Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Mol Reprod Dev ; 86(11): 1485-1504, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31518041

RESUMEN

Declining fertility rates in both human and animals is a cause for concern. While many of the infertility cases are due to known causes, idiopathic infertility is reported in 30% of the infertile couples. In such cases, 18% of the infertile males carry antisperm antibodies (ASAs). Such data are lacking in livestock, wherein 20-30% of the animals are being culled due to low fertility. In males, the blood-testis barrier (BTB) and biomolecules in the semen provide an immuno-tolerant microenvironment for spermatozoa as they traverse the immunologic milieu of both the male and female reproductive tracts. For example, insults from environmental contaminants, infections and inflammatory conditions are likely to impact the immune privilege state of the testis and fertility. The female mucosal immune system can recognize allogenic spermatozoa-specific proteins affecting sperm kinematics and sperm-zona binding leading to immune infertility. Elucidating the functions and pathways of the immune regulatory molecules associated with fertilization are prerequisites for understanding their impact on fertility. An insight into biomolecules associated with spermatozoal immune tolerance may generate inputs to develop diagnostic tools and modulate fertility. High-throughput sequencing technologies coupled with bioinformatics analyses provides a path forward to define the array of molecules influencing pregnancy outcome. This review discusses the seminal immune regulatory molecules from their origin in the testis until they traverse the uterine environment enabling fertilization and embryonic development. Well-designed experiments and the identification of biomarkers may provide a pathway to understand the finer details of reproductive immunology that will afford personalized therapies.


Asunto(s)
Barrera Hematotesticular/inmunología , Fertilidad/inmunología , Tolerancia Inmunológica , Semen/inmunología , Espermatozoides/inmunología , Animales , Femenino , Humanos , Infertilidad Masculina/inmunología , Masculino , Testículo/inmunología , Útero/inmunología
2.
J Appl Toxicol ; 39(11): 1586-1605, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31415109

RESUMEN

Food-grade titanium dioxide labeled as E171 has been approved for human consumption by the Food and Drug Administration (USA) and by the European Union for five decades. However, titanium dioxide has been classified as a possible carcinogen for humans by the International Agency of Research in Cancer raising concerns of its oral intake and the translocation to bloodstream, which could disturb barriers such as the blood-testis barrier. There is evidence that titanium dioxide by intragastric/intraperitoneal/intravenous administration induced alterations on testosterone levels, testicular function and architecture, but studies of the E171 effects on the testicle structure and blood-testis barrier are limited. E171 is contained not only in foods in liquid matrix but also in solid ones, which can exert different biological effects. We aimed to compare the effects of E171 consumption in a solid matrix (0.1%, 0.5% and 1% in pellets) and liquid suspension (5 mg/kg body weight) on testis structure, inflammation infiltrate and blood-testis barrier disruption of male BALB/c mice. Results showed that none of the administration routes had influence on body weight but an increase in germ cell sloughing and the infiltrate of inflammatory cells in seminiferous tubules, together with disruption of the blood-testis barrier were similar in testis of both groups even if the dose received in mice in liquid matrix was 136 or 260 times lower than the dose reached by oral intake in solid E171 pellets in 0.5% E171 and 1% E171, respectively. This study highlights the attention on matrix food containing E171 and possible adverse effects on testis when E171 is consumed in a liquid matrix.


Asunto(s)
Barrera Hematotesticular/efectos de los fármacos , Aditivos Alimentarios , Nanopartículas del Metal/toxicidad , Epitelio Seminífero/efectos de los fármacos , Células de Sertoli/efectos de los fármacos , Titanio/toxicidad , Alimentación Animal/análisis , Animales , Barrera Hematotesticular/inmunología , Barrera Hematotesticular/patología , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Agua Potable/química , Ingestión de Alimentos/efectos de los fármacos , Aditivos Alimentarios/toxicidad , Antígenos de Histocompatibilidad Clase II/inmunología , Masculino , Nanopartículas del Metal/administración & dosificación , Nanopartículas del Metal/química , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Epitelio Seminífero/inmunología , Epitelio Seminífero/patología , Túbulos Seminíferos/efectos de los fármacos , Túbulos Seminíferos/inmunología , Túbulos Seminíferos/ultraestructura , Células de Sertoli/inmunología , Células de Sertoli/ultraestructura , Propiedades de Superficie , Titanio/administración & dosificación , Titanio/química
3.
J Virol ; 91(22)2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28878076

RESUMEN

Confirmed reports of Zika virus (ZIKV) in human seminal fluid for months after the clearance of viremia suggest the ability of ZIKV to establish persistent infection in the seminiferous tubules, an immune-privileged site in the testis protected by the blood-testis barrier, also called the Sertoli cell (SC) barrier (SCB). However, cellular targets of ZIKV in human testis and mechanisms by which the virus enters seminiferous tubules remain unclear. We demonstrate that primary human SCs were highly susceptible to ZIKV compared to the closely related dengue virus and induced the expression of alpha interferon (IFN-α), key cytokines, and cell adhesion molecules (vascular cell adhesion molecule 1 [VCAM-1] and intracellular adhesion molecule 1 [ICAM-1]). Furthermore, using an in vitro SCB model, we show that ZIKV was released on the adluminal side of the SCB model with a higher efficiency than in the blood-brain barrier model. ZIKV-infected SCs exhibited enhanced adhesion of leukocytes that correlated with decreases in SCB integrity. ZIKV infection did not affect the expression of tight and adherens junction proteins such as ZO-1, claudin, and JAM-A; however, exposure of SCs to inflammatory mediators derived from ZIKV-infected macrophages led to the degradation of the ZO-1 protein, which correlated with increased SCB permeability. Taken together, our data suggest that infection of SCs may be one of the crucial steps by which ZIKV gains access to the site of spermatozoon development and identify SCs as a therapeutic target to clear testicular infections. The SCB model opens up opportunities to assess interactions of SCs with other testicular cells and to test the ability of anti-ZIKV drugs to cross the barrier.IMPORTANCE Recent outbreaks of ZIKV, a neglected mosquito-borne flavivirus, have identified sexual transmission as a new route of disease spread, which has not been reported for other flaviviruses. To be able to sexually transmit for months after the clearance of viremia, ZIKV must establish infection in the seminiferous tubules, the site of spermatozoon development. However, little is known about the cell types that support ZIKV infection in the human testis. Currently, there are no models to study mechanisms of virus persistence in the seminiferous tubules. We provide evidence that ZIKV infection of human Sertoli cells, which are an important component of the seminiferous tubules, is robust and induces a strong antiviral response. The use of an in vitro Sertoli cell barrier to describe how ZIKV or inflammatory mediators derived from ZIKV-infected macrophages compromise barrier integrity will enable studies to explore the interactions of other testicular cells with Sertoli cells and to test novel antivirals for clearing testicular ZIKV infection.


Asunto(s)
Barrera Hematotesticular/inmunología , Células de Sertoli/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Barrera Hematotesticular/patología , Barrera Hematotesticular/virología , Moléculas de Adhesión Celular/inmunología , Células Cultivadas , Claudinas/inmunología , Dengue/inmunología , Dengue/patología , Virus del Dengue/inmunología , Humanos , Interferón-alfa/inmunología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Receptores de Superficie Celular/inmunología , Células de Sertoli/patología , Células de Sertoli/virología , Molécula 1 de Adhesión Celular Vascular/inmunología , Infección por el Virus Zika/patología , Proteína de la Zonula Occludens-1/inmunología
4.
Reproduction ; 154(3): 293-305, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28667125

RESUMEN

Experimental autoimmune orchitis (EAO) is a rodent model of chronic testicular inflammation that mimics the pathology observed in some types of human infertility. In a previous study, testicular expression of the inflammatory/immunoregulatory cytokine, activin A, was elevated in adult mice during the onset of EAO, indicating a potential role in the regulation of the disease. Consequently, we examined the development of EAO in mice with elevated levels of follistatin, an endogenous activin antagonist, as a potential therapeutic approach to testicular inflammation. Prior to EAO induction, mice received a single intramuscular injection of a non-replicative recombinant adeno-associated viral vector carrying a gene cassette of the circulating form of follistatin, FST315 (FST group). Serum follistatin levels were increased 5-fold in the FST group compared with the control empty vector (EV) group at 30 and 50 days of EAO, but intra-testicular levels of follistatin or activin A were not significantly altered. Induction of EAO was reduced, but not prevented, with mild-to-severe damage in 75% of the EV group and 40% of the FST group, at 50 days following immunisation with testicular homogenate. However, the EAO damage score (based on disruption of the blood-testis barrier, apoptosis, testicular damage and fibrosis) and extent of intratesticular inflammation (expression of inflammatory mediators) were directly proportional to the levels of activin A measured in the testis at 50 days. These data implicate activin A in the progression of EAO, thereby providing a potential therapeutic target; however, elevating circulating follistatin levels were not sufficient to prevent EAO development.


Asunto(s)
Apoptosis , Enfermedades Autoinmunes/fisiopatología , Modelos Animales de Enfermedad , Folistatina/sangre , Orquitis/fisiopatología , Testículo/metabolismo , Regulación hacia Arriba , Activinas/antagonistas & inhibidores , Activinas/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Biomarcadores/sangre , Biomarcadores/metabolismo , Barrera Hematotesticular/inmunología , Barrera Hematotesticular/metabolismo , Barrera Hematotesticular/patología , Barrera Hematotesticular/fisiopatología , Progresión de la Enfermedad , Fibrosis , Folistatina/administración & dosificación , Folistatina/genética , Folistatina/metabolismo , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Orquitis/inmunología , Orquitis/metabolismo , Orquitis/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/sangre , Proteínas Recombinantes/metabolismo , Testículo/inmunología , Testículo/patología
5.
Semin Cell Dev Biol ; 30: 36-44, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24603046

RESUMEN

Testicular germ cells, which appear after the establishment of central tolerance, express novel cell surface and intracellular proteins that can be recognized as 'foreign antigens' by the host's immune system. However, normally these germ cells do not evoke an auto-reactive immune response. The focus of this manuscript is to review the evidence that the blood-testis-barrier (BTB)/Sertoli cell (SC) barrier along with the SCs ability to modulate the immune response is vital for protecting auto-antigenic germ cells. In normal testis, the BTB/SC barrier protects the majority of the auto-antigenic germ cells by limiting access by the immune system and sequestering these 'new antigens'. SCs also modulate testis immune cells (induce regulatory immune cells) by expressing several immunoregulatory factors, thereby creating a local tolerogenic environment optimal for survival of nonsequesetred auto-antigenic germ cells. Collectively, the fortress created by the BTB/SC barrier along with modulation of the immune response is pivotal for completion of spermatogenesis and species survival.


Asunto(s)
Barrera Hematotesticular/inmunología , Células de Sertoli/inmunología , Espermatogénesis , Animales , Autoantígenos/inmunología , Autoantígenos/metabolismo , Autoinmunidad , Humanos , Masculino , Tolerancia Periférica , Células de Sertoli/fisiología , Espermatozoides/inmunología , Espermatozoides/metabolismo , Testículo/citología , Testículo/inmunología
6.
Immunol Cell Biol ; 93(3): 311-20, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25403570

RESUMEN

The mammalian testis is an immunoprivileged organ where male germ cell autoantigens are immunologically ignored. Both systemic immune tolerance to autoantigens and local immunosuppressive milieu contribute to the testicular immune privilege. Testicular immunosuppression has been intensively studied, but information on systemic immune tolerance to autoantigens is lacking. In the present study, we aimed to determine the role of Axl and Mer receptor tyrosine kinases in maintaining the systemic tolerance to male germ cell antigens using the experimental autoimmune orchitis (EAO) model. Axl and Mer double-knockout (Axl(-/-)Mer(-/-)) mice developed evident EAO after a single immunization with germ cell homogenates emulsified with complete Freund's adjuvant. EAO was characterized by the accumulation of macrophages and T lymphocytes in the testis. Damage to the seminiferous epithelium was also observed. EAO induction was associated with pro-inflammatory cytokine upregulation in the testes, impaired permeability of the blood-testis barrier and generation of autoantibodies against germ cell antigens in Axl(-/-)Mer(-/-) mice. Immunization also induced mild EAO in Axl or Mer single-gene-knockout mice. By contrast, a single immunization failed to induce EAO in wild-type mice. The results indicate that Axl and Mer receptors cooperatively regulate the systemic immune tolerance to male germ cell antigens.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Macrófagos/inmunología , Orquitis/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Linfocitos T/inmunología , Testículo/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/genética , Barrera Hematotesticular/inmunología , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Orquitis/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
7.
Pharmacol Rev ; 64(1): 16-64, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22039149

RESUMEN

The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in the mammalian body. It divides the seminiferous epithelium into the basal and the apical (adluminal) compartments. Meiosis I and II, spermiogenesis, and spermiation all take place in a specialized microenvironment behind the BTB in the apical compartment, but spermatogonial renewal and differentiation and cell cycle progression up to the preleptotene spermatocyte stage take place outside of the BTB in the basal compartment of the epithelium. However, the BTB is not a static ultrastructure. Instead, it undergoes extensive restructuring during the seminiferous epithelial cycle of spermatogenesis at stage VIII to allow the transit of preleptotene spermatocytes at the BTB. Yet the immunological barrier conferred by the BTB cannot be compromised, even transiently, during the epithelial cycle to avoid the production of antibodies against meiotic and postmeiotic germ cells. Studies have demonstrated that some unlikely partners, namely adhesion protein complexes (e.g., occludin-ZO-1, N-cadherin-ß-catenin, claudin-5-ZO-1), steroids (e.g., testosterone, estradiol-17ß), nonreceptor protein kinases (e.g., focal adhesion kinase, c-Src, c-Yes), polarity proteins (e.g., PAR6, Cdc42, 14-3-3), endocytic vesicle proteins (e.g., clathrin, caveolin, dynamin 2), and actin regulatory proteins (e.g., Eps8, Arp2/3 complex), are working together, apparently under the overall influence of cytokines (e.g., transforming growth factor-ß3, tumor necrosis factor-α, interleukin-1α). In short, a "new" BTB is created behind spermatocytes in transit while the "old" BTB above transiting cells undergoes timely degeneration, so that the immunological barrier can be maintained while spermatocytes are traversing the BTB. We also discuss recent findings regarding the molecular mechanisms by which environmental toxicants (e.g., cadmium, bisphenol A) induce testicular injury via their initial actions at the BTB to elicit subsequent damage to germ-cell adhesion, thereby leading to germ-cell loss, reduced sperm count, and male infertility or subfertility. Moreover, we also critically evaluate findings in the field regarding studies on drug transporters in the testis and discuss how these influx and efflux pumps regulate the entry of potential nonhormonal male contraceptives to the apical compartment to exert their effects. Collectively, these findings illustrate multiple potential targets are present at the BTB for innovative contraceptive development and for better delivery of drugs to alleviate toxicant-induced reproductive dysfunction in men.


Asunto(s)
Barrera Hematotesticular/metabolismo , Anticonceptivos Masculinos/farmacocinética , Animales , Barrera Hematotesticular/inmunología , Barrera Hematotesticular/fisiología , Barrera Hematotesticular/ultraestructura , Anticonceptivos Masculinos/administración & dosificación , Sistemas de Liberación de Medicamentos , Hormonas Esteroides Gonadales/metabolismo , Humanos , Masculino , Modelos Biológicos , Células de Sertoli/efectos de los fármacos , Células de Sertoli/inmunología , Células de Sertoli/metabolismo , Células de Sertoli/ultraestructura , Espermatogénesis/efectos de los fármacos
8.
Immunol Cell Biol ; 91(6): 416-26, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23689306

RESUMEN

Tyro3, Axl and Mer (TAM) receptor tyrosine kinases triple knockout (TAM(-/-)) mice are male infertile due to impaired spermatogenesis. However, the mechanism by which TAM receptors regulate spermatogenesis remains unclear. In this study, we demonstrate that the testicular immune homeostasis was impaired in TAM(-/-) mice. As development after the onset of sexual maturity, germ cells were progressively degenerated. Macrophages and lymphocytes infiltrated into the testis as TAM(-/-) mice aged. Moreover, the integrity of blood-testis barrier was impaired, and the autoantibodies against germ cell antigens were produced. Major inflammatory cytokines, including tumor necrosis factor-α, interleukin-6 and monocyte chemotactic protein 1 were upregulated in the testis of TAM(-/-) mice, and predominantly located in Sertoli cells (SCs). In vitro assays showed that TAM(-/-) SCs secrete significantly high levels of inflammatory cytokines compared with wild-type SCs after coculture with apoptotic germ cells. These results suggest that TAM receptors are important in the maintenance of the immune homeostasis in the testis.


Asunto(s)
Linfocitos/inmunología , Macrófagos/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Células de Sertoli/inmunología , Testículo/enzimología , Animales , Autoanticuerpos/sangre , Barrera Hematotesticular/inmunología , Movimiento Celular/inmunología , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Homeostasis/genética , Homeostasis/inmunología , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Espermatogénesis/genética , Testículo/inmunología , Testículo/patología , Regulación hacia Arriba , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
9.
Adv Exp Med Biol ; 763: 237-59, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23397628

RESUMEN

The blood-testis barrier (BTB) is known for its ability to create an immune privilege site in the seminiferous epithelium, but less is known of the blood-epididymal barrier (BEB). It is already established that the fully functional BTB and BEB are much more complex and consist of anatomical/physical (tight junctions, basolateral and apical membranes), physiological and immunological components, which are all necessary to make a functioning barrier in the testis and epididymis. However, comparative data for metazoans suggest that an effective Sertoli cell barrier is not entirely necessary for the development of germ cells during spermatogenesis or that our knowledge about the barrier structure/function in metazoans is still immature. This chapter compares the unique barrier formed by the Sertoli cells of the testis to that formed by the apical junctional complexes of the epididymal epithelium.


Asunto(s)
Barrera Hematotesticular/inmunología , Epidídimo/inmunología , Testículo/inmunología , Animales , Barrera Hematotesticular/anatomía & histología , Barrera Hematotesticular/fisiología , Diferenciación Celular , Permeabilidad de la Membrana Celular , Epidídimo/anatomía & histología , Epidídimo/fisiología , Supervivencia de Injerto/inmunología , Humanos , Tolerancia Inmunológica , Masculino , Filogenia , Epitelio Seminífero/inmunología , Epitelio Seminífero/fisiología , Células de Sertoli/inmunología , Células de Sertoli/fisiología , Células de Sertoli/ultraestructura , Maduración del Esperma , Espermatogénesis , Espermatozoides/inmunología , Espermatozoides/fisiología , Testículo/anatomía & histología , Testículo/fisiología , Uniones Estrechas/inmunología , Uniones Estrechas/fisiología , Uniones Estrechas/ultraestructura , Inmunología del Trasplante
10.
Biol Reprod ; 84(5): 851-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21209417

RESUMEN

The terms blood-testis barrier (BTB) or blood-epididymis barrier (BEB), are often described as Sertoli cell-Sertoli cell tight junctions (TJs) or TJs between the epithelial cells in the epididymis, respectively. However, in reality, the BTB and BEB are much more complex than just the TJ. The focus of this minireview is to remind readers that the complete BTB and BEB are comprised of three components: anatomical, physiological, and immunological. The TJs form the anatomical (physical) barrier that restricts passage of molecules and cells from entering or exiting the lumen. The physiological barrier is comprised of transporters that regulate movement of substances in or out of the lumen, thus creating a microenvironment, which is critical for the proper development and maturation of germ cells. The immunological barrier limits access by the immune system and sequesters the majority of the autoantigenic germ cells. Combined with the overall immune-privilege of the testis, this suppresses detrimental immune responses against the autoantigenic germ cells. These three components on their own do not create a complete functional barrier; instead, it is the interaction between all three components that create a barrier of maximal competence.


Asunto(s)
Barrera Hematotesticular/fisiología , Epidídimo/fisiología , Uniones Estrechas/metabolismo , Animales , Autoantígenos/metabolismo , Barrera Hematotesticular/inmunología , Epidídimo/anatomía & histología , Epidídimo/inmunología , Humanos , Masculino , Permeabilidad , Espermatogénesis , Espermatozoides/citología , Espermatozoides/inmunología , Testículo/anatomía & histología , Testículo/inmunología
11.
Front Immunol ; 12: 582858, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679734

RESUMEN

The structural and functional destruction of the blood-testis barrier (BTB) following uropathogenic E. coli (UPEC) infection may be a critical component of the pathologic progress of orchitis. Recent findings indicate that the mammalian target of the rapamycin (mTOR)-signaling pathway is implicated in the regulation of BTB assembly and restructuring. To explore the mechanisms underlying BTB damage induced by UPEC infection, we analyzed BTB integrity and the involvement of the mTOR-signaling pathway using in vivo and in vitro UPEC-infection models. We initially confirmed that soluble virulent factors secreted from UPEC trigger a stress response in Sertoli cells and disturb adjacent cell junctions via down-regulation of junctional proteins, including occludin, zonula occludens-1 (ZO-1), F-actin, connexin-43 (CX-43), ß-catenin, and N-cadherin. The BTB was ultimately disrupted in UPEC-infected rat testes, and blood samples from UPEC-induced orchitis in these animals were positive for anti-sperm antibodies. Furthermore, we herein also demonstrated that mTOR complex 1 (mTORC1) over-activation and mTORC2 suppression contributed to the disturbance in the balance between BTB "opening" and "closing." More importantly, rapamycin (a specific mTORC1 inhibitor) significantly restored the expression of cell-junction proteins and exerted a protective effect on the BTB during UPEC infection. We further confirmed that short-term treatment with rapamycin did not aggravate spermatogenic degeneration in infected rats. Collectively, this study showed an association between abnormal activation of the mTOR-signaling pathway and BTB impairment during UPEC-induced orchitis, which may provide new insights into a potential treatment strategy for testicular infection.


Asunto(s)
Barrera Hematotesticular/inmunología , Infecciones por Escherichia coli/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Diana Mecanicista del Complejo 2 de la Rapamicina/inmunología , Infecciones Urinarias/inmunología , Escherichia coli Uropatógena/inmunología , Animales , Barrera Hematotesticular/metabolismo , Células Cultivadas , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Orquitis/inmunología , Orquitis/metabolismo , Orquitis/microbiología , Ratas Sprague-Dawley , Células de Sertoli/inmunología , Células de Sertoli/metabolismo , Células de Sertoli/microbiología , Espermatogénesis/inmunología , Testículo/inmunología , Testículo/metabolismo , Proteínas de Uniones Estrechas/inmunología , Proteínas de Uniones Estrechas/metabolismo , Infecciones Urinarias/metabolismo , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/fisiología
12.
Methods Mol Biol ; 2142: 81-92, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32367360

RESUMEN

Inflammation is part of a defense reaction of live tissues that is triggered by pathogens, chemical reagents, trauma, and radiation. Understanding the inflammatory process triggered by Zika virus (ZIKV) is important to better understand the pathogen-host interaction. The evaluation of this process can be done using tools such as enzyme-linked immunosorbent assay (ELISA) and quantitative reverse transcription PCR (RT-qPCR). Both techniques have been an indispensable tool not just for immunologists but for all interested in understanding the inflammatory process.


Asunto(s)
Inflamación/diagnóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Virus Zika/fisiología , Animales , Barrera Hematotesticular/inmunología , Barrera Hematotesticular/metabolismo , Barrera Hematotesticular/virología , Muerte Celular , Ensayo de Inmunoadsorción Enzimática/métodos , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/virología , Masculino , Ratones , Orquitis/diagnóstico , Orquitis/genética , Orquitis/inmunología , Orquitis/virología , Testículo/patología , Testículo/fisiología , Virus Zika/inmunología , Virus Zika/patogenicidad , Infección por el Virus Zika/complicaciones , Infección por el Virus Zika/genética , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/metabolismo
13.
Okajimas Folia Anat Jpn ; 85(3): 91-6, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19227199

RESUMEN

Spermatoza contain various autoimmunogenic materials, which are recognized as foreign by the self immune system. Therefore, the blood-tesits-barrier (BTB) formed by Sertoli cells, basal lamina and myoid cells protects autoimmunogeneic spermatozoa from attack by the self immune system. However, the BTB at the tubuli recti (TR) and the rete testis (RT) is known to be incomplete against humoral substances. We investigated here whether the BTB is physiologically penetrated by lymphocytes in mice. We performed light and electron microscopical observation of the seminiferous tubules (ST), the TR and the RT in normal C3H/IHe mice. Although no lymphocytes were observed inside the ST, a very few lymphocytes could be found beyond the basal lamina of the TR and the RT. These lymphocytes were close to testicular spermatozoa in the TR lumen. These findings provide a possibility that lymphocytes may gain access to autoantigens of spermatozoa inside the TR and RT under physiological conditions in mice.


Asunto(s)
Autoinmunidad/fisiología , Barrera Hematotesticular/ultraestructura , Linfocitos/ultraestructura , Red Testicular/ultraestructura , Túbulos Seminíferos/ultraestructura , Testículo/ultraestructura , Animales , Autoantígenos/inmunología , Membrana Basal/inmunología , Membrana Basal/ultraestructura , Barrera Hematotesticular/inmunología , Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C3H , Microscopía Electrónica de Transmisión , Red Testicular/inmunología , Túbulos Seminíferos/inmunología , Células de Sertoli/inmunología , Células de Sertoli/ultraestructura , Espermatogénesis/inmunología , Espermatozoides/inmunología , Espermatozoides/ultraestructura , Testículo/inmunología
14.
Protein Pept Lett ; 25(5): 440-445, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29651939

RESUMEN

BACKGROUND: Sertoli cell, over the past 30 years, have been elevated from simple mechanical elements to the rank of a "sentinel" in spermatogenesis. By delivering potent immunomodulatory and trophic proteins, Sertoli cells are unique cell type with a pivotal role in maintaining testis immune privilege and the immune-protection of the antigenic germ cells. CONCLUSIONS: The findings from SC transplantation studies utilizing experimental animal models of disease, demonstrate the presence of the same immuno-modulation properties and mechanisms at tissue and organ sites far from testis. The complex pathways that generate and maintain the immune tolerance involve the production of several immunomodulatory or immune-related proteins such as cytokines, chemokines, growth factors, mediators of the inflammation, complement inhibitors or adhesion molecules. A better definition and understanding of these Sertoli cell proteins and the mechanisms of immunoprotection should help to elucidate their role in the spermatogenic process. The demonstration of their capabilities in transplantation experiments suggests that Sertoli cells may be good candidates in cell therapy for a number of cell-mediated chronic diseases.


Asunto(s)
Barrera Hematotesticular/inmunología , Tolerancia Inmunológica , Células de Sertoli/inmunología , Espermatogénesis/inmunología , Animales , Barrera Hematotesticular/citología , Humanos , Masculino , Células de Sertoli/citología
15.
J Leukoc Biol ; 104(4): 757-766, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30265772

RESUMEN

Testicular macrophages (TM) comprise the largest immune cell population in the mammalian testis. They are characterized by a subdued proinflammatory response upon adequate stimulation, and a polarization toward the immunoregulatory and immunotolerant M2 phenotype. This enables them to play a relevant role in supporting the archetypical functions of the testis, namely spermatogenesis and steroidogenesis. During infection, the characteristic blunted immune response of TM reflects the need for a delicate balance between a sufficiently strong reaction to counteract invading pathogens, and the prevention of excessive proinflammatory cytokine levels with the potential to disturb or destroy spermatogenesis. Local microenvironmental factors that determine the special phenotype of TM have just begun to be unraveled, and are discussed in this review.


Asunto(s)
Microambiente Celular , Macrófagos/fisiología , Testículo/inmunología , Animales , Antígenos CD/análisis , Barrera Hematotesticular/inmunología , Corticosterona/metabolismo , Citocinas/metabolismo , Humanos , Sistema Inmunológico/embriología , Inmunidad Innata , Inmunofenotipificación , Macrófagos/clasificación , Masculino , Orquitis/inmunología , Orquitis/metabolismo , Prostaglandinas/metabolismo , Autotolerancia , Espermatogénesis , Espermatozoides/citología , Espermatozoides/inmunología , Testículo/citología , Testosterona/metabolismo
16.
Ann N Y Acad Sci ; 1120: 144-51, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17911411

RESUMEN

The transcription factor Ets-variant gene 5 (ETV5) is essential for spermatogonial stem cell (SSC) self-renewal, as the targeted deletion of the Etv5 gene in mice (Etv5(-/-)) results in only the first wave of spermatogenesis. Reciprocal transplants of neonatal germ cells from wild-type (WT) and Etv5(-/-) testes were performed to determine the role of ETV5 in Sertoli cells and germ cells. ETV5 appears to be needed in both cell types for normal spermatogenesis. In addition, Etv5(-/-) recipients displayed increased interstitial inflammation and tubular involution after transplantation. Preliminary studies suggest that the blood-testis barrier (Sertoli-Sertoli tight junctional complex) is abnormal in the Etv5(-/-) mouse.


Asunto(s)
Barrera Hematotesticular/fisiología , Proteínas de Unión al ADN/fisiología , Inmunidad Innata/genética , Espermatogénesis/genética , Testículo/inmunología , Factores de Transcripción/fisiología , Animales , Barrera Hematotesticular/inmunología , Proteínas de Unión al ADN/genética , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Uniones Estrechas/inmunología , Uniones Estrechas/fisiología , Factores de Transcripción/genética
17.
Invest Clin ; 48(1): 107-21, 2007 Mar.
Artículo en Español | MEDLINE | ID: mdl-17432549

RESUMEN

The pathogenesis of male infertility can be reflected in alterations of spermatogenesis caused by testicular cancer, aplasia of the germinal cells, varicocele, environmental factors or defect in the transport of the sperms, among others. In general, 48% of men suffer unexplained infertility. During a long time, the masculine reproductive tract and the immune system have been studied as different and independent systems. However, in the last two decades a particular interest has arisen in the interaction of both systems on masculine infertility, in particular in the evaluation of antisperm antibodies as a common cause of infertility. Also, the inflammation due to genital or systemic infections can cause alterations in the testicular function. The recognition of intratesticular antigens provokes the production of antibodies by B lymphocytes. Then, the immune system induces a cellular response, by cytokines secretion, activation of complement and T lymphocytes activation. In this review the components and the immune system response mechanism, the organization of the testicle as a reproductive organ and the mediators of the immunologic response will be examined: interleukin-1 (IL-1), IL-6, leukaemia Inhibitory factor, tumor necrosis factor-alpha, the molecule FasL (CD95L) and Fas (CD95), macrophage migration-inhibitory factor, mononuclear phagocyte colony stimulating factor, Granulocyte/macrophage colony stimulating factor, as well as stem cell factor, interferon, transforming growth factor B and activins.


Asunto(s)
Citocinas/fisiología , Infertilidad Masculina/etiología , Testículo/fisiopatología , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Barrera Hematotesticular/inmunología , Activación de Complemento , Humanos , Infertilidad Masculina/inmunología , Inflamación/complicaciones , Activación de Linfocitos , Masculino , Espermatozoides/inmunología
18.
Artículo en Inglés | MEDLINE | ID: mdl-28680856

RESUMEN

Flaviviruses including Dengue virus (DENV), Yellow fever virus (YFV), West Nile virus (WNV), and Japanese encephalitis virus (JEV) are global health problems that caused several serious diseases such as fever, hemorrhagic fever, and encephalitis in the past century. Recently, Zika virus (ZIKV) which spreads from Asia to American and causes millions of infections emerges as a new dangerous member of the genus of Flavivirus. Unlike other well-known flaviviruses, ZIKV can be transmitted sexually and infect testes in murine models. Its impacts on sperm functions, and the exact susceptible cells, however, are not entirely clear. To investigate these issues, we infected interferon α/ß and γ receptors deficient AG6 mice with ZIKV and examined the outcomes of infection using an assortment of physiological, histopathological, immunological, and virological techniques. We found that infected mice displayed signs of reproductive system disorder, altered androgen levels in serum, and high viral load in semen and testes. Additionally, histopathological examinations revealed marked atrophy of seminiferous tubules and significant reduction in lumen size. Notably, these were accompanied by positive staining of ZIKV antigens on sertoli cells, detection of viral particles and vacuole changes within cytoplasm of sertoli cells. The susceptibility of sertoli cells to ZIKV was further validated in vitro study using cell lines. Importantly, the disruption of tight junctions within testis and altered sperm morphology were also observed in ZIKV infected mice. It is well-known that tight junctions formed by adjacent sertoli cells are major component of blood testis barrier, which plays important roles in maintenance of microenvironment for spermagenesis in testis. Taken together, these results demonstrate that sertoli cells are susceptible to ZIKV infection, which results in the disruption of tight junctions in testis and causes abnormal spermatogenesis in mice. These results also imply that long-term impact of ZIKV infection on human male reproductive system requires close monitoring.


Asunto(s)
Células de Sertoli/inmunología , Células de Sertoli/patología , Testículo/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/patogenicidad , Animales , Antígenos Virales , Barrera Hematotesticular/inmunología , Barrera Hematotesticular/patología , Barrera Hematotesticular/virología , Línea Celular , Dengue/inmunología , Dengue/patología , Virus del Dengue/inmunología , Modelos Animales de Enfermedad , Masculino , Ratones , Túbulos Seminíferos/patología , Túbulos Seminíferos/virología , Células de Sertoli/virología , Espermatogénesis , Tasa de Supervivencia , Testículo/patología , Testículo/ultraestructura , Testículo/virología , Proteínas de Uniones Estrechas/metabolismo , Transcriptoma , Carga Viral , Replicación Viral , Virus Zika/inmunología , Infección por el Virus Zika/patología , Infección por el Virus Zika/virología
19.
J Reprod Immunol ; 69(1): 23-34, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16380167

RESUMEN

On most tissues, multiple membrane complement regulators (CReg) protect self-cells from damage by complement. An exception is the brain, where the blood-brain barrier provides a protected environment within which cells survive with little or no protection from complement. The testis has a functionally similar structure, the blood-testis barrier (BTB). Here, we have investigated the expression of C3/C5 convertase CReg and C3 in the normal rat testis at different ages and different spermatogenetic stages, as well as in rats in which spermatogenesis and the BTB were impaired due to a developmental deficit. Immature testis, prior to BTB formation at puberty, displayed broad expression of the ubiquitous rodent CReg Crry on all elements and no expression of CD46 or CD55. Within days of BTB formation, CReg expression was dramatically altered; Crry was expressed only in the spermatogenetic cells external to the BTB in basal layers of adult seminal epithelium. Spermatogenic cells immediately distal to the BTB at first expressed no C3/C5 convertase regulators but later acquired expression of CD46 and CD55. Staining for C3 was widespread pre-puberty, but absent distal to the BTB in mature rats. In rats with defects in spermatogenesis and BTB integrity, expression patterns of CReg and C3 resembled those in pre-pubertal normals. The relative paucity of CReg and absence of C3 synthesis distal to the BTB suggest the presence of a complement-protected environment analogous to that described in the brain, and suggest also that cells enclosed by the BTB may be susceptible to complement damage when the barrier is breached.


Asunto(s)
Barrera Hematotesticular/inmunología , Complemento C3/metabolismo , Proteínas Inactivadoras de Complemento/metabolismo , Espermatogénesis , Testículo/crecimiento & desarrollo , Testículo/inmunología , Animales , Complemento C3/análisis , Convertasas de Complemento C3-C5/metabolismo , Proteínas Inactivadoras de Complemento/análisis , Proteínas Inactivadoras de Complemento/genética , Masculino , Mutación , Ratas , Ratas Wistar , Epitelio Seminífero/química , Epitelio Seminífero/crecimiento & desarrollo , Epitelio Seminífero/inmunología , Espermatogénesis/genética , Testículo/citología
20.
Oncotarget ; 7(51): 84907-84923, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27783995

RESUMEN

Sertoli cells create a local tolerogenic microenvironment to maintain testicular immune privilege especially through the formation of a blood-testis barrier (BTB). However, the molecular mechanisms underlying the immune modulation function and BTB dynamics of Sertoli cells remained elusive. MAP phosphatase (MKP)-1 acts as a crucial negative regulator of the inflammatory response. Nevertheless, the role of MKP-1 in regulating Sertoli cells has not been elucidated. In this study, we have for the first time uncovered distinct cellular localization of MKP-1 in the cells at different stages of mouse testis, and the level of MKP-1 expression was significantly up-regulated by LPS-induced acute testis inflammation. In addition, MKP-1 staining was strongly detected in nuclei and peri-nuclear regions of cytoplasm in the Sertoli cells, and it was presented at Sertoli cell tight junctions (TJs) at stages VII-VIII after LPS treatment. Moreover, we demonstrated that MKP-1 was capable of attenuating LPS-induced decrease of occludin by interaction with p38 MAP kinase and IκBα molecules. Taken together, our data highlight that MKP-1 was an important endogenous suppressor of innate immune responses involved in the regulation of BTB barrier dynamic. This study thus might offer novel targets for treating inflammatory diseases in the testis.


Asunto(s)
Barrera Hematotesticular/inmunología , Núcleo Celular/metabolismo , Fosfatasa 1 de Especificidad Dual/metabolismo , Epitelio Seminífero/metabolismo , Células de Sertoli/fisiología , Testículo/inmunología , Uniones Estrechas/metabolismo , Animales , Células Cultivadas , Microambiente Celular , Privilegio Inmunológico , Inflamación , Lipopolisacáridos/inmunología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa/metabolismo , Ocludina/metabolismo , Transporte de Proteínas , Epitelio Seminífero/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA