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











Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 13(8)2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923536

RESUMEN

The fallopian tube epithelium is the site of origin for a majority of high grade serous ovarian carcinomas (HGSOC). The chemical communication between the fallopian tube and the ovary in the development of HGSOC from the fallopian tube is of interest since the fimbriated ends in proximity of the ovary harbor serous tubal intraepithelial carcinoma (STICs). Epidemiological data indicates that androgens play a role in ovarian carcinogenesis; however, the oncogenic impact of androgen exposure on the fallopian tube, or tubal neoplastic precursor lesions, has yet to be explored. In this report, imaging mass spectrometry identified that testosterone is produced by the ovary when exposed to tumorigenic fallopian tube derived PTEN deficient cells. Androgen exposure increased cellular viability, proliferation, and invasion of murine cell models of healthy fallopian tube epithelium and PAX2 deficient models of the preneoplastic secretory cell outgrowths (SCOUTs). Proliferation and invasion induced by androgen was reversed by co-treatment with androgen receptor (AR) antagonist, bicalutamide. Furthermore, ablation of phosphorylated ERK reversed proliferation, but not invasion. Investigation of two hyperandrogenic rodent models of polycystic ovarian syndrome revealed that peripheral administration of androgens does not induce fallopian proliferation in vivo. These data suggest that tumorigenic lesions in the fallopian tube may induce an androgenic microenvironment proximal to the ovary, which may in turn promote proliferation of the fallopian tube epithelium and preneoplastic lesions.

2.
Am J Respir Cell Mol Biol ; 56(6): 708-715, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28118023

RESUMEN

Obliterative bronchiolitis (OB), characterized by fibrous obliteration of the small airways, is a major impediment to long-term survival in lung allograft recipients. We found previously that IL-17A is produced primarily by CD4+ T cells and γδ T cells after lung transplant in a mouse model of orthotopic lung transplant. The absence of either subset of T cells was compensated for by expansion of the other subset, which suggested that systemic blockade of IL-17A was necessary. To determine the specific role of IL-17A in the development of OB, we treated lung allograft recipients with an IL-17A antagonistic antibody. After IL-17A blockade, the incidence of OB was significantly reduced in lung allografts. IL-17A blockade also significantly attenuated the severity of acute rejection and overall lung fibrosis. The decreased OB incidence was associated with reduced lymphocyte recruitment, particularly CD8+ T cells and other IFN-γ-producing lymphocytes, to the lung allograft. Interestingly, IL-17A blockade led to an increase in the frequency of IL-17A-producing T-helper cell type 17 cells and γδ T cells in lung allografts, suggesting that IL-17A is a negative regulator of these T cells. Our data suggest that blocking IL-17A after lung transplant reduces the overall IFN-γ-mediated lymphocyte response and decreases the development of OB.


Asunto(s)
Aloinjertos/inmunología , Bronquiolitis Obliterante/inmunología , Inmunidad Celular/inmunología , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Trasplante de Pulmón , Animales , Bronquiolitis Obliterante/complicaciones , Bronquiolitis Obliterante/patología , Quimiocinas/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Células Th17/inmunología
3.
Immunol Cell Biol ; 95(3): 280-286, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27649780

RESUMEN

γδ T cells producing interleukin-17A (γδT17) are thought to develop spontaneously in the thymus and to be maintained in the periphery. Previous studies suggested a role for T-helper 17 (Th17) cells in the maintenance of γδT17 via the expression of transforming growth factor-ß1 (TGFß1). However, we have previously found that Th17 cells were not required for expansion of γδT17 cells after lung transplant in a mouse model. Using mice deficient in signal transducer and activator of transcription 3 (STAT3) in CD4+ T cells, which are unable to develop Th17 cells, we investigated the requirement for Th17 cells and TGFß1 to maintain γδT17 cells in the lung and lymphoid tissues. At steady state, we found no defect in γδT17 cells in the thymus or periphery of these mice. Further, STAT3-deficient CD4+ T cells produced significantly higher levels of TGFß1 than wild-type CD4+ T cells under Th17 differentiation conditions in vitro. To determine whether STAT3-deficient CD4+ T cells could expand γδT17 cells in vivo, we used TCRß-/- mice, which are known to have a defect in γδT17 cells that can be rescued by Th17 cells. However, adoptive transfer of wild-type Th17 cells or bulk CD4+ T cells did not expand γδT17 cells in TCRß-/- mice. In contrast, interferon-γ+ γδ T cells preferentially expanded, particularly in the lungs. Interestingly, we found in vivo and in vitro that TGFß1 may negatively regulate the pool of γδT17 cells. Our data suggest that Th17 cells and TGFß1 are not required for the maintenance of γδT17 cells.


Asunto(s)
Interleucina-17/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Células Th17/metabolismo , Animales , Diferenciación Celular , Polaridad Celular , Proliferación Celular , Homeostasis , Interferón gamma , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
4.
Am J Transplant ; 15(7): 1793-1804, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25773063

RESUMEN

Lung transplant survival is limited by obliterative bronchiolitis (OB), but the mechanisms of OB development are unknown. Previous studies in a mouse model of orthotopic lung transplantation suggested a requirement for IL-17. We have used this orthotopic mouse model to investigate the source of IL-17A and the requirement for T cells producing IL-17A. The major sources of IL-17A were CD4(+) T cells and γδ T cells. Depletion of CD4(+) T cells led to a significantly decreased frequency and number of IL-17A(+) lymphocytes and was sufficient to prevent acute rejection and OB. However, mice with STAT3-deficient T cells, which are unable to differentiate into Th17 cells, rejected lung allografts and developed OB similar to control mice. The frequency of IL-17A(+) cells was not decreased in mice with STAT3-deficient T cells due mainly to the presence of IL-17A(+) γδ T cells. Deficiency of γδ T cells also did not affect the development of airway fibrosis. Our data suggest that CD4(+) T cells are required for OB development and expansion of IL-17A responses in the lung, while Th17 and γδ T cells are not absolutely required and may compensate for each other.


Asunto(s)
Bronquiolitis Obliterante/inmunología , Linfocitos T CD4-Positivos/inmunología , Supervivencia de Injerto/inmunología , Interleucina-17/inmunología , Trasplante de Pulmón , Células Th17/inmunología , Animales , Bronquiolitis Obliterante/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Interferón gamma/metabolismo , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT3/fisiología
5.
J Immunol ; 191(8): 4431-9, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043901

RESUMEN

Obliterative bronchiolitis (OB) post-lung transplantation involves IL-17-regulated autoimmunity to type V collagen and alloimmunity, which could be enhanced by complement activation. However, the specific role of complement activation in lung allograft pathology, IL-17 production, and OB is unknown. The current study examines the role of complement activation in OB. Complement-regulatory protein (CRP) (CD55, CD46, complement receptor 1-related protein y/CD46) expression was downregulated in human and murine OB; and C3a, a marker of complement activation, was upregulated locally. IL-17 differentially suppressed complement receptor 1-related protein y expression in airway epithelial cells in vitro. Neutralizing IL-17 recovered CRP expression in murine lung allografts and decreased local C3a production. Exogenous C3a enhanced IL-17 production from alloantigen- or autoantigen (type V collagen)-reactive lymphocytes. Systemically neutralizing C5 abrogated the development of OB, reduced acute rejection severity, lowered systemic and local levels of C3a and C5a, recovered CRP expression, and diminished systemic IL-17 and IL-6 levels. These data indicated that OB induction is in part complement dependent due to IL-17-mediated downregulation of CRPs on airway epithelium. C3a and IL-17 are part of a feed-forward loop that may enhance CRP downregulation, suggesting that complement blockade could be a therapeutic strategy for OB.


Asunto(s)
Bronquiolitis Obliterante/inmunología , Activación de Complemento , Rechazo de Injerto/inmunología , Interleucina-17/metabolismo , Trasplante de Pulmón/efectos adversos , Animales , Autoinmunidad , Líquido del Lavado Bronquioalveolar , Antígenos CD55/biosíntesis , Colágeno Tipo V/inmunología , Complemento C3a/biosíntesis , Complemento C5 , Regulación hacia Abajo , Humanos , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Interleucina-6/biosíntesis , Prueba de Cultivo Mixto de Linfocitos , Proteína Cofactora de Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Receptores de Complemento/biosíntesis , Receptores de Complemento 3b
6.
PLoS One ; 8(8): e72955, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967339

RESUMEN

Acute rejection, a common complication of lung transplantation, may promote obliterative bronchiolitis leading to graft failure in lung transplant recipients. During acute rejection episodes, CD8(+) T cells can contribute to lung epithelial injury but the mechanisms promoting and controlling CD8-mediated injury in the lung are not well understood. To study the mechanisms regulating CD8(+) T cell-mediated lung rejection, we used a transgenic model in which adoptively transferred ovalbumin (OVA)-specific cytotoxic T lymphocytes (CTL) induce lung injury in mice expressing an ovalbumin transgene in the small airway epithelium of the lungs (CC10-OVA mice). The lung pathology is similar to findings in humans with acute lung transplant. In the presence of an intact immune response the inflammation resolves by day 30. Using CC10-OVA.RAG(-/-) mice, we found that CD4(+) T cells and ICOS(+/+) T cells were required for protection against lethal lung injury, while neutrophil depletion was not protective. In addition, CD4(+)Foxp3 (+) ICOS(+) T cells were enriched in the lungs of animals surviving lung injury and ICOS(+/+) Tregs promoted survival in animals that received ICOS(-/-) T cells. Direct comparison of ICOS(-/-) Tregs to ICOS(+/+) Tregs found defects in vitro but no differences in the ability of ICOS(-/-) Tregs to protect from lethal lung injury. These data suggest that ICOS affects Treg development but is not necessarily required for Treg effector function.


Asunto(s)
Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Lesión Pulmonar/genética , Lesión Pulmonar/inmunología , Trasplante de Pulmón/efectos adversos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Animales , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Silenciamiento del Gen , Rechazo de Injerto/mortalidad , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Lesión Pulmonar/patología , Subgrupos Linfocitarios/citología , Ratones , Neutrófilos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA