Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Immunol ; 17(5): 538-44, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27043413

RESUMEN

Acidic mammalian chitinase (AMCase) is known to be induced by allergens and helminths, yet its role in immunity is unclear. Using AMCase-deficient mice, we show that AMCase deficiency reduced the number of group 2 innate lymphoid cells during allergen challenge but was not required for establishment of type 2 inflammation in the lung in response to allergens or helminths. In contrast, AMCase-deficient mice showed a profound defect in type 2 immunity following infection with the chitin-containing gastrointestinal nematodes Nippostrongylus brasiliensis and Heligmosomoides polygyrus bakeri. The impaired immunity was associated with reduced mucus production and decreased intestinal expression of the signature type 2 response genes Il13, Chil3, Retnlb, and Clca1. CD103(+) dendritic cells, which regulate T cell homing, were also reduced in mesenteric lymph nodes of infected AMCase-deficient mice. Thus, AMCase functions as a critical initiator of protective type 2 responses to intestinal nematodes but is largely dispensable for allergic responses in the lung.


Asunto(s)
Quitinasas/inmunología , Tracto Gastrointestinal/inmunología , Inmunidad/inmunología , Infecciones por Strongylida/inmunología , Animales , Quitinasas/genética , Quitinasas/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/inmunología , Canales de Cloruro/metabolismo , Citometría de Flujo , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/parasitología , Expresión Génica/inmunología , Hormonas Ectópicas/genética , Hormonas Ectópicas/inmunología , Hormonas Ectópicas/metabolismo , Interacciones Huésped-Parásitos/inmunología , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Inmunidad/genética , Péptidos y Proteínas de Señalización Intercelular , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-13/metabolismo , Lectinas/genética , Lectinas/inmunología , Lectinas/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Nematospiroides dubius/inmunología , Nematospiroides dubius/fisiología , Nippostrongylus/inmunología , Nippostrongylus/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/inmunología , beta-N-Acetilhexosaminidasas/metabolismo
2.
Immunity ; 45(1): 145-58, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27421703

RESUMEN

Fibroproliferative diseases are driven by dysregulated tissue repair responses and are a major cause of morbidity and mortality because they affect nearly every organ system. Type 2 cytokine responses are critically involved in tissue repair; however, the mechanisms that regulate beneficial regeneration versus pathological fibrosis are not well understood. Here, we have shown that the type 2 effector cytokine interleukin-13 simultaneously, yet independently, directed hepatic fibrosis and the compensatory proliferation of hepatocytes and biliary cells in progressive models of liver disease induced by interleukin-13 overexpression or after infection with Schistosoma mansoni. Using transgenic mice with interleukin-13 signaling genetically disrupted in hepatocytes, cholangiocytes, or resident tissue fibroblasts, we have revealed direct and distinct roles for interleukin-13 in fibrosis, steatosis, cholestasis, and ductular reaction. Together, these studies show that these mechanisms are simultaneously controlled but distinctly regulated by interleukin-13 signaling. Thus, it may be possible to promote interleukin-13-dependent hepatobiliary expansion without generating pathological fibrosis. VIDEO ABSTRACT.


Asunto(s)
Hígado Graso/inmunología , Fibroblastos/inmunología , Interleucina-13/metabolismo , Cirrosis Hepática Biliar/inmunología , Hígado/patología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Animales , Ácidos y Sales Biliares/biosíntesis , Proliferación Celular , Células Cultivadas , Fibrosis , Humanos , Interleucina-13/genética , Interleucina-13/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Células Th2/inmunología
3.
J Pathol ; 248(1): 16-29, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30536905

RESUMEN

Fibroproliferative diseases affect a significant proportion of the world's population. Despite this, core mechanisms driving organ fibrosis of diverse etiologies remain ill defined. Recent studies suggest that integrin-alpha V serves as a master driver of fibrosis in multiple organs. Although diverse mechanisms contribute to the progression of fibrosis, TGF-ß and IL-13 have emerged as central mediators of fibrosis during type 1/type 17, and type 2 polarized inflammatory responses, respectively. To investigate if integrin-alpha V interactions or signaling is critical to the development of type 2 fibrosis, we analyzed fibroblast-specific integrin-alpha V knockout mice in three type 2-driven inflammatory disease models. While we confirmed a role for integrin-alpha V in type 17-associated fibrosis, integrin-alpha V was not critical to the development of type 2-driven fibrosis. Additionally, our studies support a novel mechanism through which fibroblasts, via integrin-alpha V expression, are capable of regulating immune polarization. We show that when integrin-alpha V is deleted on fibroblasts, initiation of type 17 inflammation is inhibited leading to a deregulation of type 2 inflammation. This mechanism is most evident in a model of severe asthma, which is characterized by a mixed type 2/type 17 inflammatory response. Together, these findings suggest dual targeting of integrin-alpha V and type 2 pathways may be needed to ameliorate fibrosis and prevent rebound of opposing pro-fibrotic and inflammatory mechanisms. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Fibroblastos/metabolismo , Inflamación/metabolismo , Integrina alfa5/fisiología , Animales , Asma/metabolismo , Asma/prevención & control , Modelos Animales de Enfermedad , Femenino , Fibrosis , Eliminación de Gen , Inflamación/patología , Integrina alfa5/genética , Interleucina-13/antagonistas & inhibidores , Interleucina-13/inmunología , Cirrosis Hepática/inmunología , Cirrosis Hepática/patología , Cirrosis Hepática/prevención & control , Masculino , Ratones Noqueados , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control
4.
PLoS Pathog ; 10(9): e1004372, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25211233

RESUMEN

Mice expressing a Cre recombinase from the lysozyme M-encoding locus (Lyz2) have been widely used to dissect gene function in macrophages and neutrophils. Here, we show that while naïve resident tissue macrophages from IL-4Rαf(lox/delta)LysM(Cre) mice almost completely lose IL-4Rα function, a large fraction of macrophages elicited by sterile inflammatory stimuli, Schistosoma mansoni eggs, or S. mansoni infection, fail to excise Il4rα. These F4/80(hi)CD11b(hi) macrophages, in contrast to resident tissue macrophages, express lower levels of Lyz2 explaining why this population resists LysM(Cre)-mediated deletion. We show that in response to IL-4 and IL-13, Lyz2(lo)IL-4Rα(+) macrophages differentiate into an arginase 1-expressing alternatively-activated macrophage (AAM) population, which slows the development of lethal fibrosis in schistosomiasis. In contrast, we identified Lyz2(hi)IL-4Rα(+) macrophages as the key subset of AAMs mediating the downmodulation of granulomatous inflammation in chronic schistosomiasis. Our observations reveal a limitation on using a LysMCre mouse model to study gene function in inflammatory settings, but we utilize this limitation as a means to demonstrate that distinct populations of alternatively activated macrophages control inflammation and fibrosis in chronic schistosomiasis.


Asunto(s)
Fibrosis/inmunología , Inflamación/inmunología , Macrófagos Peritoneales/inmunología , Receptores de Superficie Celular/fisiología , Schistosoma mansoni/patogenicidad , Esquistosomiasis/inmunología , Animales , Células Cultivadas , Enfermedad Crónica , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrosis/parasitología , Fibrosis/patología , Inflamación/parasitología , Inflamación/patología , Integrasas/metabolismo , Macrófagos Peritoneales/parasitología , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Neutrófilos/parasitología , Neutrófilos/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esquistosomiasis/parasitología , Esquistosomiasis/patología
5.
Immunol Cell Biol ; 92(6): 499-508, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24613975

RESUMEN

Expression of the chemokine receptor CX3CR1 has been used to identify distinct populations within the monocyte, macrophage and dendritic cell lineages. Recent evidence indicates that CX3CR1-positive subsets of myeloid cells play distinct and important roles in a wide range of immunological maladies, and thus the use of CX3CR1 expression has leveraged our understanding of the myeloid contribution to a multitude of diseases. Here we use CX3CR1 expression as a means to identify a novel nongranulocytic CX3CR1-negative myeloid population that is functionally distinct from the previously described CX3CR1-positive cellular subsets within the CD11b-positive cellular compartment of ascites from ovarian tumor-bearing mice. We functionally identify CX3CR1-negative cells as myeloid suppressor cells and as a cellular subset with pathological specificity. Importantly, the CX3CR1-negative cells exhibit early IL-10 production in the ovarian tumor microenvironment, which we have shown to be critically tied to suppression and additional myeloid-derived suppressor cell accumulation, and we now show that this cellular population actively contributes to tumor progression. Furthermore, we demonstrate that the CX3CR1-negative population is derived from the recently described CX3CR1-positive macrophage/dendritic cell precursor cell. These studies provide a greater understanding of the generation and maintenance of regulatory myeloid subsets and have broad implications for the elucidation of myeloid function and contributions within the tumor microenvironment.


Asunto(s)
Células Mieloides/inmunología , Neoplasias Experimentales/inmunología , Neoplasias Ováricas/inmunología , Receptores de Quimiocina/inmunología , Microambiente Tumoral/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Femenino , Interleucina-10/genética , Interleucina-10/inmunología , Ratones , Ratones Transgénicos , Células Mieloides/patología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Receptores de Quimiocina/genética , Microambiente Tumoral/genética
6.
Sci Immunol ; 8(82): eadd8945, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37027478

RESUMEN

Macrophages are central orchestrators of the tissue response to injury, with distinct macrophage activation states playing key roles in fibrosis progression and resolution. Identifying key macrophage populations found in human fibrotic tissues could lead to new treatments for fibrosis. Here, we used human liver and lung single-cell RNA sequencing datasets to identify a subset of CD9+TREM2+ macrophages that express SPP1, GPNMB, FABP5, and CD63. In both human and murine hepatic and pulmonary fibrosis, these macrophages were enriched at the outside edges of scarring and adjacent to activated mesenchymal cells. Neutrophils expressing MMP9, which participates in the activation of TGF-ß1, and the type 3 cytokines GM-CSF and IL-17A coclustered with these macrophages. In vitro, GM-CSF, IL-17A, and TGF-ß1 drive the differentiation of human monocytes into macrophages expressing scar-associated markers. Such differentiated cells could degrade collagen IV but not collagen I and promote TGF-ß1-induced collagen I deposition by activated mesenchymal cells. In murine models blocking GM-CSF, IL-17A or TGF-ß1 reduced scar-associated macrophage expansion and hepatic or pulmonary fibrosis. Our work identifies a highly specific macrophage population to which we assign a profibrotic role across species and tissues. It further provides a strategy for unbiased discovery, triage, and preclinical validation of therapeutic targets based on this fibrogenic macrophage population.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Fibrosis Pulmonar , Humanos , Ratones , Animales , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Interleucina-17/metabolismo , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Cicatriz , Macrófagos/patología , Inflamación/patología , Proteínas de Unión a Ácidos Grasos/metabolismo , Glicoproteínas de Membrana , Receptores Inmunológicos
7.
Sci Transl Med ; 9(396)2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28659437

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is now the most common progressive liver disease in developed countries and is the second leading indication for liver transplantation due to the extensive fibrosis it causes. NAFLD progression is thought to be tied to chronic low-level type 1 inflammation originating in the adipose tissue during obesity; however, the specific immunological mechanisms regulating the progression of NAFLD-associated fibrosis in the liver are unclear. To investigate the immunopathogenesis of NAFLD more completely, we investigated adipose dysfunction, nonalcoholic steatohepatitis (NASH), and fibrosis in mice that develop polarized type 1 or type 2 immune responses. Unexpectedly, obese interleukin-10 (IL-10)/IL-4-deficient mice (type 1-polarized) were highly resistant to NASH. This protection was associated with an increased hepatic interferon-γ (IFN-γ) signature. Conversely, IFN-γ-deficient mice progressed rapidly to NASH with evidence of fibrosis dependent on transforming growth factor-ß (TGF-ß) and IL-13 signaling. Unlike increasing type 1 inflammation and the marked loss of eosinophils seen in expanding adipose tissue, progression of NASH was associated with increasing eosinophilic type 2 liver inflammation in mice and human patient biopsies. Finally, simultaneous inhibition of TGF-ß and IL-13 signaling attenuated the fibrotic machinery more completely than TGF-ß alone in NAFLD-associated fibrosis. Thus, although type 2 immunity maintains healthy metabolic signaling in adipose tissues, it exacerbates the progression of NAFLD collaboratively with TGF-ß in the liver.


Asunto(s)
Progresión de la Enfermedad , Inmunidad , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/prevención & control , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Factor de Crecimiento Transformador beta/metabolismo , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa , Eosinófilos/patología , Humanos , Inflamación/patología , Interferón gamma/deficiencia , Interferón gamma/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/patología
8.
Sci Transl Med ; 8(337): 337ra65, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27147589

RESUMEN

Thymic stromal lymphopoietin (TSLP), interleukin-25 (IL-25), and IL-33 are important initiators of type 2-associated mucosal inflammation and immunity. However, their role in the maintenance of progressive type 2 inflammation and fibrosis is much less clear. Using chronic models of helminth infection and allergic lung inflammation, we show that collective disruption of TSLP, IL-25, and IL-33 signaling suppresses chronic and progressive type 2 cytokine-driven inflammation and fibrosis. In a schistosome lung granuloma model or during chronic Schistosoma mansoni infection in the liver, individual ablation of TSLP, IL-25, or IL-33/ST2 had no impact on the development of IL-4/IL-13-dependent inflammation or fibrosis. However, significant reductions in granuloma-associated eosinophils, hepatic fibrosis, and IL-13-producing type 2 innate lymphoid cells (ILC2s) were observed when signaling of all three mediators was simultaneously disrupted. Combined blockade through monoclonal antibody (mAb) treatment also reduced IL-5 and IL-13 expression during primary and secondary granuloma formation in the lungs. In a model of chronic house dust mite-induced allergic lung inflammation, combined mAb treatment did not decrease established inflammation or fibrosis. TSLP/IL-33 double-knockout mice treated with anti-IL-25 mAb during priming, however, displayed decreased inflammation, mucus production, and lung remodeling in the chronic phase. Together, these studies reveal partially redundant roles for TSLP, IL-25, and IL-33 in the maintenance of type 2 pathology and suggest that in some settings, early combined targeting of these mediators is necessary to ameliorate progressive type 2-driven disease.


Asunto(s)
Citocinas/metabolismo , Fibrosis/inmunología , Inflamación/inmunología , Inflamación/terapia , Interleucina-17/metabolismo , Interleucina-33/metabolismo , Neoplasias Pulmonares/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Citocinas/antagonistas & inhibidores , Citocinas/genética , Femenino , Fibrosis/tratamiento farmacológico , Fibrosis/terapia , Granuloma/tratamiento farmacológico , Granuloma/inmunología , Granuloma/parasitología , Granuloma/terapia , Inflamación/tratamiento farmacológico , Interleucina-13/antagonistas & inhibidores , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-17/antagonistas & inhibidores , Interleucina-17/genética , Interleucina-33/antagonistas & inhibidores , Interleucina-33/genética , Interleucina-4/antagonistas & inhibidores , Interleucina-4/genética , Interleucina-4/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/parasitología , Neoplasias Pulmonares/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Schistosoma mansoni/inmunología , Schistosoma mansoni/patogenicidad , Linfopoyetina del Estroma Tímico
9.
Sci Transl Med ; 7(301): 301ra129, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26290411

RESUMEN

Increasing evidence suggests that asthma is a heterogeneous disorder regulated by distinct molecular mechanisms. In a cross-sectional study of asthmatics of varying severity (n = 51), endobronchial tissue gene expression analysis revealed three major patient clusters: TH2-high, TH17-high, and TH2/17-low. TH2-high and TH17-high patterns were mutually exclusive in individual patient samples, and their gene signatures were inversely correlated and differentially regulated by interleukin-13 (IL-13) and IL-17A. To understand this dichotomous pattern of T helper 2 (TH2) and TH17 signatures, we investigated the potential of type 2 cytokine suppression in promoting TH17 responses in a preclinical model of allergen-induced asthma. Neutralization of IL-4 and/or IL-13 resulted in increased TH17 cells and neutrophilic inflammation in the lung. However, neutralization of IL-13 and IL-17 protected mice from eosinophilia, mucus hyperplasia, and airway hyperreactivity and abolished the neutrophilic inflammation, suggesting that combination therapies targeting both pathways may maximize therapeutic efficacy across a patient population comprising both TH2 and TH17 endotypes.


Asunto(s)
Asma/inmunología , Asma/metabolismo , Células Th17/metabolismo , Células Th2/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Interleucina-13/metabolismo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos BALB C , Transducción de Señal
11.
Front Immunol ; 2: 29, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22566819

RESUMEN

Elevated levels of IL-10 in the microenvironment of human ovarian cancer and murine models of ovarian cancer are well established and correlate with poor clinical prognosis. However, amongst a myriad of immunosuppressive factors, the actual contribution of IL-10 to the ovarian tumor microenvironment, the mechanisms by which it acts, and its possible functional redundancy are unknown. We previously demonstrated that elimination of the myeloid-derived suppressor cell (MDSC) compartment within the ovarian tumor ascites inhibited tumor progression and, intriguingly, significantly decreased local IL-10 levels. Here we identify a novel pathway in which the tumor-infiltrating MDSC are the predominant producers of IL-10 and, importantly, require it to develop their immunosuppressive function in vivo. Importantly, we demonstrate that the role of IL-10 is critical, and not redundant with other immunosuppressive molecules, to in vivo tumor progression: blockade of the IL-10 signaling network results in alleviation of MDSC-mediated immunosuppression, altered T cell phenotype and activity, and improved survival. These studies define IL-10 as a fundamental modulator of both MDSC and T cells within the ovarian tumor microenvironment. Importantly, IL-10 signaling is shown to be necessary to the development and maintenance of a permissive tumor microenvironment and represents a viable target for anti-tumor strategies.

12.
Neoplasia ; 11(6): 564-73, 1 p following 573, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19484145

RESUMEN

Ovarian tumor progression is marked by the peritoneal accumulation of leukocytes. Among these leukocytes, an immunosuppressive CD11b(+)CD11c(+) population has been identified in both human and ovarian tumors. The use of transplantable models of murine ovarian tumors has demonstrated that this population promotes ovarian tumor growth, whereas elimination of this population has been shown to inhibit ovarian tumor progression. Despite the demonstrated importance of these cells to ovarian tumor progression, the mechanisms by which these cells are recruited to the peritoneal tumor are largely unknown. Therefore, this study analyzes the mechanisms these cells use to migrate to the peritoneum with the goal of therapeutically blocking their recruitment and subsequent immunosuppressive activity. Recent studies have identified that CX(3)CR1, Gr-1, and CCR2 delineate phenotypic and functional murine monocyte subsets. Here, we report that CX(3)CR1(lo)Gr-1(hi) cells dominate the population of peritoneal CD11b(+) leukocytes early in murine tumor development; however, the CX(3)CR1(hi) population of cells present in the peritoneum dramatically increases in both total numbers and percentage during tumor progression. Functional analyses reveal that both of these CX(3)CR1 subsets are immunosuppressive to naive CD8(+) and CD4(+) T-cell responses. Importantly, we demonstrate that CCR2 is a critical functional facilitator of leukocyte recruitment to the ovarian tumor microenvironment, and its genetic deletion results in a reduced tumor burden compared with wild-type mice. These results demonstrate that subsets of immunosuppressive leukocytes are recruited to the ovarian tumor environment through the CCR2 pathway, which offers a viable therapeutic target to inhibit their migration to the tumor site.


Asunto(s)
Monocitos/metabolismo , Neoplasias Ováricas/patología , Receptores de Quimiocina/metabolismo , Animales , Ascitis/inmunología , Ascitis/metabolismo , Ascitis/patología , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Receptor 1 de Quimiocinas CX3C , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunofenotipificación , Interferón gamma/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Leucocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Carga Tumoral
13.
J Reprod Immunol ; 80(1-2): 33-40, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19406482

RESUMEN

The human female reproductive tract (FRT) must balance the requirements of procreation with the demands of protection from pathogen invasion. We hypothesize that the FRT expresses functional pattern recognition receptors (PRRs), including Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD) proteins that may mediate these tasks. Expression of PRRs was evaluated in FRT tissues by RT-PCR. PRR function within FRT tissue cells was determined by CXCL8 (IL-8) production in response to treatment with PRR agonists. We now report that TLRs7-9 are expressed in Fallopian tube, uterine endometrium, cervix and ectocervix, while TLR10 expression is restricted to Fallopian tube. NOD1 and NOD2 and the signal transducer RICK were detected in all FRT tissues. Stimulation of FRT tissue cells with PRR ligands resulted in secretion of CXCL8. Results of these studies indicate that PRRs are functionally expressed in FRT tissues, and suggest that these receptors mediate microbial recognition and immune defense in the reproductive tract.


Asunto(s)
Trompas Uterinas/metabolismo , Interleucina-8/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Receptores Toll-Like/metabolismo , Útero/metabolismo , Adulto , Anciano , Aminoquinolinas/farmacología , Apoptosis/inmunología , Trompas Uterinas/efectos de los fármacos , Trompas Uterinas/inmunología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Imiquimod , Interleucina-8/genética , Interleucina-8/inmunología , Persona de Mediana Edad , Proteína Adaptadora de Señalización NOD1/inmunología , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/inmunología , Proteína Adaptadora de Señalización NOD2/metabolismo , Especificidad de Órganos , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/inmunología , Transducción de Señal , Receptores Toll-Like/agonistas , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Útero/efectos de los fármacos , Útero/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA