Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Plast Reconstr Surg ; 139(4): 933e-940e, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28350671

RESUMEN

BACKGROUND: Inflammation is integral to the injury response. The inflammatory response is essential to the host defense against infection and also to tissue regeneration and repair. Toll-like receptors (TLRs) are critical activators of the innate immune response and present attractive therapeutic targets for inflammation-modulated tissue regeneration. The authors' previous study showed that depletion of TLR4 resulted in accelerated skull bone healing concurrent with increased expression of osteoclastogenic genes. As such, in the present study, the authors used various knockout mouse models for TLR4 and its associated signaling mediators as tools to further understand the role of Toll-like receptor-mediated inflammation in calvarial bone healing. METHODS: Calvarial defects (1.8-mm diameter) were created in wild-type, TLR4 knockout (TLR4), TLR2, MyD88, TRIF, TLR4 knockout in myeloid cell (Lyz-TLR4), and TLR4 knockout in dendritic-lineage cell (CD11c-TLR4) mice. Bone healing was examined using micro-computed tomographic, histologic, and histomorphometric analyses. RESULTS: Micro-computed tomographic and histomorphometric analyses revealed that TLR4-deficient mice (TLR4, Lyz-TLR4, and CD11c-TLR4) exhibited a faster intramembraneous healing response at postoperative day 7, whereas MyD88 and CD11c-TLR4 mice showed enhanced bone healing at day 28. CONCLUSIONS: The authors' data suggest a detrimental role for TLR4 in CD11c cells, mediated by Myd88 signaling, during calvarial bone healing. The authors have demonstrated that Toll-like receptor signaling components affect calvarial bone healing, establishing a link between the skeletal and immune systems during craniofacial bone healing. Toll-like receptor signaling components might be used to initiate enhanced healing in bone defects to improve clinical outcomes.


Asunto(s)
Antígeno CD11c/fisiología , Factor 88 de Diferenciación Mieloide/fisiología , Cráneo/lesiones , Receptor Toll-Like 4/fisiología , Cicatrización de Heridas/fisiología , Animales , Antígeno CD11c/genética , Femenino , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Transducción de Señal , Receptor Toll-Like 4/genética
2.
Gastroenterology ; 145(6): 1347-57, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23973919

RESUMEN

BACKGROUND & AIMS: Intestinal epithelial cells aid in mucosal defense by providing a physical barrier against entry of pathogenic bacteria and secreting antimicrobial peptides (AMPs). Autophagy is an important component of immune homeostasis. However, little is known about its role in specific cell types during bacterial infection in vivo. We investigated the role of autophagy in the response of intestinal epithelial and antigen-presenting cells to Salmonella infection in mice. METHODS: We generated mice deficient in Atg16l1 in epithelial cells (Atg16l1(f/f) × Villin-cre) or CD11c(+) cells (Atg16l1(f/f) × CD11c-cre); these mice were used to assess cell type-specific antibacterial autophagy. All responses were compared with Atg16l1(f/f) mice (controls). Mice were infected with Salmonella enterica serovar typhimurium; cecum and small-intestine tissues were collected for immunofluorescence, histology, and quantitative reverse-transcription polymerase chain reaction analyses of cytokines and AMPs. Modulators of autophagy were screened to evaluate their effects on antibacterial responses in human epithelial cells. RESULTS: Autophagy was induced in small intestine and cecum after infection with S typhimurium, and required Atg16l1. S typhimurium colocalized with microtubule-associated protein 1 light chain 3ß (Map1lc3b or LC3) in the intestinal epithelium of control mice but not in Atg16l1(f/f) × Villin-cre mice. Atg16l1(f/f) × Villin-cre mice also had fewer Paneth cells and abnormal granule morphology, leading to reduced expression of AMPs. Consistent with these defective immune responses, Atg16l1(f/f) × Villin-cre mice had increased inflammation and systemic translocation of bacteria compared with control mice. In contrast, we observed few differences between Atg16l1(f/f) × CD11c-cre and control mice. Trifluoperazine promoted autophagy and bacterial clearance in HeLa cells; these effects were reduced upon knockdown of ATG16L1. CONCLUSIONS: Atg16l1 regulates autophagy in intestinal epithelial cells and is required for bacterial clearance. It also is required to prevent systemic infection of mice with enteric bacteria.


Asunto(s)
Autofagia/fisiología , Proteínas Portadoras/fisiología , Mucosa Intestinal/fisiología , Salmonelosis Animal/prevención & control , Animales , Proteínas Relacionadas con la Autofagia , Antígeno CD11c/fisiología , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Células HeLa , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Salmonelosis Animal/patología , Salmonelosis Animal/fisiopatología , Salmonella typhimurium/aislamiento & purificación
3.
J Immunol ; 191(1): 25-9, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23733880

RESUMEN

Dendritic cell (DC)-T cell interactions that underlie inducible/adaptive regulatory T cell generation and airway tolerance are not well understood. In this study, we show that mice lacking CD11c(hi) lung DCs, but containing plasmacytoid DCs (pDCs), fail tolerization with inhaled Ag and cannot support Foxp3 induction in vivo in naive CD4(+) T cells. CD103(+) DCs from tolerized mice efficiently induced Foxp3 in cocultured naive CD4(+) T cells but pDCs and lung macrophages failed to do so. CD103(+) DCs, but not pDCs or lung macrophages, upregulated the expression of retinaldehyde dehydrogenase 2 (aldh1a2), which is key for the production of retinoic acid, a cofactor for TGF-ß for Foxp3 induction. Batf3(-/-) mice, selectively lacking CD103(+) DCs, failed tolerization by inhaled Ag. Collectively, our data show that pulmonary tolerance is dependent on CD103(+) DCs, correlating with their ability to upregulate aldh1a2, which can promote Foxp3 expression in T cells.


Asunto(s)
Aldehído Deshidrogenasa/biosíntesis , Antígenos CD/biosíntesis , Antígeno CD11c/fisiología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/biosíntesis , Cadenas alfa de Integrinas/biosíntesis , Pulmón/inmunología , Regulación hacia Arriba/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/enzimología , Células Dendríticas/metabolismo , Tolerancia Inmunológica/inmunología , Pulmón/enzimología , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
4.
J Immunol ; 189(8): 3869-77, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22972929

RESUMEN

Patients with allergic asthma have more lung mast cells, which likely worsens the symptoms. In experimental asthma, CD11c(+) cells have to be present during the challenge phase for several features of allergic inflammation to occur. Whether CD11c(+) cells play a role for Ag-induced increases of lung mast cells is unknown. In this study, we used diphtheria toxin treatment of sensitized CD11c-diphtheria toxin receptor transgenic mice to deplete CD11c(+) cells. We demonstrate that recruitment of mast cell progenitors to the lung is substantially reduced when CD11c(+) cells are depleted during the challenge phase. This correlated with an impaired induction of endothelial VCAM-1 and led to a significantly reduced number of mature mast cells 1 wk after challenge. Collectively, these data suggest that Ag challenge stimulates CD11c(+) cells to produce cytokines and/or chemokines required for VCAM-1 upregulation on the lung endothelium, which in turn is crucial for the Ag-induced mast cell progenitor recruitment and the increase in mast cell numbers.


Asunto(s)
Antígeno CD11c/fisiología , Movimiento Celular/inmunología , Pulmón/citología , Pulmón/inmunología , Mastocitos/citología , Mastocitos/inmunología , Regulación hacia Arriba/inmunología , Animales , Antígeno CD11c/biosíntesis , Antígeno CD11c/genética , Quimiocinas/biosíntesis , Quimiocinas/fisiología , Toxina Diftérica/administración & dosificación , Femenino , Humanos , Recuento de Leucocitos , Pulmón/metabolismo , Depleción Linfocítica , Masculino , Mastocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Células Madre/citología , Células Madre/inmunología , Células Madre/metabolismo , Regulación hacia Arriba/genética , Molécula 1 de Adhesión Celular Vascular/biosíntesis
5.
Eur J Immunol ; 42(6): 1459-67, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22622847

RESUMEN

Under homeostatic conditions, skin DCs migrate to regional LNs transporting self-antigens (self-Ags). The transport of self-Ags is considered to be critical for maintaining peripheral tolerance. Although the chemokine receptor CCR7 potently induces the migration of skin DCs to regional LNs, Ccr7(-/-) (Ccr7-KO) mice do not show skin auto-immune diseases. To resolve this inconsistency, we examined Ccr7-KO epidermis- or dermis-hyperpigmented transgenic (Tg) mice, in which the transport of skin self-Ags is traceable by melanin granules (MGs). Under CCR7-deficient conditions, the transport of epidermal MGs to regional LNs was impaired at 7 weeks of age. However, epidermal MGs could be transported when they had accumulated in the dermis. Ccr7-KO-dermis-pigmented Tg mice confirmed the presence of CCR7-independent transport from the dermis. Compared with WT-dermis-pigmented Tg mice, the amount of transported melanin and number of MG-laden CD11c(+) cells were both approximately 40% of the WT levels, while the number of MG-laden CD205(+) or CD207(+) cells decreased to about 10% in skin regional LNs of Ccr7-KO-dermis-pigmented Tg mice. Cell sorting highlighted the involvement of CD11c(+) cells in the CCR7-independent transport. Here, we show that CCR7-independent transport of skin self-Ags occurs in the dermis. This system might contribute to the continuous transport of self-Ags, and maintain peripheral tolerance.


Asunto(s)
Autoantígenos/metabolismo , Dermis/inmunología , Receptores CCR7/fisiología , Animales , Transporte Biológico , Antígeno CD11c/análisis , Antígeno CD11c/fisiología , Tolerancia Inmunológica , Ganglios Linfáticos/inmunología , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Células Madre/fisiología
6.
Ross Fiziol Zh Im I M Sechenova ; 97(6): 601-8, 2011 Jun.
Artículo en Ruso | MEDLINE | ID: mdl-21874872

RESUMEN

The role of beta2-integrins CD11b/CD18 and CD 11c/CD 18 in adhesion and migration of leukocytes on fibrinogen was studied. The monoclonal antibodies against CD11b inhibited the spontaneous adhesion of monocytic THP-1 cells on fibrinogen, whereas antibodies to CD11c more effectively inhibited the adhesion stimulated by chemokine MCP-1. By the RNA-interference method the clones of THP-1 with reduced expression of CD11b and general beta2-subunit CD18 were obtained. MCP-I stimulated the adhesion to fibrinogen of THP-1 cells of wild-type and mutant cells with reduced expression of CD11b (THP-1-CD11b-low), but not of cells with low expression of CD18 (THP-1-CD18-low). THP-1-CD18-low cells were also characterized by the impaired chemotaxis in presence of MCP-1. The data obtained suggest that spontaneous cell adhesion to fibrinogen is mediated to a greater extent by CD11b/CD18 integrins, while chemokine-stimulated adhesion and migration is mostly dependent on CD11c/CD18 molecules.


Asunto(s)
Antígeno CD11b/fisiología , Antígeno CD11c/fisiología , Movimiento Celular/fisiología , Antígeno CD11b/genética , Antígeno CD11c/genética , Antígenos CD18/genética , Antígenos CD18/fisiología , Adhesión Celular/genética , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , Fibrinógeno/metabolismo , Humanos , Leucocitos/fisiología , Monocitos/fisiología
7.
J Immunol ; 186(4): 2003-12, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21239711

RESUMEN

γδ T cells are considered to be innate lymphocytes that play an important role in host defense against tumors and infections. We recently reported that IL-18 markedly amplified γδ T cell responses to zoledronate (ZOL)/IL-2. In an extension of this finding, we analyzed the mechanism underlying the IL-18-mediated expansion of γδ T cells. After incubation of PBMCs with ZOL/IL-2/IL-18, the majority of the cells expressed γδ TCR, and the rest mostly exhibited CD56(bright)CD11c(+) under the conditions used in this study. CD56(bright)CD11c(+) cells were derived from a culture of CD56(int)CD11c(+) cells and CD14(+) cells in the presence of IL-2 and IL-18 without the addition of ZOL. They expressed IL-18Rs, HLA-DR, CD25, CD80, CD83, CD86, and CD11a/CD18. In addition, they produced IFN-γ, TNF-α, but not IL-12, when treated with IL-2/IL-18, and they exerted cytotoxicity against K562 cells, thus exhibiting characteristics of both NK cells and dendritic cells. Incubation of purified γδ T cells with CD56(bright)CD11c(+) cells in the presence of ZOL/IL-2/IL-18 resulted in the formation of massive cell clusters and led to the marked expansion of γδ T cells. However, both conventional CD56(-/int)CD11c(high) dendritic cells induced by GM-CSF/IL-4 and CD56(+)CD11c(-) NK cells failed to support the expansion of γδ T cells. These results strongly suggest that CD56(bright)CD11c(+) cells play a key role in the IL-18-mediated proliferation of γδ T cells.


Asunto(s)
Antígeno CD11c/biosíntesis , Antígeno CD56/metabolismo , Proliferación Celular , Interleucina-18/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta/biosíntesis , Subgrupos de Linfocitos T/inmunología , Adulto , Antígeno CD11c/fisiología , Antígeno CD56/fisiología , Diferenciación Celular/inmunología , Células Cultivadas , Células Clonales , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Inmunofenotipificación , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo
8.
J Immunol ; 185(1): 532-41, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20505139

RESUMEN

The selective targeting of the tumor-associated death-inducing receptors DR4 and DR5 with agonistic mAbs has demonstrated preclinical and clinical antitumor activity. However, the cellular and molecular mechanisms contributing to this efficacy remain poorly understood. In this study, using the first described C57BL/6 (B6) TRAIL-sensitive experimental tumor models, we have characterized the innate and adaptive immune components involved in the primary rejection phase of an anti-mouse DR5 (mDR5) mAb, MD5-1 in established MC38 colon adenocarcinomas. FcR mediated cross-linking of MD5-1 significantly inhibited the growth of MC38 colon adenocarcinomas through the induction of TRAIL-R-dependent tumor cell apoptosis. The loss of host DR5, TRAIL, perforin, FasL, or TNF did not compromise anti-DR5 therapy in vivo. By contrast, anti-DR5 therapy was completely abrogated in mice deficient of B cells or CD11c(+) dendritic cells (DCs), providing the first direct evidence that these cells play a critical role. Importantly, the requirement for an intact B cell compartment for optimal anti-DR5 antitumor efficacy was also observed in established AT-3 mammary tumors. Interestingly, MD5-1-mediated apoptosis as measured by early TUNEL activity was completely lost in B cell-deficient microMT mice, but intact in mice deficient in CD11c(+) DCs. Overall, these data show that Ab-mediated targeting of DR5 triggers tumor cell apoptosis in established tumors in a B cell-dependent manner and that CD11c(+) DCs make a critical downstream contribution to anti-DR5 antitumor activity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Subgrupos de Linfocitos B/inmunología , Antígeno CD11c/fisiología , Células Dendríticas/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma/terapia , Animales , Anticuerpos Monoclonales/toxicidad , Subgrupos de Linfocitos B/metabolismo , Antígeno CD11c/biosíntesis , Línea Celular , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/inmunología , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/terapia , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/terapia , Mastocitoma/inmunología , Mastocitoma/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/deficiencia , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética
9.
J Clin Invest ; 120(4): 1324-36, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20200448

RESUMEN

Peptide loading of MHC class II (MHCII) molecules is directly catalyzed by the MHCII-like molecule HLA-DM (DM). Another MHCII-like molecule, HLA-DO (DO), associates with DM, thereby modulating DM function. The biological role of DO-mediated regulation of DM activity in vivo remains unknown; however, it has been postulated that DO expression dampens presentation of self antigens, thereby preventing inappropriate T cell activation that ultimately leads to autoimmunity. To test the idea that DO modulation of the MHCII self-peptide repertoire mediates self tolerance, we generated NOD mice that constitutively overexpressed DO in DCs (referred to herein as NOD.DO mice). NOD mice are a mouse model for type 1 diabetes, an autoimmune disease mediated by the destruction of insulin-secreting pancreatic beta cells. Our studies showed that diabetes development was completely blocked in NOD.DO mice. Similar to NOD mice, NOD.DO animals selected a diabetogenic T cell repertoire, and the numbers and function of Tregs were normal. Indeed, immune system function in NOD.DO mice was equivalent to that in NOD mice. NOD.DO DCs, however, presented an altered MHCII-bound self-peptide repertoire, thereby preventing the activation of diabetogenic T cells and subsequent diabetes development. These studies show that DO expression can shape the overall MHCII self-peptide repertoire to promote T cell tolerance.


Asunto(s)
Presentación de Antígeno , Autoantígenos/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Inmunocompetencia , Animales , Antígeno CD11c/fisiología , Diabetes Mellitus Tipo 1/inmunología , Femenino , Antígenos HLA-D/fisiología , Antígenos de Histocompatibilidad Clase II/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T Reguladores/inmunología
10.
J Immunol ; 184(8): 4247-57, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20231692

RESUMEN

Lymph node expansion during immune responses is accompanied by rapid vascular expansion. The re-establishment of quiescence and stabilization of the newly expanded vasculature and the regulatory mechanisms involved have not been well studied. We show that although initiation of vascular expansion in immune-stimulated nodes is associated with upregulated endothelial cell proliferation, increased high endothelial venule trafficking efficiency and VCAM-1 expression, and disrupted perivascular fibroblastic reticular cell organization, the re-establishment of vascular quiescence and stabilization postexpansion is characterized by reversal of these phenomena. Although CD11c(med) cells are associated with the initiation of vascular expansion, CD11c(hi)MHC class II (MHC II)(med) dendritic cells (DCs) accumulate later, and their short-term depletion in mice abrogates the re-establishment of vascular quiescence and stabilization. CD11c(hi)MHC II(med) cells promote endothelial cell quiescence in vitro and, in vivo, mediate quiescence at least in part by mediating reduced lymph node vascular endothelial growth factor. Disrupted vascular quiescence and stabilization in expanded nodes is associated with attenuated T cell-dependent B cell responses. These results describe a novel mechanism whereby CD11c(hi)MHC II(med) DCs regulate the re-establishment of vascular quiescence and stabilization after lymph node vascular expansion and suggest that these DCs function in part to orchestrate the microenvironmental alterations required for successful immunity.


Asunto(s)
Antígeno CD11c/fisiología , Células Dendríticas/inmunología , Endotelio Vascular/inmunología , Antígenos de Histocompatibilidad Clase II/biosíntesis , Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/inmunología , Vasos Linfáticos/inmunología , Activación de Linfocitos/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígeno CD11c/biosíntesis , Línea Celular Tumoral , Movimiento Celular/inmunología , Proliferación Celular , Células Cultivadas , Células Dendríticas/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Ganglios Linfáticos/citología , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transporte de Proteínas/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Regulación hacia Arriba/inmunología
11.
Int J Cancer ; 126(4): 896-908, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19653277

RESUMEN

The development of autochtonous mammary tumors in HER-2/neu transgenic mice is facilitated by immune tolerance to the neu-transgene. However, appropriate vaccination strategies can initiate immune system-mediated antitumor response by a process that requires IFN-gamma. We investigated the role of inducible nitric oxide synthase (iNOS) induction by IFN-gamma to promote tumor cell apoptosis. Tumors from FVBN202 mice expressing the normal neu gene under the control of the MMTV-LTR were treated in slice cultures with IFN-gamma for up to 24 hr. Apoptosis was induced, which depended on iNOS enzymatic activity. iNOS expression was predominantly found in infiltrating CD11b(+)CD11c(+) myeloid cells and at much lower levels in the tumor epithelium. By contrast, IFN-gamma treatment of explant cultures of tumor epithelial cells was not sufficient to efficiently induce iNOS, emphasizing an important role of the integrity of tumor tissue architecture, which was preserved in the slice cultures. This notion was further supported by the upregulation of iNOS costimulatory cytokines TNF-alpha and IL-1beta in slice cultures but not in explants and the capability of purified CD11b(+)CD11c(+) cells to enhance iNOS expression of tumor cells in cocultures. The findings suggest that tumor-infiltrating myeloid cells in immuno-tolerant HER-2/neu transgenic mice possess tumor killing ability via induction of iNOS and underline the capacity of antitumor strategies designed to stimulate infiltrating myeloid cells.


Asunto(s)
Antígeno CD11b/fisiología , Antígeno CD11c/fisiología , Neoplasias Mamarias Experimentales/patología , Óxido Nítrico Sintasa de Tipo II/genética , Receptor ErbB-2/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis/inmunología , Apoptosis/fisiología , Muerte Celular/genética , Línea Celular Tumoral , Femenino , Amplificación de Genes , Interferón gamma/inmunología , Interferón gamma/farmacología , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Transgénicos , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Factor de Necrosis Tumoral alfa/inmunología
12.
J Exp Med ; 206(13): 3073-88, 2009 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-20008527

RESUMEN

The inflammatory response is one of several host alert mechanisms that recruit neutrophils from the circulation to the area of infection. We demonstrate that Bordetella, a bacterial pathogen, exploits an antiinflammatory cytokine, interleukin-10 (IL-10), to evade the host immune system. We identified a Bordetella effector, BopN, that is translocated into the host cell via the type III secretion system, where it induces enhanced production of IL-10. Interestingly, the BopN effector translocates itself into the nucleus and is involved in the down-regulation of mitogen-activated protein kinases. Using pharmacological blockade, we demonstrated that BopN-induced IL-10 production is mediated, at least in part, by its ability to block the extracellular signal-regulated kinase pathway. We also showed that BopN blocks nuclear translocation of nuclear factor kappaB p65 (NF-kappaBp65) but, in contrast, promotes nuclear translocation of NF-kappaBp50. A BopN-deficient strain was unable to induce IL-10 production in mice, resulting in the elimination of bacteria via neutrophil infiltration into the pulmonary alveoli. Furthermore, IL-10-deficient mice effectively eliminated wild-type as well as BopN mutant bacteria. Thus, Bordetella exploits BopN as a stealth strategy to shut off the host inflammatory reaction. These results explain the ability of Bordetella species to avoid induction of the inflammatory response.


Asunto(s)
Proteínas Bacterianas/fisiología , Bordetella/inmunología , Evasión Inmune , Interleucina-10/biosíntesis , Animales , Bordetella/patogenicidad , Antígeno CD11c/fisiología , Interleucina-10/genética , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología , ARN Mensajero/análisis , Transducción de Señal , Tráquea/microbiología , Factores de Virulencia/fisiología
13.
J Immunol ; 183(1): 209-20, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19542432

RESUMEN

Allergic inflammation in the airway is generally considered a Th2-type immune response. However, recent studies demonstrated that Th1- and Th17-type immune responses also play important roles in this process. IFN-gamma is a Th1-type cytokine that generally counteracts the Th2 response. Although previous studies suggest that exogenous IFN-gamma suppresses allergic airway inflammation, the mechanism of suppression has not been fully clarified. In this study, we elucidated whether IFN-gamma suppresses Ag-induced immune responses including the production of Th1- and Th17-type cytokines in the lung, and examined its mechanism of action. BALB/c mice were sensitized and challenged with OVA-Ag to induce airway inflammation. An IFN-gamma-producing plasmid vector was delivered before systemic Ag sensitization. IFN-gamma suppressed indicators of Th2-type immune responses such as airway eosinophilia, IL-5 and IL-13 production in the lung, and bronchial mucus production. Moreover, IFN-gamma also suppressed the production of IL-17 and IFN-gamma itself. The suppression was not mediated by inducing regulatory T cells or by inducing apoptosis in immunocytes. Instead, IFN-gamma suppressed the Ag-presenting capacity and cytokine production of splenic dendritic cells and thus subsequently suppressed OVA-induced activation of CD4(+) T cells. Furthermore, IFN-gamma also attenuated allergic airway inflammation when delivered during the OVA challenge. Various functions of lung CD11c(+) APCs and their migration to regional lymph nodes were also suppressed. These results suggest that the Th1 cytokine IFN-gamma has broad immune regulatory potential through suppressing APC functions. They also suggest that delivery of IFN-gamma could be an effective strategy for regulating Ag-induced immune responses in the lung.


Asunto(s)
Antígenos/administración & dosificación , Antígenos/inmunología , Citocinas/antagonistas & inhibidores , Interferón gamma/fisiología , Pulmón/inmunología , Células TH1/inmunología , Animales , Células Presentadoras de Antígenos/metabolismo , Antígeno CD11c/biosíntesis , Antígeno CD11c/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Citocinas/fisiología , Interferón gamma/biosíntesis , Interferón gamma/genética , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Plásmidos/administración & dosificación , Plásmidos/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/prevención & control , Células TH1/metabolismo
14.
Circulation ; 119(20): 2708-17, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19433759

RESUMEN

BACKGROUND: Monocyte activation and migration into the arterial wall are key events in atherogenesis associated with hypercholesterolemia. CD11c/CD18, a beta2 integrin expressed on human monocytes and a subset of mouse monocytes, has been shown to play a distinct role in human monocyte adhesion on endothelial cells, but the regulation of CD11c in hypercholesterolemia and its role in atherogenesis are unknown. METHODS AND RESULTS: Mice genetically deficient in CD11c were generated and crossbred with apolipoprotein E (apoE)-/- mice to generate CD11c-/-/apoE-/- mice. Using flow cytometry, we examined CD11c on blood leukocytes in apoE-/- hypercholesterolemic mice and found that compared with wild-type and apoE-/- mice on a normal diet, apoE-/- mice on a Western high-fat diet had increased CD11c+ monocytes. Circulating CD11c+ monocytes from apoE-/- mice fed a high-fat diet exhibited cytoplasmic lipid vacuoles and expressed higher levels of CD11b and CD29. Deficiency of CD11c decreased firm arrest of mouse monocytes on vascular cell adhesion molecule-1 and E-selectin in a shear flow assay, reduced monocyte/macrophage accumulation in atherosclerotic lesions, and decreased atherosclerosis development in apoE-/- mice on a high-fat diet. CONCLUSIONS: CD11c, which increases on blood monocytes during hypercholesterolemia, plays an important role in monocyte recruitment and atherosclerosis development in an apoE-/- mouse model of hypercholesterolemia.


Asunto(s)
Aterosclerosis/etiología , Antígeno CD11c/fisiología , Hipercolesterolemia/complicaciones , Monocitos/fisiología , Animales , Apolipoproteínas E/deficiencia , Antígeno CD11c/análisis , Antígeno CD11c/genética , Quimiotaxis de Leucocito , Selectina E/metabolismo , Ratones , Ratones Noqueados , Monocitos/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
Eur Respir J ; 34(3): 749-56, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19282343

RESUMEN

The present study verified the hypothesis that enhanced maturation of antigen-presenting CD11c(+) cells could explain the viral-induced exacerbated immune response to Saccharopolyspora rectivirgula (SR), the main antigen responsible for farmer's lung, a classic form of hypersensitivity pneumonitis (HP). Four groups of mice were studied: group 1 received intranasal instillations of saline; group 2 received instillations of SR for 12 weeks; group 3 received instillations of saline and a single infection with Sendai virus on week 3; and group 4 received instillations of SR for 12 weeks with a single administration of Sendai virus on week 3. On week 13, mice were sacrificed and bronchoalveolar lavage was performed. Lungs were harvested, digested with enzymes, and CD11c(+) cells were analysed in flow cytometry with anti-CD11c, anti-CD86 and anti-major histocompatibility complex class II markers. Immunofluorescence studies were also performed with the same cell surface markers. Both flow cytometry and immunofluorescence results demonstrate that mature CD11c(+) cells are significantly enhanced in SR-challenged mice simultaneously infected with Sendai virus, compared with other groups. These CD11c(+) cells persist in the lung for 9 weeks after the virus infection. Maturation of CD11c(+) cells could explain, at least in part, the virus-induced increased immune response to SR antigens in this model of HP, but mechanisms have still to be elucidated.


Asunto(s)
Alveolitis Alérgica Extrínseca/inmunología , Alveolitis Alérgica Extrínseca/patología , Antígenos Bacterianos/inmunología , Antígeno CD11c/fisiología , Saccharopolyspora/inmunología , Alveolitis Alérgica Extrínseca/microbiología , Animales , Antígeno B7-2/metabolismo , Modelos Animales de Enfermedad , Femenino , Antígenos HLA-D/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones por Respirovirus/complicaciones , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/metabolismo , Virus Sendai/inmunología
16.
Blood ; 112(12): 4546-54, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18812469

RESUMEN

Interleukin-15 (IL-15) is crucial for the development of naive and memory CD8 T cells and is delivered through a mechanism called transpresentation. Previous studies showed that memory CD8 T cells require IL-15 transpresentation by an as yet unknown cell of hematopoietic origin. We hypothesized that dendritic cells (DCs) transpresent IL-15 to CD8 T cells, and we examined this by developing a transgenic model that limits IL-15 transpresentation to DCs. In this study, IL-15 transpresentation by DCs had little effect on restoring naive CD8 T cells but contributed to the development of memory-phenotype CD8 T cells. The generation of virus-specific, memory CD8 T cells was partially supported by IL-15Ralpha(+) DCs through the preferential enhancement of a subset of KLRG-1(+)CD27(-) CD8 T cells. In contrast, these DCs were largely sufficient in driving normal homeostatic proliferation of established memory CD8 T cells, suggesting that memory CD8 T cells grow more dependent on IL-15 transpresentation by DCs. Overall, our study clearly supports a role for DCs in memory CD8 T-cell homeostasis but also provides evidence that other hematopoietic cells are involved in this function. The identification of DCs fulfilling this role will enable future studies to better focus on mechanisms regulating T-cell homeostasis.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/fisiología , Memoria Inmunológica/fisiología , Interleucina-15/inmunología , Animales , Antígeno CD11c/genética , Antígeno CD11c/fisiología , Proliferación Celular , Células Dendríticas/metabolismo , Homeostasis/inmunología , Memoria Inmunológica/genética , Interleucina-15/genética , Subunidad alfa del Receptor de Interleucina-15/genética , Subunidad alfa del Receptor de Interleucina-15/inmunología , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas
17.
Mol Immunol ; 45(13): 3671-81, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18541300

RESUMEN

Integrin-mediated adhesion of human monocytes to fibrinogen regulated by CD11b/CD18 and the closely related integrin CD11c/CD18, play a key role in inflammation. Peripheral blood monocytes isolated from human donors despite expressing CD11c primarily utilized CD11b to mediate adhesion to fibrinogen upon stimulation with granulocyte macrophage-colony stimulating factor (GM-CSF) and fMLP. Blocking with anti-CD11b resulted in 90% (p<0.001, n=3) inhibition of monocyte adhesion. Monocytes cultured in human serum showed a shift in the participation of integrins, adhesion to fibrinogen involving both CD11b and CD11c. The participation of CD11c in cultured monocytes corresponded to a 3.4-fold increase in expression in CD11c. Blocking cultured monocytes with anti-CD11b or anti-CD11c alone showed no significant effect on adhesion. Treatment with both anti-CD11b and anti-CD11c resulted in inhibition of adhesion by 85% (p<0.001, n=3). Abrogation in adhesion upon treatment with PP1 or PP2 showed that Src family kinase activity was required for CD11b and CD11c mediated adhesion of cultured monocytes to fibrinogen upon stimulation with GM-CSF and fMLP. The clustering of CD11c on cultured monocytes upon adhesion to fibrinogen was diminished on inhibition with PP2 indicating a role for Src family kinase activity in regulating CD11c avidity. CD11b was critical to cytoskeletal events leading to increased spreading and formation of actin foci in cultured monocytes following adhesion to fibrinogen. Blocking cultured monocytes with anti-CD11b or anti-CD11c alone showed that the increase in spread area was diminished by 67+/-3% and 36+/-9%, respectively. The differential involvement of CD11c and CD11b in adhesion and subsequent cytoskeletal changes in monocytes exposed to different conditions indicates the importance of each integrin in distinct responses during inflammation.


Asunto(s)
Antígeno CD11b/fisiología , Antígeno CD11c/fisiología , Monocitos/fisiología , Familia-src Quinasas/fisiología , Antígenos de Superficie/metabolismo , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Adhesión Celular , Técnicas de Cultivo de Célula , Proliferación Celular , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/fisiología , Humanos , Modelos Biológicos , Monocitos/metabolismo , Transducción de Señal/inmunología , Familia-src Quinasas/metabolismo
18.
Trends Immunol ; 28(8): 333-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17597006

RESUMEN

Immunological memory is a critical feature of the adaptive immune system and the underlying principal behind vaccination. The mechanisms that maintain memory T cell survival between the initial and subsequent encounter with antigen remain incompletely defined. Although the cytokines IL-15 and IL-7 are important in memory T cell homeostasis, additional signals by way of TNFR family members are required for maximal maintenance of T cell memory. Here we propose a unifying model in which subsets of TNF family ligands distinguish the competitive niches for maintenance of CD4 versus CD8 T cell memory. Understanding the unique 'memory niches' defined by TNF family ligand expression will provide new insights into the mechanisms of memory T cell maintenance.


Asunto(s)
Memoria Inmunológica , Receptores del Factor de Necrosis Tumoral/fisiología , Linfocitos T/inmunología , Ligando 4-1BB/fisiología , Animales , Antígeno CD11c/fisiología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/inmunología , Citocinas/fisiología , Humanos , Antígeno Ki-1/fisiología , Receptores OX40/fisiología , Factor 1 Asociado a Receptor de TNF/fisiología
20.
Nat Immunol ; 7(7): 740-6, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16715101

RESUMEN

The function of interleukin 15 (IL-15) in unmethylated CpG oligodeoxynucleotide (CpG)-induced immune responses remains unknown. Here, in response to CpG, both wild-type and natural killer cell-depleted mice produced IL-12 and became resistant to a lethal dose of Listeria monocytogenes. In contrast, CpG-treated IL-15-deficient mice produced little IL-12 and succumbed to L. monocytogenes. CpG-stimulated conventional dendritic cells (cDCs) were the main producers of both IL-15 and IL-12, but cDCs did not produce IL-12 in the absence of plasmacytoid DCs (pDCs). The cDC-derived IL-15 induced CD40 expression by cDCs. Interaction between CD40 on cDCs and CD40 ligand on pDCs led to IL-12 production by cDCs. Thus, IL-15-dependent crosstalk between cDCs and pDCs is essential for CpG-induced immune activation.


Asunto(s)
Islas de CpG/inmunología , Células Dendríticas/fisiología , Interleucina-12/biosíntesis , Interleucina-15/fisiología , Listeriosis/inmunología , Animales , Animales Congénicos , Antígeno CD11c/genética , Antígeno CD11c/fisiología , Antígenos CD40/genética , Antígenos CD40/inmunología , Ligando de CD40/genética , Ligando de CD40/inmunología , Comunicación Celular , Células Cultivadas/inmunología , Islas de CpG/genética , Células Dendríticas/clasificación , Células Dendríticas/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Memoria Inmunológica , Interferón gamma/fisiología , Interleucina-12/genética , Interleucina-15/biosíntesis , Interleucina-15/deficiencia , Interleucina-15/genética , Listeria monocytogenes/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Quimera por Radiación , Proteínas Recombinantes de Fusión/fisiología , Subgrupos de Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA