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










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 10(8): e0134219, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26261989

RESUMEN

Asthma is a heterogeneous disease whose etiology is poorly understood but is likely to involve innate responses to inhaled microbial components that are found in allergens. The influence of these components on pulmonary inflammation has been largely studied in the context of individual agonists, despite knowledge that they can have synergistic effects when used in combination. Here we have explored the effects of LPS and ß-glucan, two commonly-encountered microbial agonists, on the pathogenesis of allergic and non-allergic respiratory responses to house dust mite allergen. Notably, sensitization with these microbial components in combination acted synergistically to promote robust neutrophilic inflammation, which involved both Dectin-1 and TLR-4. This pulmonary neutrophilic inflammation was corticosteroid-refractory, resembling that found in patients with severe asthma. Thus our results provide key new insights into how microbial components influence the development of respiratory pathology.


Asunto(s)
Asma/etiología , Lipopolisacáridos/inmunología , Neutrófilos/inmunología , beta-Glucanos/inmunología , Animales , Asma/tratamiento farmacológico , Asma/metabolismo , Asma/patología , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Ratones Noqueados , Infiltración Neutrófila , Neutrófilos/metabolismo , Neutrófilos/patología , Pyroglyphidae/inmunología , Esteroides/administración & dosificación , Esteroides/farmacología , Células Th17/inmunología , Células Th17/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
2.
Vet Immunol Immunopathol ; 163(3-4): 134-45, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25540877

RESUMEN

The lymphatic endothelium (LE) serves as a conduit for transport of immune cells and soluble antigens from peripheral tissues to draining lymph nodes (LNs), contributing to development of host immune responses and possibly dissemination of microbes. Lymphatic endothelial cells (LECs) are major constituents of the lymphatic endothelium. These specialized cells could play important roles in initiation of host innate immune responses through sensing of pathogen-associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs), including toll-like receptors (TLRs). LECs secrete pro-inflammatory cytokines and chemokines to create local inflammatory conditions for recruitment of naïve antigen presenting cells (APCs) such as dendritic cells (DCs) to sites of infection and/or vaccine administration. In this study, we examined the innate immune potential of primary LEC populations derived from multiple tissues of an animal model for human infectious diseases - the ferret. We generated a total of six primary LEC populations from lung, tracheal, and mesenteric LN tissues from three different ferrets. Standard RT-PCR characterization of these primary LECs showed that they varied in their expression of LEC markers. The ferret LECs were examined for their ability to respond to poly I:C (TLR3 and RIG-I ligand) and other known TLR ligands as measured by production of proinflammatory cytokine (IFNα, IL6, IL10, Mx1, and TNFα) and chemokine (CCL5, CCL20, and CXCL10) mRNAs using real time RT-PCR. Poly I:C exposure induced robust proinflammatory responses by all of the primary ferret LECs. Chemotaxis was performed to determine the functional activity of CCL20 produced by the primary lung LECs and showed that the LEC-derived CCL20 was abundant and functional. Taken together, our results continue to reveal the innate immune potential of primary LECs during pathogen-host interactions and expand our understanding of the roles LECs might play in health and disease in animal models.


Asunto(s)
Células Endoteliales/citología , Hurones/fisiología , Animales , Biomarcadores , Técnicas de Cultivo de Célula , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Clonación Molecular , ADN Complementario/genética , ADN Complementario/metabolismo , Células Endoteliales/fisiología , Femenino , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/fisiología , Pulmón , Filogenia , Receptores Toll-Like/metabolismo
3.
PLoS Pathog ; 10(5): e1004099, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24831696

RESUMEN

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), infects one third of the world's population. Among these infections, clinical isolates belonging to the W-Beijing appear to be emerging, representing about 50% of Mtb isolates in East Asia, and about 13% of all Mtb isolates worldwide. In animal models, infection with W-Beijing strain, Mtb HN878, is considered "hypervirulent" as it results in increased mortality and causes exacerbated immunopathology in infected animals. We had previously shown the Interleukin (IL) -17 pathway is dispensable for primary immunity against infection with the lab adapted Mtb H37Rv strain. However, it is not known whether IL-17 has any role to play in protective immunity against infection with clinical Mtb isolates. We report here that lab adapted Mtb strains, such as H37Rv, or less virulent Mtb clinical isolates, such as Mtb CDC1551, do not require IL-17 for protective immunity against infection while infection with Mtb HN878 requires IL-17 for early protective immunity. Unexpectedly, Mtb HN878 induces robust production of IL-1ß through a TLR-2-dependent mechanism, which supports potent IL-17 responses. We also show that the role for IL-17 in mediating protective immunity against Mtb HN878 is through IL-17 Receptor signaling in non-hematopoietic cells, mediating the induction of the chemokine, CXCL-13, which is required for localization of T cells within lung lymphoid follicles. Correct T cell localization within lymphoid follicles in the lung is required for maximal macrophage activation and Mtb control. Since IL-17 has a critical role in vaccine-induced immunity against TB, our results have far reaching implications for the design of vaccines and therapies to prevent and treat emerging Mtb strains. In addition, our data changes the existing paradigm that IL-17 is dispensable for primary immunity against Mtb infection, and instead suggests a differential role for IL-17 in early protective immunity against emerging Mtb strains.


Asunto(s)
Inmunidad Innata/genética , Interleucina-17/fisiología , Mycobacterium tuberculosis/inmunología , Animales , Células Cultivadas , Enfermedades Transmisibles Emergentes/genética , Enfermedades Transmisibles Emergentes/inmunología , Citoprotección/genética , Citoprotección/inmunología , Femenino , Interleucina-17/genética , Interleucina-1beta/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/patogenicidad , Receptores Tipo I de Interleucina-1/genética , Receptor Toll-Like 2/fisiología , Tuberculosis/genética , Tuberculosis/inmunología
4.
Am J Pathol ; 184(1): 55-63, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24183780

RESUMEN

Mucosal vaccines are thought to confer superior protection against mucosal infectious diseases. In addition, mucosal routes of vaccine delivery preferentially induce the generation of T helper 17 (Th17) cells, which produce the cytokine IL-17. Th17 cells are critical in mediating vaccine-induced immunity against several mucosal infectious diseases. However, IL-17 is also a potent proinflammatory cytokine, and we recently showed that IL-17 mediates immunopathology and lung injury after influenza infection in mice. In the present study, we tested the hypothesis that mucosal pre-exposure to Th17-inducing adjuvants can promote disease exacerbation upon subsequent infection with influenza virus. Mice mucosally pre-exposed to Th17-inducing adjuvants, such as type II heat-labile enterotoxin or cholera toxin, resulted in increased morbidity and exacerbated lung inflammation upon subsequent infection with influenza virus. Furthermore, the increased morbidity was accompanied by increased expression of inflammatory chemokines and increased accumulation of neutrophils. Importantly, blockade of the IL-17 pathway in mice pre-exposed to Th17-inducing adjuvants resulted in attenuation of the inflammatory phenotype seen in influenza-infected mice. Our findings indicate that, before mucosal Th17-inducing adjuvants can be used in vaccine strategies, the short- and long-term detrimental effects of such adjuvants on disease exacerbation and lung injury in response to infections, such as influenza, should be carefully studied.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Infecciones por Orthomyxoviridae/inmunología , Células Th17/inmunología , Animales , Femenino , Citometría de Flujo , Inmunohistoquímica , Hibridación in Situ , Virus de la Influenza A , Vacunas contra la Influenza/inmunología , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/inmunología , Infecciones por Orthomyxoviridae/patología , Reacción en Cadena de la Polimerasa
5.
Am J Respir Crit Care Med ; 188(9): 1137-46, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24047412

RESUMEN

RATIONALE: A hallmark of pulmonary tuberculosis (TB) is the formation of granulomas. However, the immune factors that drive the formation of a protective granuloma during latent TB, and the factors that drive the formation of inflammatory granulomas during active TB, are not well defined. OBJECTIVES: The objective of this study was to identify the underlying immune mechanisms involved in formation of inflammatory granulomas seen during active TB. METHODS: The immune mediators involved in inflammatory granuloma formation during TB were assessed using human samples and experimental models of Mycobacterium tuberculosis infection, using molecular and immunologic techniques. MEASUREMENTS AND MAIN RESULTS: We demonstrate that in human patients with active TB and in nonhuman primate models of M. tuberculosis infection, neutrophils producing S100 proteins are dominant within the inflammatory lung granulomas seen during active TB. Using the mouse model of TB, we demonstrate that the exacerbated lung inflammation seen as a result of neutrophilic accumulation is dependent on S100A8/A9 proteins. S100A8/A9 proteins promote neutrophil accumulation by inducing production of proinflammatory chemokines and cytokines, and influencing leukocyte trafficking. Importantly, serum levels of S100A8/A9 proteins along with neutrophil-associated chemokines, such as keratinocyte chemoattractant, can be used as potential surrogate biomarkers to assess lung inflammation and disease severity in human TB. CONCLUSIONS: Our results thus show a major pathologic role for S100A8/A9 proteins in mediating neutrophil accumulation and inflammation associated with TB. Thus, targeting specific molecules, such as S100A8/A9 proteins, has the potential to decrease lung tissue damage without impacting protective immunity against TB.


Asunto(s)
Calgranulina A/inmunología , Calgranulina B/inmunología , Granuloma del Sistema Respiratorio/inmunología , Mediadores de Inflamación/inmunología , Neutrófilos/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Quimiocinas/inmunología , Factores Quimiotácticos/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL
6.
Lymphat Res Biol ; 11(1): 26-34, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23531182

RESUMEN

Abstract LYVE-1 is a marker expressed by lymphatic endothelial cells (LECs) that line the lymphatic endothelium. Through studies designed to examine potential changes in expression of LYVE-1 in cynomolgus macaque colon tissues during the course of simian immunodeficiency virus (SIV) infection, we discovered that LYVE-1 was expressed by heterogenous populations of cells. As revealed by in situ hybridization (ISH), LYVE-1 mRNA levels in colon were decreased in macaques with AIDS compared with acutely infected or uninfected macaques. In the submucosal layer of the colon, approximately half of the LYVE-1-expressing cells co-expressed the dendritic cell (DC) marker, DC-SIGN/CD209, and this percentage did not change appreciably during infection. Subsets of cells expressing LYVE-1 also co-expressed macrophage markers, such as CD68 and the macrophage mannose receptor (MMR)/CD206, in both the colon and lymph nodes. LECs, DCs, and macrophages that co-expressed LYVE-1 were observed in colon and lymph node from uninfected, healthy animals as well as in tissues with SIV-driven inflammation. These findings provide further definition of the phenotypic overlap between LECs and antigen presenting cells, reveal the heterogeneity within the population of cells expressing the lymphatic marker LYVE-1, and show that SIV modulates this population of cells in a mucosal surface across which the virus is acquired.


Asunto(s)
Moléculas de Adhesión Celular/inmunología , Intestino Grueso/inmunología , Lectinas Tipo C/inmunología , Receptores de Superficie Celular/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Proteínas de Transporte Vesicular/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/inmunología , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Colon/inmunología , Colon/metabolismo , Colon/virología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Endotelio Linfático/citología , Endotelio Linfático/inmunología , Endotelio Linfático/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Hibridación in Situ , Intestino Grueso/metabolismo , Intestino Grueso/virología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/virología , Macaca fascicularis , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Receptor de Manosa , Lectinas de Unión a Manosa/inmunología , Lectinas de Unión a Manosa/metabolismo , Microscopía Confocal , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
7.
Cytokine ; 61(3): 924-32, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23360828

RESUMEN

CCL20 is currently the only known chemokine ligand for the receptor CCR6, and is a mucosal chemokine involved in normal and pathological immune responses. Although nucleotide sequence data are available for ccl20 and ccr6 sequences from multiple species, the ferret ccl20 and ccr6 sequences have not been determined. To increase our understanding of immune function in ferret models of infection and vaccination, we have used RT-PCR to obtain the ferret ccl20 and ccr6 cDNA sequences and functionally characterize the encoded proteins. The open reading frames of both genes were highly conserved across species and mostly closely related to canine sequences. For functional analyses, single cell clones expressing ferret CCR6 were generated, a ferret CCL20/mouse IgG(2a) fusion protein (fCCL20-mIgG(2a)) was produced, and fCCL20 was chemically synthesized. Cell clones expressing ferret CCR6 responded chemotactically to fCCL20-mIgG2a fusion protein and synthetic ferret CCL20. Chemotaxis inhibition studies identified the polyphenol epigallocatechin-3-gallate and the murine γ-herpesvirus 68 M3 protein as inhibitors of fCCL20. Surface plasmon resonance studies revealed that EGCG bound directly to fCCL20. These results provide molecular characterization of previously unreported ferret immune gene sequences and for the first time identify a broad-spectrum small molecule inhibitor of CCL20 and reveal CCL20 as a target for the herpesviral M3 protein.


Asunto(s)
Quimiocina CCL20/metabolismo , Quimiotaxis , Hurones/metabolismo , Receptores CCR6/metabolismo , Secuencia de Aminoácidos , Animales , Catequina/análogos & derivados , Catequina/farmacología , Quimiocina CCL20/química , Quimiotaxis/efectos de los fármacos , Clonación Molecular , ADN Complementario/genética , Perros , Ratones , Datos de Secuencia Molecular , Sistemas de Lectura Abierta/genética , Filogenia , Unión Proteica/efectos de los fármacos , Receptores CCR6/química , Alineación de Secuencia , Análisis de Secuencia de ADN , Proteínas Virales/farmacología
8.
J Clin Invest ; 123(2): 712-26, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23281399

RESUMEN

One third of the world's population is infected with Mycobacterium tuberculosis (Mtb). Although most infected people remain asymptomatic, they have a 10% lifetime risk of developing active tuberculosis (TB). Thus, the current challenge is to identify immune parameters that distinguish individuals with latent TB from those with active TB. Using human and experimental models of Mtb infection, we demonstrated that organized ectopic lymphoid structures containing CXCR5+ T cells were present in Mtb-infected lungs. In addition, we found that in experimental Mtb infection models, the presence of CXCR5+ T cells within ectopic lymphoid structures was associated with immune control. Furthermore, in a mouse model of Mtb infection, we showed that activated CD4+CXCR5+ T cells accumulated in Mtb-infected lungs and produced proinflammatory cytokines. Mice deficient in Cxcr5 had increased susceptibility to TB due to defective T cell localization within the lung parenchyma. We demonstrated that CXCR5 expression in T cells mediated correct T cell localization within TB granulomas, promoted efficient macrophage activation, protected against Mtb infection, and facilitated lymphoid follicle formation. These data demonstrate that CD4+CXCR5+ T cells play a protective role in the immune response against TB and highlight their potential use for future TB vaccine design and therapy.


Asunto(s)
Tuberculosis Latente/inmunología , Receptores CXCR5/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Granuloma del Sistema Respiratorio/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Activación de Linfocitos , Tejido Linfoide/inmunología , Activación de Macrófagos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Subgrupos de Linfocitos T/inmunología
9.
Proc Natl Acad Sci U S A ; 108(13): 5360-5, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21402950

RESUMEN

Aspergillus fumigatus is commonly associated with allergic bronchopulmonary aspergillosis in patients with severe asthma in which chronic airway neutrophilia predicts a poor outcome. We were able to recapitulate fungus-induced neutrophilic airway inflammation in a mouse model in our efforts to understand the underlying mechanisms. However, neutrophilia occurred in a mouse strain-selective fashion, providing us with an opportunity to perform a comparative study to elucidate the mechanisms involved. Here we show that TNF-α, largely produced by Ly6c(+)CD11b(+) dendritic cells (DCs), plays a central role in promoting IL-17A from CD4(+) T cells and collaborating with it to induce airway neutrophilia. Compared with C57BL/6 mice, BALB/c mice displayed significantly more TNF-α-producing DCs and macrophages in the lung. Lung TNF-α levels were drastically reduced in CD11c-DTR BALB/c mice depleted of CD11c+ cells, and TNF-α-producing Ly6c(+)CD11b(+) cells were abolished in Dectin-1(-/-) and MyD88(-/-) BALB/c mice. TNF-α deficiency itself blunted accumulation of inflammatory Ly6c(+)CD11b(+) DCs. Also, lack of TNF-α decreased IL-17A but promoted IL-5 levels, switching inflammation from a neutrophil to eosinophil bias resembling that in C57BL/6 mice. The TNF-α(low) DCs in C57BL/6 mice contained more NF-κB p50 homodimers, which are strong repressors of TNF-α transcription. Functionally, collaboration between TNF-α and IL-17A triggered significantly higher levels of the neutrophil chemoattractants keratinocyte cytokine and macrophage inflammatory protein 2 in BALB/c mice. Our study identifies TNF-α as a molecular switch that orchestrates a sequence of events in DCs and CD4 T cells that promote neutrophilic airway inflammation.


Asunto(s)
Células Dendríticas/inmunología , Eosinofilia/inmunología , Interleucina-17/inmunología , Interleucina-5/inmunología , Pulmón/inmunología , Neutrófilos/inmunología , Aspergilosis Pulmonar/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Antígenos CD/inmunología , Linfocitos T CD4-Positivos/inmunología , Humanos , Pulmón/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptor Toll-Like 2/inmunología
10.
Am J Pathol ; 177(3): 1274-85, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20671263

RESUMEN

Infection by HIV-1 frequently leads to pulmonary complications, including alterations to local immune environments. To better understand these alterations, we have examined in detail the patterns and levels of expression of chemokine, cytokine, and chemokine receptor mRNAs in lung tissues from 16 uninfected or simian immunodeficiency virus (SIV)/DeltaB670 infected cynomolgus macaques at different stages of infection. Among the most up-regulated immune genes were interferon (IFN)-gamma, IFN-gamma-inducible CXCR3 ligands, and CCR5 ligands, as well as the cognate chemokine receptors. These changes were greatest in animals with clear Pneumocystis carinii coinfection. Immunohistochemistry and in situ hybridization revealed monocytes/macrophages to be the predominant type of cell infiltrating into lung tissues and serving as the major cellular source of chemokines. To explore the causes of chemokine alterations, we treated macaque lung cells with IFN-gamma, lipopolysaccharide, Poly(I:C), and P. carinii in vitro, and results revealed that these stimuli can induce the expression of CXCR3 ligand and/or CCR5 ligand mRNAs. Taken together, these studies provide a comprehensive definition of the chemokine networks available to modulate cellular recruitment to lung tissues during SIV infection and implicate both cytokines (IFN-gamma) and pathogens (SIV and P. carinii) as contributors to increased expression of pro-inflammatory chemokines.


Asunto(s)
Quimiocinas/inmunología , Pulmón/inmunología , Pneumocystis carinii/inmunología , Neumonía por Pneumocystis/inmunología , Receptores de Quimiocina/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Quimiocinas/metabolismo , Inmunohistoquímica , Hibridación in Situ , Pulmón/metabolismo , Pulmón/virología , Macaca fascicularis , Neumonía por Pneumocystis/complicaciones , Neumonía por Pneumocystis/metabolismo , Neumonía por Pneumocystis/virología , Receptores de Quimiocina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Estadísticas no Paramétricas
11.
Cancer Immunol Immunother ; 59(9): 1401-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20549206

RESUMEN

Stimulation of double-stranded (ds)RNA receptors can increase the effectiveness of cancer vaccines, but the underlying mechanisms are not completely elucidated. In this study, we sought to determine critical roles of host IFN-alpha and IFN-gamma pathways in the enhanced therapeutic efficacy mediated by peptide vaccines and polyinosinic-polycytidylic acid [poly(I:C)] stabilized by lysine and carboxymethylcellulose (poly-ICLC) in the murine central nervous system (CNS) GL261 glioma. C57BL/6-background wild type (WT), IFN-alpha receptor-1 (IFN-alphaR1)(-/-) or IFN-gamma(-/-) mice bearing syngeneic CNS GL261 glioma received subcutaneous (s.c.) vaccinations with synthetic peptides encoding CTL epitopes with or without intramuscular (i.m.) injections of poly-ICLC. The combinational treatment induced a robust transcription of CXCL10 in the glioma site. Blockade of CXCL10 with a specific monoclonal antibody (mAb) abrogated the efficient CNS homing of antigen-specific type-1 CTL (Tc1). Both IFN-alphaR(-/-) and IFN-gamma(-/-) hosts failed to up-regulate the CXCL10 mRNA and recruit Tc1 cells to the tumor site, indicating non-redundant roles of type-1 and type-2 IFNs in the effects of poly-ICLC-assisted vaccines. The efficient trafficking of Tc1 also required Tc1-derived IFN-gamma. Our data point to critical roles of the host-IFN-alpha and IFN-gamma pathways in the modulation of CNS glioma microenvironment, and the therapeutic effectiveness of poly-ICLC-assisted glioma vaccines.


Asunto(s)
Vacunas contra el Cáncer , Carboximetilcelulosa de Sodio/análogos & derivados , Neoplasias del Sistema Nervioso Central/terapia , Glioma/terapia , Poli I-C/administración & dosificación , Polilisina/análogos & derivados , Linfocitos T Citotóxicos/efectos de los fármacos , Animales , Anticuerpos Bloqueadores/administración & dosificación , Carboximetilcelulosa de Sodio/administración & dosificación , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Movimiento Celular/inmunología , Neoplasias del Sistema Nervioso Central/inmunología , Neoplasias del Sistema Nervioso Central/patología , Quimiocina CXCL10/biosíntesis , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Epítopos de Linfocito T/química , Glioma/inmunología , Glioma/patología , Inmunización , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Polilisina/administración & dosificación , Receptor de Interferón alfa y beta/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología
12.
J Immunol ; 180(5): 3399-405, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18292566

RESUMEN

The lymphatic endothelium is the preferred route for the drainage of interstitial fluid from tissues and also serves as a conduit for peripheral dendritic cells (DCs) to reach draining lymph nodes. Lymphatic endothelial cells (LECs) are known to produce chemokines that recruit Ag-loaded DCs to lymphatic vessels and therefore are likely to regulate the migration of DCs to lymph nodes. TLRs are immune receptors that recognize pathogen associated molecular patterns and then signal and stimulate production of inflammatory chemokines and cytokines that contribute to innate and adaptive immune responses. TLRs are known to be expressed by a wide variety of cell types including leukocytes, epithelial cells, and endothelial cells. Because the TLR expression profile of LECs remains largely unexamined, we have undertaken a comprehensive study of the expression of TLR1-10 mRNAs and protein in primary human dermal (HD) and lung LECs as well as in htert-HDLECs, which display a longer life-span than HDLECs. We found that all three cell types expressed TLR1-6 and TLR9. The responsiveness of these LECs to a panel of ligands for TLR1-9 was measured by real-time RT-PCR, ELISA, and flow cytometry, and revealed that the LECs responded to most but not all TLR ligands by increasing expression of inflammatory chemokines, cytokines, and adhesion molecules. These findings provide insight into the ability of cells of the lymphatic vasculature to respond to pathogens and potential vaccine adjuvants and shape peripheral environments in which DCs will acquire Ag and environmental cues.


Asunto(s)
Endotelio Linfático/inmunología , Endotelio Linfático/metabolismo , Receptores Toll-Like/biosíntesis , Receptores Toll-Like/fisiología , Animales , Moléculas de Adhesión Celular/biosíntesis , Línea Celular , Células Cultivadas , Quimiocina CCL21/biosíntesis , Quimiocina CCL21/genética , Quimiocinas/biosíntesis , Quimiocinas/fisiología , Citocinas/biosíntesis , Citocinas/fisiología , Relación Dosis-Respuesta Inmunológica , Endotelio Linfático/citología , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Ligandos , Pulmón/citología , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Microcirculación/citología , Microcirculación/inmunología , Microcirculación/metabolismo , Piel/citología , Piel/inmunología , Piel/metabolismo , Receptores Toll-Like/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...