Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Am J Pathol ; 191(9): 1592-1609, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34126085

RESUMEN

Male factors, including those of autoimmune origin, contribute to approximately 50% of infertility cases in humans. However, the mechanisms underlying autoimmune male infertility are poorly understood. Deficiency in autoimmune regulator (AIRE) impairs central immune tolerance because of diminished expression of self-antigens in the thymus. Humans with AIRE mutations and mice with engineered ablation of Aire develop multiorgan autoimmunity and infertility. To determine the immune targets contributing to infertility in male Aire-deficient (-/-) mice, Aire-/- or wild-type (WT) males were paired with WT females. Aire-/- males exhibited dramatically reduced mating frequency and fertility, hypogonadism, and reduced serum testosterone. Approximately 15% of mice exhibited lymphocytic infiltration into the testis, accompanied by atrophy, azoospermia, and reduced numbers of mitotically active germ cells; the remaining mice showed normal testicular morphology, sperm counts, and motility. However, spermatozoa from all Aire-/- mice were defective in their ability to fertilize WT oocytes in vitro. Lymphocytic infiltration into the epididymis, seminal vesicle, and prostate gland was evident. Aire-/- male mice generated autoreactive antibodies in an age-dependent manner against sperm, testis, epididymis, prostate gland, and seminal vesicle. Finally, expression of Aire was evident in the seminiferous epithelium in an age-dependent manner, as well as in the prostate gland. These findings suggest that Aire-dependent central tolerance plays a critical role in maintaining male fertility by stemming autoimmunity against multiple reproductive targets.


Asunto(s)
Infertilidad Masculina/inmunología , Poliendocrinopatías Autoinmunes/patología , Factores de Transcripción/metabolismo , Animales , Femenino , Infertilidad Masculina/genética , Masculino , Ratones , Ratones Noqueados , Poliendocrinopatías Autoinmunes/genética , Factores de Transcripción/genética , Proteína AIRE
2.
Biol Reprod ; 100(6): 1492-1504, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30770532

RESUMEN

Autoimmune Regulator (AIRE) regulates central immune tolerance by inducing expression of tissue-restricted antigens in thymic medullary epithelial cells, thereby ensuring elimination of autoreactive T cells. Aire mutations in humans and targeted Aire deletion in mice result in multiorgan autoimmune disease, known in humans as autoimmune polyglandular syndrome type 1 (APS-1). APS-1 is characterized by the presence of adrenal insufficiency, chronic mucosal candidiasis, and/or hypoparathyroidism. Additionally, females often present with gonadal insufficiency and infertility. Aire-deficiency (KO) in mice results in oophoritis and age-dependent depletion of follicular reserves. Here, we found that while the majority of young 6-week-old Aire-KO females had normal follicular reserves, mating behavior, and ovulation rates, 50% of females experienced embryonic loss between gestation day (GD) 5.5 and 7.5 that could not be attributed to insufficient progesterone production or decidualization. The quality of GD0.5 embryos recovered from Aire KO mice was reduced, and when cultured in vitro, embryos displayed limited developmental capacity in comparison to those recovered from wild-type (WT) mice. Further, embryos flushed from Aire KO dams at GD3.5 were developmentally delayed in comparison to WT controls and had reduced trophoblastic outgrowth in vitro. We conclude that AIRE does not play a direct role in uterine decidualization. Rather, reduced fertility of Aire-deficient females is likely due to multiple factors, including oophoritis, delayed preimplantation development, and compromised implantation. These effects may be explained by autoimmune targeting of the ovary, embryo, or both. Alternatively, altered embryonic development could be due to a direct role for AIRE in early embryogenesis.


Asunto(s)
Implantación del Embrión/fisiología , Desarrollo Embrionario/fisiología , Factores de Transcripción/metabolismo , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Factores de Transcripción/genética , Proteína AIRE
3.
J Immunol ; 195(8): 3737-47, 2015 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-26342030

RESUMEN

Hepatitis C virus (HCV) is the world's most common blood-borne viral infection for which there is no vaccine. The rates of vertical transmission range between 3 and 6% with odds 90% higher in the presence of HIV coinfection. Prevention of vertical transmission is not possible because of lack of an approved therapy for use in pregnancy or an effective vaccine. Recently, HCV has been identified as an independent risk factor for preterm delivery, perinatal mortality, and other complications. In this study, we characterized the immune responses that contribute to the control of viral infection at the maternal-fetal interface (MFI) in the early gestational stages. In this study, we show that primary human trophoblast cells and an extravillous trophoblast cell line (HTR8), from first and second trimester of pregnancy, express receptors relevant for HCV binding/entry and are permissive for HCV uptake. We found that HCV-RNA sensing by human trophoblast cells induces robust upregulation of type I/III IFNs and secretion of multiple chemokines that elicit recruitment and activation of decidual NK cells. Furthermore, we observed that HCV-RNA transfection induces a proapoptotic response within HTR8 that could affect the morphology of the placenta. To our knowledge, for the first time, we demonstrate that HCV-RNA sensing by human trophoblast cells elicits a strong antiviral response that alters the recruitment and activation of innate immune cells at the MFI. This work provides a paradigm shift in our understanding of HCV-specific immunity at the MFI as well as novel insights into mechanisms that limit vertical transmission but may paradoxically lead to virus-related pregnancy complications.


Asunto(s)
Hepacivirus/inmunología , Hepatitis C/inmunología , Inmunidad Materno-Adquirida , Células Asesinas Naturales/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Trofoblastos/inmunología , Adulto , Femenino , Hepatitis C/patología , Hepatitis C/transmisión , Humanos , Inmunidad Innata , Transmisión Vertical de Enfermedad Infecciosa , Células Asesinas Naturales/patología , Embarazo , Complicaciones Infecciosas del Embarazo/virología , Trofoblastos/patología
5.
Biol Reprod ; 86(4): 110, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22219212

RESUMEN

Loss-of-function mutations in the autoimmune regulator (AIRE) gene are responsible for autoimmune polyglandular syndrome type 1 (APS-1), which commonly manifests as infertility in women. AIRE is a transcriptional regulator that promotes expression of tissue-restricted antigens in the thymus, including antigens specific to the ovary. Thymic expression of ovarian genes under AIRE's control may be critical for preventing ovarian autoimmune disease. Because mice lacking Aire are an important APS-1 model, we examined the reproductive properties of female Aire-deficient (Aire(-/-)) mice. Female Aire(-/-) mice on the BALB/c background were examined for reproductive parameters, including fertility, litter sizes, and ovarian follicular reserves. Although delayed puberty was observed in Aire(-/-) mice, all mice entered puberty and exhibited mating behavior. Only 50% of Aire(-/-) females gave an initial litter, and only 16% were able to produce two litters. Ovarian histopathologic examination revealed that 83% of previously bred females lost all ovarian follicular reserves. Among virgin females, follicular depletion was observed in 25% by 8 wk, and by 20 wk, 50%-60% of mice lost all follicles. This was associated with elevated serum follicle-stimulating hormone level and ovarian infiltration of proliferating CD3+ T lymphocytes. Ovulation rates of 6-wk-old Aire(-/-) mice were reduced by 22%, but this difference was not statistically significant. Finally, transplantation experiments revealed that follicular loss depended on factors extrinsic to the ovary. These results suggest that immune-mediated ovarian follicular depletion is a mechanism of infertility in Aire(-/-) mice. The results have important implications in the pathogenesis of ovarian autoimmune disease in women.


Asunto(s)
Envejecimiento Prematuro/genética , Infertilidad Femenina/genética , Folículo Ovárico/inmunología , Poliendocrinopatías Autoinmunes/genética , Factores de Transcripción/genética , Envejecimiento Prematuro/inmunología , Animales , Complejo CD3 , Femenino , Fertilización/inmunología , Hormona Folículo Estimulante/sangre , Infertilidad Femenina/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Modelos Animales , Folículo Ovárico/patología , Ovulación/inmunología , Poliendocrinopatías Autoinmunes/inmunología , Subgrupos de Linfocitos T , Factores de Transcripción/inmunología , Proteína AIRE
6.
Chest ; 150(3): e81-5, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27613994

RESUMEN

A 64-year-old male former smoker with a history of prostate cancer presented to our pulmonary clinic, complaining of nonproductive cough for 10 years. Prior evaluation included treatment for upper airway cough syndrome and gastroesophageal reflux, stopping angiotensin-converting enzyme inhibitor, and initiation of inhaled ß-agonists. Esophageal pH monitoring indicated silent reflux, and proton pump inhibitor therapy was started. He continued to cough and complain of dyspnea. Physical examination produced unremarkable results, with no evidence of lymphadenopathy. Pulmonary function tests showed a pseudo-restrictive pattern with air trapping, hyperreactivity, and incomplete bronchodilator responsiveness: FEV1, 2.48 L (69% of predicted); FVC, 3.57 L (75% of predicted); FEV1/FVC, 92%; total lung capacity, 7.00 L (100% of predicted); and residual volume, 3.05 L (136% of predicted). Laboratory studies, including a complete metabolic panel, prostate-specific antigen test, and complete blood count, yielded normal results.


Asunto(s)
Tumor Carcinoide/diagnóstico por imagen , Neoplasias Pulmonares/diagnóstico por imagen , Pulmón/diagnóstico por imagen , Nódulos Pulmonares Múltiples/diagnóstico por imagen , Tumor Carcinoide/complicaciones , Tumor Carcinoide/patología , Tumor Carcinoide/fisiopatología , Enfermedad Crónica , Tos/etiología , Disnea/etiología , Fluorodesoxiglucosa F18 , Volumen Espiratorio Forzado , Humanos , Pulmón/fisiopatología , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/fisiopatología , Masculino , Persona de Mediana Edad , Nódulos Pulmonares Múltiples/complicaciones , Nódulos Pulmonares Múltiples/patología , Nódulos Pulmonares Múltiples/fisiopatología , Tomografía de Emisión de Positrones , Radiofármacos , Volumen Residual , Factores de Tiempo , Tomografía Computarizada por Rayos X , Capacidad Pulmonar Total , Capacidad Vital
7.
Cell Mol Immunol ; 11(6): 510-21, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25327908

RESUMEN

The ovary is not an immunologically privileged organ, but a breakdown in tolerogenic mechanisms for ovary-specific antigens has disastrous consequences on fertility in women, and this is replicated in murine models of autoimmune disease. Isolated ovarian autoimmune disease is rare in women, likely due to the severity of the disease and the inability to transmit genetic information conferring the ovarian disease across generations. Nonetheless, autoimmune oophoritis is often observed in association with other autoimmune diseases, particularly autoimmune adrenal disease, and takes a toll on both society and individual health. Studies in mice have revealed at least two mechanisms that protect the ovary from autoimmune attack. These mechanisms include control of autoreactive T cells by thymus-derived regulatory T cells, as well as a role for the autoimmune regulator (AIRE), a transcriptional regulator that induces expression of tissue-restricted antigens in medullary thymic epithelial cells during development of T cells. Although the latter mechanism is incompletely defined, it is well established that failure of either results in autoimmune-mediated targeting and depletion of ovarian follicles. In this review, we will address the clinical features and consequences of autoimmune-mediated ovarian infertility in women, as well as the possible mechanisms of disease as revealed by animal models.


Asunto(s)
Enfermedades de las Glándulas Suprarrenales/inmunología , Enfermedades Autoinmunes/inmunología , Infertilidad Femenina/inmunología , Enfermedades del Ovario/inmunología , Linfocitos T Reguladores/inmunología , Enfermedades de las Glándulas Suprarrenales/complicaciones , Animales , Enfermedades Autoinmunes/complicaciones , Modelos Animales de Enfermedad , Femenino , Humanos , Infertilidad Femenina/etiología , Ratones , Enfermedades del Ovario/complicaciones , Factores de Transcripción/metabolismo , Proteína AIRE
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA