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










Intervalo de ano de publicação
1.
Front Immunol ; 12: 582858, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679734

RESUMO

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.


Assuntos
Barreira Hematotesticular/imunologia , Infecções por Escherichia coli/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/imunologia , Alvo Mecanístico do Complexo 2 de Rapamicina/imunologia , Infecções Urinárias/imunologia , Escherichia coli Uropatogênica/imunologia , Animais , Barreira Hematotesticular/metabolismo , Células Cultivadas , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Orquite/imunologia , Orquite/metabolismo , Orquite/microbiologia , Ratos Sprague-Dawley , Células de Sertoli/imunologia , Células de Sertoli/metabolismo , Células de Sertoli/microbiologia , Espermatogênese/imunologia , Testículo/imunologia , Testículo/metabolismo , Proteínas de Junções Íntimas/imunologia , Proteínas de Junções Íntimas/metabolismo , Infecções Urinárias/metabolismo , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/fisiologia
2.
Methods Mol Biol ; 2142: 81-92, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32367360

RESUMO

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.


Assuntos
Inflamação/diagnóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Zika virus/fisiologia , Animais , Barreira Hematotesticular/imunologia , Barreira Hematotesticular/metabolismo , Barreira Hematotesticular/virologia , Morte Celular , Ensaio de Imunoadsorção Enzimática/métodos , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/virologia , Masculino , Camundongos , Orquite/diagnóstico , Orquite/genética , Orquite/imunologia , Orquite/virologia , Testículo/patologia , Testículo/fisiologia , Zika virus/imunologia , Zika virus/patogenicidade , Infecção por Zika virus/complicações , Infecção por Zika virus/genética , Infecção por Zika virus/imunologia , Infecção por Zika virus/metabolismo
3.
Mol Reprod Dev ; 86(11): 1485-1504, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31518041

RESUMO

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.


Assuntos
Barreira Hematotesticular/imunologia , Fertilidade/imunologia , Tolerância Imunológica , Sêmen/imunologia , Espermatozoides/imunologia , Animais , Feminino , Humanos , Infertilidade Masculina/imunologia , Masculino , Testículo/imunologia , Útero/imunologia
4.
J Appl Toxicol ; 39(11): 1586-1605, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31415109

RESUMO

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.


Assuntos
Barreira Hematotesticular/efeitos dos fármacos , Aditivos Alimentares , Nanopartículas Metálicas/toxicidade , Epitélio Seminífero/efeitos dos fármacos , Células de Sertoli/efeitos dos fármacos , Titânio/toxicidade , Ração Animal/análise , Animais , Barreira Hematotesticular/imunologia , Barreira Hematotesticular/patologia , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Água Potável/química , Ingestão de Alimentos/efeitos dos fármacos , Aditivos Alimentares/toxicidade , Antígenos de Histocompatibilidade Classe II/imunologia , Masculino , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos BALB C , Tamanho da Partícula , Epitélio Seminífero/imunologia , Epitélio Seminífero/patologia , Túbulos Seminíferos/efeitos dos fármacos , Túbulos Seminíferos/imunologia , Túbulos Seminíferos/ultraestrutura , Células de Sertoli/imunologia , Células de Sertoli/ultraestrutura , Propriedades de Superfície , Titânio/administração & dosagem , Titânio/química
5.
J Leukoc Biol ; 104(4): 757-766, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30265772

RESUMO

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.


Assuntos
Microambiente Celular , Macrófagos/fisiologia , Testículo/imunologia , Animais , Antígenos CD/análise , Barreira Hematotesticular/imunologia , Corticosterona/metabolismo , Citocinas/metabolismo , Humanos , Sistema Imunitário/embriologia , Imunidade Inata , Imunofenotipagem , Macrófagos/classificação , Masculino , Orquite/imunologia , Orquite/metabolismo , Prostaglandinas/metabolismo , Tolerância a Antígenos Próprios , Espermatogênese , Espermatozoides/citologia , Espermatozoides/imunologia , Testículo/citologia , Testosterona/metabolismo
6.
Protein Pept Lett ; 25(5): 440-445, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29651939

RESUMO

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.


Assuntos
Barreira Hematotesticular/imunologia , Tolerância Imunológica , Células de Sertoli/imunologia , Espermatogênese/imunologia , Animais , Barreira Hematotesticular/citologia , Humanos , Masculino , Células de Sertoli/citologia
7.
J Virol ; 91(22)2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28878076

RESUMO

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.


Assuntos
Barreira Hematotesticular/imunologia , Células de Sertoli/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Barreira Hematotesticular/patologia , Barreira Hematotesticular/virologia , Moléculas de Adesão Celular/imunologia , Células Cultivadas , Claudinas/imunologia , Dengue/imunologia , Dengue/patologia , Vírus da Dengue/imunologia , Humanos , Interferon-alfa/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Receptores de Superfície Celular/imunologia , Células de Sertoli/patologia , Células de Sertoli/virologia , Molécula 1 de Adesão de Célula Vascular/imunologia , Infecção por Zika virus/patologia , Proteína da Zônula de Oclusão-1/imunologia
8.
Reproduction ; 154(3): 293-305, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28667125

RESUMO

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.


Assuntos
Apoptose , Doenças Autoimunes/fisiopatologia , Modelos Animais de Doenças , Folistatina/sangue , Orquite/fisiopatologia , Testículo/metabolismo , Regulação para Cima , Ativinas/antagonistas & inibidores , Ativinas/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Biomarcadores/sangue , Biomarcadores/metabolismo , Barreira Hematotesticular/imunologia , Barreira Hematotesticular/metabolismo , Barreira Hematotesticular/patologia , Barreira Hematotesticular/fisiopatologia , Progressão da Doença , Fibrose , Folistatina/administração & dosagem , Folistatina/genética , Folistatina/metabolismo , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Orquite/imunologia , Orquite/metabolismo , Orquite/patologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Proteínas Recombinantes/metabolismo , Testículo/imunologia , Testículo/patologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-28680856

RESUMO

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.


Assuntos
Células de Sertoli/imunologia , Células de Sertoli/patologia , Testículo/imunologia , Infecção por Zika virus/imunologia , Zika virus/patogenicidade , Animais , Antígenos Virais , Barreira Hematotesticular/imunologia , Barreira Hematotesticular/patologia , Barreira Hematotesticular/virologia , Linhagem Celular , Dengue/imunologia , Dengue/patologia , Vírus da Dengue/imunologia , Modelos Animais de Doenças , Masculino , Camundongos , Túbulos Seminíferos/patologia , Túbulos Seminíferos/virologia , Células de Sertoli/virologia , Espermatogênese , Taxa de Sobrevida , Testículo/patologia , Testículo/ultraestrutura , Testículo/virologia , Proteínas de Junções Íntimas/metabolismo , Transcriptoma , Carga Viral , Replicação Viral , Zika virus/imunologia , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
10.
Oncotarget ; 7(51): 84907-84923, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-27783995

RESUMO

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.


Assuntos
Barreira Hematotesticular/imunologia , Núcleo Celular/metabolismo , Fosfatase 1 de Especificidade Dupla/metabolismo , Epitélio Seminífero/metabolismo , Células de Sertoli/fisiologia , Testículo/imunologia , Junções Íntimas/metabolismo , Animais , Células Cultivadas , Microambiente Celular , Privilégio Imunológico , Inflamação , Lipopolissacarídeos/imunologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidor de NF-kappaB alfa/metabolismo , Ocludina/metabolismo , Transporte Proteico , Epitélio Seminífero/patologia
11.
Sci Rep ; 6: 18896, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26744177

RESUMO

Sertoli cells, can function as non-professional tolerogenic antigen-presenting cells, and sustain the blood-testis barrier formed by their tight junctions. The NOD-like receptor family members and the NALP3 inflammasome play a key role in pro-inflammatory innate immunity signalling pathways. Limited data exist on NOD1 and NOD2 expression in human and mouse Sertoli cells. Currently, there is no data on inflammasome expression or function in Sertoli cells. We found that in primary pre-pubertal Sertoli cells and in adult Sertoli line, TLR4\NOD1 and NOD2 crosstalk converged in NFκB activation and elicited a NALP3 activation, leading to de novo synthesis and inflammasome priming. This led to caspase-1 activation and IL-1ß secretion. We demonstrated this process was controlled by mechanisms linked to autophagy. NOD1 promoted pro-IL-1ß restriction and autophagosome maturation arrest, while NOD2 promoted caspase-1 activation, IL-1ß secretion and autophagy maturation. NALP3 modulated NOD1 and pro-IL-1ß expression, while NOD2 inversely promoted IL-1ß. This study is proof of concept that Sertoli cells, upon specific stimulation, could participate in male infertility pathogenesis via inflammatory cytokine induction.


Assuntos
Inflamassomos/imunologia , Interleucina-1beta/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD2/genética , Células de Sertoli/imunologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Autofagia/genética , Autofagia/imunologia , Barreira Hematotesticular/imunologia , Caspase 1/genética , Caspase 1/imunologia , Regulação da Expressão Gênica , Imunidade Inata , Inflamassomos/genética , Interleucina-1beta/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/genética , NF-kappa B/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Proteína Adaptadora de Sinalização NOD1/imunologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Células de Sertoli/citologia , Transdução de Sinais , Junções Íntimas/imunologia , Junções Íntimas/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
12.
Radiat Prot Dosimetry ; 167(1-3): 316-20, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25948832

RESUMO

Exposure to ionising radiation induces male infertility, accompanied by increasing permeability of the blood-testis barrier. However, the effect on male fertility by low-dose-rate chronic radiation has not been investigated. In this study, the effects of low-dose-rate chronic radiation on male mice were investigated by measuring the levels of tight-junction-associated proteins (ZO-1 and occludin-1), Niemann-Pick disease type 2 protein (NPC-2) and antisperm antibody (AsAb) in serum. BALB/c mice were exposed to low-dose-rate radiation (3.49 mGy h(-1)) for total exposures of 0.02 (6 h), 0.17 (2 d) and 1.7 Gy (21 d). Based on histological examination, the diameter and epithelial depth of seminiferous tubules were significantly decreased in 1.7-Gy-irradiated mice. Compared with those of the non-irradiated group, 1.7-Gy-irradiated mice showed significantly decreased ZO-1, occludin-1 and NPC-2 protein levels, accompanied with increased serum AsAb levels. These results suggest potential blood-testis barrier injury and immune infertility in male mice exposed to low-dose-rate chronic radiation.


Assuntos
Barreira Hematotesticular/lesões , Barreira Hematotesticular/efeitos da radiação , Infertilidade Masculina/imunologia , Exposição à Radiação/efeitos adversos , Lesões Experimentais por Radiação/imunologia , Testículo/efeitos da radiação , Animais , Barreira Hematotesticular/imunologia , Relação Dose-Resposta à Radiação , Infertilidade Masculina/etiologia , Infertilidade Masculina/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Doses de Radiação , Lesões por Radiação , Lesões Experimentais por Radiação/etiologia , Lesões Experimentais por Radiação/patologia
13.
Immunol Cell Biol ; 93(3): 311-20, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25403570

RESUMO

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.


Assuntos
Doenças Autoimunes/imunologia , Macrófagos/imunologia , Orquite/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Linfócitos T/imunologia , Testículo/imunologia , Animais , Autoanticorpos/imunologia , Autoantígenos/imunologia , Doenças Autoimunes/genética , Barreira Hematotesticular/imunologia , Modelos Animais de Doenças , Humanos , Tolerância Imunológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orquite/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
14.
PLoS One ; 9(4): e96120, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24760014

RESUMO

The testis is an organ with immune privilege. The comprehensive blood-testis barrier formed by Sertoli cells protects autoimmunogenic spermatozoa and spermatids from attack by the body's immune system. The interleukin (IL)-6/IL-12 family cytokines IL-12 (p35/p40), IL-23 (p19/p40), IL-27 (p28/Epstein-Barr virus-induced gene 3 [EBI3]), and IL-35 (p35/EBI3) play critical roles in the regulation of various immune responses, but their roles in testicular immune privilege are not well understood. In the present study, we investigated whether these cytokines are expressed in the testes and whether they function in the testicular immune privilege by using mice deficient in their subunits. Expression of EBI3 was markedly increased at both mRNA and protein levels in the testes of 10- or 12-week-old wild-type mice as compared with levels in 2-week-old mice, whereas the mRNA expression of p40 was markedly decreased and that of p35 was conserved between these two groups. Lack of EBI3, p35, and IL-12 receptor ß2 caused enhanced infiltration of lymphocytes into the testicular interstitium, with increased interferon-γ expression in the testes and autoantibody production against mainly acrosomal regions of spermatids. Spermatogenic disturbance was more frequently observed in the seminiferous tubules, especially when surrounded by infiltrating lymphocytes, of these deficient mice than in those of wild-type mice. In particular, p35-deficient mice showed the most severe spermatogenic disturbance. Immunohistochemical analyses revealed that endothelial cells and peritubular cells in the interstitium were highly positive for p35 at both ages, and CD163+ resident macrophages positive for p35 and EBI3, possibly producing IL-35, were also detected in the interstitium of 12-week-old mice but not those of 2-week-old mice. These results suggest that p35 helps in maintaining the testicular immune privilege, in part in an IL-35-dependent manner.


Assuntos
Barreira Hematotesticular/imunologia , Subunidade p35 da Interleucina-12/imunologia , Interleucinas/imunologia , Receptores de Citocinas/imunologia , Testículo/imunologia , Animais , Autoanticorpos/metabolismo , Células Endoteliais/metabolismo , Subunidade p35 da Interleucina-12/genética , Interleucinas/genética , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Antígenos de Histocompatibilidade Menor , Receptores de Citocinas/genética , Túbulos Seminíferos/citologia , Túbulos Seminíferos/metabolismo , Testículo/crescimento & desenvolvimento
15.
Semin Cell Dev Biol ; 30: 36-44, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24603046

RESUMO

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.


Assuntos
Barreira Hematotesticular/imunologia , Células de Sertoli/imunologia , Espermatogênese , Animais , Autoantígenos/imunologia , Autoantígenos/metabolismo , Autoimunidade , Humanos , Masculino , Tolerância Periférica , Células de Sertoli/fisiologia , Espermatozoides/imunologia , Espermatozoides/metabolismo , Testículo/citologia , Testículo/imunologia
16.
Immunol Cell Biol ; 91(6): 416-26, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23689306

RESUMO

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.


Assuntos
Linfócitos/imunologia , Macrófagos/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Células de Sertoli/imunologia , Testículo/enzimologia , Animais , Autoanticorpos/sangue , Barreira Hematotesticular/imunologia , Movimento Celular/imunologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Homeostase/genética , Homeostase/imunologia , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Espermatogênese/genética , Testículo/imunologia , Testículo/patologia , Regulação para Cima , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
17.
Nat Rev Immunol ; 13(3): 206-18, 2013 03.
Artigo em Inglês | MEDLINE | ID: mdl-23435332

RESUMO

Complex barriers separate immune-privileged tissues from the circulation. Here, we propose that cell entry to immune-privileged sites through barriers composed of tight junction-interconnected endothelium is associated with destructive inflammation, whereas border structures comprised of fenestrated vasculature enveloped by tightly regulated epithelium serve as active and selective immune-skewing gates in the steady state. Based on emerging knowledge of the central nervous system and information from other immune-privileged sites, we propose that these sites are endowed either with absolute endothelial-based barriers and epithelial gates that enable selective and educative transfer of trafficking leukocytes or with selective epithelial gates only.


Assuntos
Quimiotaxia de Leucócito , Vigilância Imunológica/fisiologia , Modelos Imunológicos , Junções Íntimas/fisiologia , Animais , Barreira Hematoaquosa/imunologia , Barreira Hematoaquosa/fisiologia , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/fisiologia , Barreira Hematorretiniana/imunologia , Barreira Hematorretiniana/fisiologia , Barreira Hematotesticular/imunologia , Barreira Hematotesticular/fisiologia , Fusão Celular , Quimerismo , Células Epiteliais/fisiologia , Células Epiteliais/ultraestrutura , Epitélio/imunologia , Epitélio/fisiologia , Feminino , Humanos , Tolerância Imunológica/imunologia , Vigilância Imunológica/imunologia , Inflamação/imunologia , Inflamação/fisiopatologia , Masculino , Troca Materno-Fetal/imunologia , Infiltração de Neutrófilos , Especificidade de Órgãos , Gravidez , Migração Transendotelial e Transepitelial/fisiologia
18.
Pharmacol Rev ; 64(1): 16-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22039149

RESUMO

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.


Assuntos
Barreira Hematotesticular/metabolismo , Anticoncepcionais Masculinos/farmacocinética , Animais , Barreira Hematotesticular/imunologia , Barreira Hematotesticular/fisiologia , Barreira Hematotesticular/ultraestrutura , Anticoncepcionais Masculinos/administração & dosagem , Sistemas de Liberação de Medicamentos , Hormônios Esteroides Gonadais/metabolismo , Humanos , Masculino , Modelos Biológicos , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/imunologia , Células de Sertoli/metabolismo , Células de Sertoli/ultraestrutura , Espermatogênese/efeitos dos fármacos
19.
Adv Exp Med Biol ; 763: 237-59, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23397628

RESUMO

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.


Assuntos
Barreira Hematotesticular/imunologia , Epididimo/imunologia , Testículo/imunologia , Animais , Barreira Hematotesticular/anatomia & histologia , Barreira Hematotesticular/fisiologia , Diferenciação Celular , Permeabilidade da Membrana Celular , Epididimo/anatomia & histologia , Epididimo/fisiologia , Sobrevivência de Enxerto/imunologia , Humanos , Tolerância Imunológica , Masculino , Filogenia , Epitélio Seminífero/imunologia , Epitélio Seminífero/fisiologia , Células de Sertoli/imunologia , Células de Sertoli/fisiologia , Células de Sertoli/ultraestrutura , Maturação do Esperma , Espermatogênese , Espermatozoides/imunologia , Espermatozoides/fisiologia , Testículo/anatomia & histologia , Testículo/fisiologia , Junções Íntimas/imunologia , Junções Íntimas/fisiologia , Junções Íntimas/ultraestrutura , Imunologia de Transplantes
20.
Expert Rev Clin Immunol ; 7(5): 627-34, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21895475

RESUMO

Increased numbers of mast cells (MCs) were described in the testes of males exhibiting infertility many years ago. Since beneficial effects of treatment with MC blockers on impaired male fertility were reported, more attention has been drawn on the role of MCs in the male reproductive tract. The main interest is focused on testicular MCs, however MCs also occur in the epididymis and seminal fluid, which may be relevant for fertility as well. The increase in testicular MCs in close contact to the seminiferous tubules indicates a relationship between MC proliferation and a dysfunction of the blood-testis barrier. Activated MCs not only coincide with fibrotic events, but also with elevated numbers of several types of immune cells in the testes of infertile men and may, therefore, be involved in the pathogenesis of testicular inflammatory processes as well. Outside the testis, MCs have really been assigned a key role in chronic protatitis/chronic pelvic pain syndrome. The occurrence of MCs in the seminal plasma of fertile/infertile men and negative effects on sperm functions has not been clarified so far and require further investigation. Optimistic reports on the beneficial effects of the treatment with MC blockers on disturbed male fertility also warrant further confirmation.


Assuntos
Barreira Hematotesticular/imunologia , Infertilidade Masculina/imunologia , Mastócitos/imunologia , Animais , Barreira Hematotesticular/patologia , Humanos , Infertilidade Masculina/patologia , Infertilidade Masculina/terapia , Inflamação/imunologia , Inflamação/patologia , Masculino , Mastócitos/patologia , Sêmen/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...