactions.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/fisiología , Isoenzimas/deficiencia , Fosfatidilinositol 3-Quinasas/deficiencia , Animales , Movimiento Celular , Quimiocinas/farmacología , Fosfatidilinositol 3-Quinasa Clase Ib , Células Dendríticas/clasificación , Células Dendríticas/efectos de los fármacos , Dermatitis por Contacto , Hipersensibilidad Tardía , Técnicas In Vitro , Isoenzimas/genética , Isoenzimas/inmunología , Isoenzimas/fisiología , Células de Langerhans/efectos de los fármacos , Células de Langerhans/inmunología , Células de Langerhans/fisiología , Masculino , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/fisiología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA