Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 22(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374605

ABSTRACT

The immune privilege of the testes is necessary to prevent immune attacks to gamete-specific antigens and paternal major histocompatibility complex (MHC) antigens, allowing for normal spermatogenesis. However, infection and inflammation of the male genital tract can break the immune tolerance and represent a significant cause of male infertility. Different T cell subsets have been identified in mammalian testes, which may be involved in the maintenance of immune tolerance and pathogenic immune responses in testicular infection and inflammation. We reviewed the evidence in the published literature on different T subtypes (regulatory T cells, helper T cells, cytotoxic T cells, γδ T cells, and natural killer T cells) in human and animal testes that support their regulatory roles in infertility and the orchitis pathology. While many in vitro studies have indicated the regulation potential of functional T cell subsets and their possible interaction with Sertoli cells, Leydig cells, and spermatogenesis, both under physiological and pathological processes, there have been no in situ studies to date. Nevertheless, the normal distribution and function of T cell subsets are essential for the immune privilege of the testes and intact spermatogenesis, and T cell-mediated immune response drives testicular inflammation. The distinct function of different T cell subsets in testicular homeostasis and the orchitis pathology suggests a considerable potential of targeting specific T cell subsets for therapies targeting chronic orchitis and immune infertility.


Subject(s)
Immunity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Testis/immunology , Testis/metabolism , Animals , Autoimmunity , Biomarkers , Disease Management , Disease Susceptibility , Homeostasis , Humans , Immunomodulation , Leydig Cells/immunology , Leydig Cells/metabolism , Male , Sertoli Cells/immunology , Sertoli Cells/metabolism , Spermatogenesis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
J Appl Toxicol ; 39(11): 1586-1605, 2019 11.
Article in English | MEDLINE | ID: mdl-31415109

ABSTRACT

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.


Subject(s)
Blood-Testis Barrier/drug effects , Food Additives , Metal Nanoparticles/toxicity , Seminiferous Epithelium/drug effects , Sertoli Cells/drug effects , Titanium/toxicity , Animal Feed/analysis , Animals , Blood-Testis Barrier/immunology , Blood-Testis Barrier/pathology , Body Weight/drug effects , Dose-Response Relationship, Drug , Drinking Water/chemistry , Eating/drug effects , Food Additives/toxicity , Histocompatibility Antigens Class II/immunology , Male , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Particle Size , Seminiferous Epithelium/immunology , Seminiferous Epithelium/pathology , Seminiferous Tubules/drug effects , Seminiferous Tubules/immunology , Seminiferous Tubules/ultrastructure , Sertoli Cells/immunology , Sertoli Cells/ultrastructure , Surface Properties , Titanium/administration & dosage , Titanium/chemistry
3.
J Virol ; 91(22)2017 11 15.
Article in English | MEDLINE | ID: mdl-28878076

ABSTRACT

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.


Subject(s)
Blood-Testis Barrier/immunology , Sertoli Cells/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Blood-Testis Barrier/pathology , Blood-Testis Barrier/virology , Cell Adhesion Molecules/immunology , Cells, Cultured , Claudins/immunology , Dengue/immunology , Dengue/pathology , Dengue Virus/immunology , Humans , Interferon-alpha/immunology , Macrophages/immunology , Macrophages/pathology , Male , Receptors, Cell Surface/immunology , Sertoli Cells/pathology , Sertoli Cells/virology , Vascular Cell Adhesion Molecule-1/immunology , Zika Virus Infection/pathology , Zonula Occludens-1 Protein/immunology
4.
Microb Pathog ; 123: 60-67, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29959039

ABSTRACT

It is conceivable that early developing germ cells must across the basal to the luminal region of seminiferous tubules (STs) during spermatogenesis is associated with extensive restructuring of junctional complex. However, very limited information is documented about these junctional complexes in reptiles. In the present study we have determined the localization of inter-Sertoli cell tight junctions (TJ's), protein CLDN11 and gap junction protein Cx43 during spermatogenesis in the testis. In early spermatogenesis, weak immunoreactivity of CLDN11and focal localization of Cx43 was observed around the Sertoli cell in the luminal region, but completely delaminated from the basal compartment of STs. In late spermatogenesis, strong focal to linear localization of CLDN11and Cx43 was detected at the points of contact between two Sertoli cells and around the early stages of primary spermatocytes in the basal compartment of STs. In late spermatogenesis, localization of CLDN11and Cx43 was drastically reduced and seen only around Sertoli cells and spermatogonia near the basal lamina. However, transmission electron microscopy revealed that inter-Sertoli cell tight junctions were present within the basal compartment of STs, leaving the spermatogonia and early primary spermatocytes in the basal region during mid spermatogenesis. Gap junctions were observed between Sertoli cells, and Sertoli cells with spermatogonia and primary spermatocytes throughout spermatogenesis. Moreover, adherens and hemidesmosomes junctions were observed during spermatogenesis. The above findings collectively suggest that the intensity and localization of TJ's and gap junctions vary according to the spermatogenetic stages that might be protected the developing germ cells from own immune response.


Subject(s)
Adherens Junctions/physiology , Autoimmunity/immunology , Hemidesmosomes/physiology , Sertoli Cells/cytology , Sertoli Cells/immunology , Spermatogenesis/physiology , Tight Junctions/physiology , Animals , Claudins/metabolism , Connexin 43/metabolism , Male , Microscopy, Electron, Transmission , Spermatocytes/physiology , Spermatogonia/physiology , Turtles
5.
Semin Cell Dev Biol ; 30: 36-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24603046

ABSTRACT

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.


Subject(s)
Blood-Testis Barrier/immunology , Sertoli Cells/immunology , Spermatogenesis , Animals , Autoantigens/immunology , Autoantigens/metabolism , Autoimmunity , Humans , Male , Peripheral Tolerance , Sertoli Cells/physiology , Spermatozoa/immunology , Spermatozoa/metabolism , Testis/cytology , Testis/immunology
6.
Immunology ; 148(3): 253-65, 2016 07.
Article in English | MEDLINE | ID: mdl-26878424

ABSTRACT

Sertoli cells (SCs) possess inherent immunosuppressive properties and are major contributors to the immunoprivileged status of mammalian testis. SCs have been reported to inhibit the activation of B cells, T cells and natural killer cells but not dendritic cells (DCs). Herein, we present evidence that co-culture with SCs results in a persistent state of DC immaturity characterized by down-regulation of the surface molecules I-A/E, CD80, CD83, CD86, CCR7 and CD11c, as well as reduced production of pro-inflammatory cytokines. SC-conditioned DCs (SC-DCs) displayed low immunogenicity and enhanced immunoregulatory functions, including the inhibition of T-cell proliferation and the promotion of Foxp3(+) regulatory T-cell development. Mechanistically, the activation of p38, extracellular signal-regulated kinase 1/2, and signal transducer and activator of transcription 3 was suppressed in SC-DCs. More importantly, we demonstrate that galectin-1 secreted by SCs plays a pivotal role in the differentiation of functionally tolerogenic SC-DCs. These findings further support the role of SCs in maintaining the immunoprivileged environment of the testis and provide a novel approach to derive tolerogenic DCs, which may lead to alternative therapeutic strategies for the treatment of immunopathogenic diseases.


Subject(s)
Dendritic Cells/immunology , Galectin 1/metabolism , Immune Tolerance , Sertoli Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Forkhead Transcription Factors/metabolism , Galectin 1/immunology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Reprod Fertil Dev ; 28(6): 757-64, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25399480

ABSTRACT

Genital tract infection and inflammation may affect male fertility, causing germ and Sertoli cell loss. We determined if testicular cell transplantation is effective at repairing testicular injury induced by herpes simplex virus (HSV) orchitis. ROSA26 mice were used as donors and the recipients were C57BL/6 mice after HSV testicular inoculation; some of the recipients were treated with the antiviral drug acyclovir (ACV). ACV reduced the amount of HSV antigen in testes on Day 3 after transplantation and enhanced the efficacy of transplantation at Day 30. In recipient testes, donor Sertoli cells formed new seminiferous tubules; significantly more new tubules were observed in the testes of ACV-treated mice compared with mice not treated with ACV (17.8% vs 3.6%). Over half (50.4%) of new tubules in ACV-treated testes contained germ cells and round spermatids were detected in 14.2% of new tubules compared with 15.9% and 5.3% in testes not treated with ACV, respectively. At Day 150 the seminiferous epithelium was completely recovered in some donor tubules and elongated spermatids were observed inside it. Thus, our findings reveal the effectiveness of the combination of antiviral therapy with neonatal testis-cell transplantation for the restoration of spermatogenesis damaged by viral infection.


Subject(s)
Cell Transplantation/methods , Herpes Genitalis/physiopathology , Infertility, Male/therapy , Orchitis/etiology , Spermatogenesis , Testis/transplantation , Acyclovir/adverse effects , Acyclovir/therapeutic use , Animals , Animals, Newborn , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Cell Transplantation/adverse effects , Combined Modality Therapy/adverse effects , Herpes Genitalis/drug therapy , Herpes Genitalis/immunology , Herpes Genitalis/virology , Immunohistochemistry , Infertility, Male/etiology , Infertility, Male/pathology , Infertility, Male/prevention & control , Kinetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Orchitis/immunology , Orchitis/metabolism , Orchitis/prevention & control , Seminiferous Tubules/drug effects , Seminiferous Tubules/immunology , Seminiferous Tubules/metabolism , Seminiferous Tubules/pathology , Sertoli Cells/drug effects , Sertoli Cells/immunology , Sertoli Cells/metabolism , Sertoli Cells/pathology , Simplexvirus/drug effects , Simplexvirus/immunology , Simplexvirus/isolation & purification , Spermatids/drug effects , Spermatids/immunology , Spermatids/metabolism , Spermatids/pathology , Spermatogenesis/drug effects , Testis/drug effects , Testis/metabolism , Testis/pathology
8.
Pharmacol Rev ; 64(1): 16-64, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22039149

ABSTRACT

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.


Subject(s)
Blood-Testis Barrier/metabolism , Contraceptive Agents, Male/pharmacokinetics , Animals , Blood-Testis Barrier/immunology , Blood-Testis Barrier/physiology , Blood-Testis Barrier/ultrastructure , Contraceptive Agents, Male/administration & dosage , Drug Delivery Systems , Gonadal Steroid Hormones/metabolism , Humans , Male , Models, Biological , Sertoli Cells/drug effects , Sertoli Cells/immunology , Sertoli Cells/metabolism , Sertoli Cells/ultrastructure , Spermatogenesis/drug effects
9.
Biol Reprod ; 91(1): 18, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24876407

ABSTRACT

The mechanism by which noninfectious testicular inflammation results in infertility is poorly understood. Here the infiltration of CD11b+ immunoreactive testicular interstitial cells (neutrophil, macrophages, dendritic cells) in immature (Postnatal Day [PND] 21, 28, and 35) and adult (PND 56) Fischer rats is described at 12, 24, and 48 h after an oral dose of 1 g/kg mono-(2-ethylhexyl) phthalate (MEHP), a well-described Sertoli cell toxicant. Increases of CD11b+ cells are evident 12 h after MEHP exposure in PND 21 and 28 rats. In PND 28 rats, CD11b+ cells remained significantly elevated at 48 h, while in PND 21 rats, it returned to control levels by 24 h. The peak number of CD11b+ cells in PND 35 rat testis is delayed until 24 h, but remains significantly elevated at 48 h. In PND 56 rats, no increase in CD11b+ cells occurs after MEHP exposure. In PND 21, 28, and 35 rats, a significant increase in monocyte chemoattractant protein-1 (MCP-1) by peritubular myoid cells occurs 12 h after MEHP. Interestingly, MEHP treatment of C57BL/6J mice did not incite an infiltration of CD11b+ cells at either PND 21 or 28. The peak level of germ cell apoptosis observed 24 h after MEHP exposure in young rats is not seen in mice at any age or in PND 56 rats. Taken together, these findings implicate MCP-1 released by peritubular myoid cells in provoking the migration of CD11b+ cells into the immature rat testis early after MEHP exposure and point to a role for CD11b+ cells in triggering germ cell apoptosis in an age- and species-dependent manner.


Subject(s)
Diethylhexyl Phthalate/analogs & derivatives , Macrophages/drug effects , Sertoli Cells/drug effects , Testis/drug effects , Age Factors , Animals , Cell Movement/physiology , Chemokine CCL2/metabolism , Diethylhexyl Phthalate/pharmacology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Rats , Sertoli Cells/immunology , Sertoli Cells/metabolism , Species Specificity , Testis/immunology , Testis/metabolism
10.
J Urol ; 191(2): 458-64, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24012581

ABSTRACT

PURPOSE: Undescended testis is the most common defect in male newborns. This condition is associated with increased risks of infertility and testicular malignancy due to abnormal germ cell development in the testes. Early surgery may limit such risks. We analyzed germ cell development vs age at orchiopexy using a germ cell marker and a Sertoli cell marker on testicular biopsies. MATERIALS AND METHODS: A total of 22 testicular biopsies at orchiopexy in 20 patients 5 to 24.5 months old were fixed and embedded in paraffin. Sections were processed and labeled with AMH antibody for Sertoli cells and MVH antibody for germ cells for immunofluorescent histochemical analysis. Confocal images were counted using ImageJ (National Institutes of Health, Bethesda, Maryland) for germ cells and testicular tubules. The data were analyzed using linear regression. RESULTS: Sertoli cells were clearly distinguished from MVH positive and negative germ cells located centrally or on basement membranes of tubules. Percentage of tubules with MVH negative germ cells significantly decreased with increasing age at orchiopexy (ß = -0.03, p = 0.03). Total tubular numbers and "empty" tubules without germ cells significantly increased with age at orchiopexy (ß = 1.15, p = 0.02 and ß = 0.44, p = 0.04, respectively). CONCLUSIONS: AMH antibody distinguished Sertoli cells from germ cells, and MVH antibody distinguished 2 types of germ cells at different developmental stages. Biopsy at orchiopexy in older patients showed significant germ cell depletion. These results lend support to early surgery to optimize germ cell number.


Subject(s)
Cryptorchidism/pathology , Cryptorchidism/surgery , Germ Cells/pathology , Orchiopexy , Sertoli Cells/pathology , Age Factors , Anti-Mullerian Hormone/immunology , Basement Membrane/pathology , Biopsy , Cell Death , Child, Preschool , DEAD-box RNA Helicases/immunology , Fibrosis , Germ Cells/immunology , Humans , Immunohistochemistry , Infant , Male , Microscopy, Confocal , Sertoli Cells/immunology , Testis/pathology
11.
Reproduction ; 148(3): 259-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24920664

ABSTRACT

Sertoli cells (SCs) play an important physiological role in the testis, as they support, nourish, and protect the germ cells. As protection of the developing spermatozoa is an emerging aspect of reproductive physiology, this study examined the expression pattern of innate immune-related genes, including avian ß-defensins (AvBDs), Toll-like receptors (TLRs), and cytokines, and investigated the time course of an inflammatory response in rooster SCs triggered by exposure to the bacterial endotoxin lipopolysaccharide (LPS). SCs were isolated from 6-week-old chicken, cultured in vitro, and stimulated with 1 µg/ml LPS at different time courses (0, 6, 12, 24, and 48  h). Data on expression analysis revealed that all ten members of the chicken TLR family, nine members of the AvBD family, as well as eight cytokine genes were expressed in SCs. Quantitative real-time PCR analysis revealed that LPS treatment resulted in significant induction of the expression levels of six TLRs, six AvBDs, and four cytokine genes, while two cytokine genes were downregulated and two other genes were unchanged. The increasing interleukin 1ß (IL1ß) production was confirmed in the conditioned medium. Furthermore, the phagocytosis of SCs was increased after LPS treatment. In conclusion, these findings provide evidence that SCs express innate immune-related genes and respond directly to bacterial ligands. These genes represent an important component of the immune system, which could be integrated into semen, and present a distinctive constituent of the protective repertoire of the testis against ascending infections.


Subject(s)
Cytokines/metabolism , Immunity, Innate/immunology , Lipopolysaccharides/pharmacology , Sertoli Cells/immunology , Animals , Cells, Cultured , Chickens , Cytokines/genetics , Immunity, Innate/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Phagocytosis/immunology , Sertoli Cells/drug effects , Sertoli Cells/metabolism
12.
J Reprod Immunol ; 165: 104291, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986230

ABSTRACT

The male reproductive system provides a distinctive shield to the immune system, safeguarding germ cells (GCs) from autoimmune harm. The testis in mammals creates a unique immunological setting due to its exceptional immune privilege and potent local innate immunity. which can result from a number of different circumstances, including disorders of the pituitary gland, GC aplasia, and immunological elements. Apoptosis, or programmed cell death (PCD), is essential for mammalian spermatogenesis to maintain and ensure an appropriate number of GCs that correspond with the supporting capability of the Sertoli cells. Apoptosis is substantial in controlling the number of GCs in the testis throughout spermatogenesis, and any dysregulation of this process has been linked to male infertility. There is a number of evidence about the potential of PCD in designing novel therapeutic approaches in the treatment of infertility. A detailed understanding of PCD and the processes that underlie immunological infertility can contribute to the progress in designing strategies to prevent and treat male infertility. This review will provide a summary of the role of immune cell death in male reproduction and infertility and describe the therapeutic strategies and agents for treatment based on immune cell death.


Subject(s)
Apoptosis , Infertility, Male , Spermatogenesis , Male , Humans , Spermatogenesis/immunology , Infertility, Male/immunology , Animals , Apoptosis/immunology , Testis/immunology , Testis/pathology , Sertoli Cells/immunology , Sertoli Cells/pathology , Fertility/immunology , Immunity, Innate
13.
Immunol Cell Biol ; 91(6): 416-26, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23689306

ABSTRACT

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.


Subject(s)
Lymphocytes/immunology , Macrophages/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Sertoli Cells/immunology , Testis/enzymology , Animals , Autoantibodies/blood , Blood-Testis Barrier/immunology , Cell Movement/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Homeostasis/genetics , Homeostasis/immunology , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Spermatogenesis/genetics , Testis/immunology , Testis/pathology , Up-Regulation , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
14.
Biol Reprod ; 86(1): 1-14, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21900683

ABSTRACT

Primary Sertoli cells isolated from mouse testes survive when transplanted across immunological barriers and protect cotransplanted allogeneic and xenogeneic cells from rejection in rodent models. In contrast, the mouse Sertoli cell line (MSC-1) lacks immunoprotective properties associated with primary Sertoli cells. In this study, enriched primary Sertoli cells or MSC-1 cells were transplanted as allografts into the renal subcapsular area of naive BALB/c mice, and their survival in graft sites was compared. While Sertoli cells were detected within the grafts with 100% graft survival throughout the 20-day study, MSC-1 cells were rejected between 11 and 14 days, with 0% graft survival at 20 days posttransplantation. Nonetheless, the mechanism for primary Sertoli cell survival and immunoprotection remains unresolved. To identify immune factors or functional pathways potentially responsible for immune privilege, gene expression profiles of enriched primary Sertoli cells were compared with those of MSC-1 cells. Microarray analysis identified 2369 genes in enriched primary Sertoli cells that were differentially expressed at ±4-fold or higher levels than in MSC-1 cells. Ontological analyses identified multiple immune pathways, which were used to generate a list of 340 immune-related genes. Three functions were identified in primary Sertoli cells as potentially important for establishing immune privilege: suppression of inflammation by specific cytokines and prostanoid molecules, slowing of leukocyte migration by controlled cell junctions and actin polymerization, and inhibition of complement activation and membrane-associated cell lysis. These results increase our understanding of testicular immune privilege and, in the long-term, could lead to improvements in transplantation success.


Subject(s)
Sertoli Cells/immunology , Sertoli Cells/transplantation , Adherens Junctions , Animals , Apoptosis , Cell Adhesion , Cell Line , Gene Expression Profiling , Gene Expression Regulation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tight Junctions
15.
Adv Exp Med Biol ; 763: 237-59, 2012.
Article in English | MEDLINE | ID: mdl-23397628

ABSTRACT

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.


Subject(s)
Blood-Testis Barrier/immunology , Epididymis/immunology , Testis/immunology , Animals , Blood-Testis Barrier/anatomy & histology , Blood-Testis Barrier/physiology , Cell Differentiation , Cell Membrane Permeability , Epididymis/anatomy & histology , Epididymis/physiology , Graft Survival/immunology , Humans , Immune Tolerance , Male , Phylogeny , Seminiferous Epithelium/immunology , Seminiferous Epithelium/physiology , Sertoli Cells/immunology , Sertoli Cells/physiology , Sertoli Cells/ultrastructure , Sperm Maturation , Spermatogenesis , Spermatozoa/immunology , Spermatozoa/physiology , Testis/anatomy & histology , Testis/physiology , Tight Junctions/immunology , Tight Junctions/physiology , Tight Junctions/ultrastructure , Transplantation Immunology
16.
Viruses ; 14(2)2022 02 11.
Article in English | MEDLINE | ID: mdl-35215967

ABSTRACT

Zika virus (ZIKV), a re-emerging virus, causes congenital brain abnormalities and Guillain-Barré syndrome. It is mainly transmitted by Aedes mosquitoes, but infections are also linked to sexual transmissions. Infectious ZIKV has been isolated, and viral RNA has been detected in semen over a year after the onset of initial symptoms, but the mode of long-term persistence is not yet understood. ZIKV can proliferate in human Sertoli cells (HSerC) for several weeks in vitro, suggesting that it might be a reservoir for persistent ZIKV infection. This study determined proteomic changes in HSerC during ZIKV infections by TMT-mass spectrometry analysis. Levels of 4416 unique Sertoli cell proteins were significantly altered at 3, 5, and 7 days after ZIKV infection. The significantly altered proteins include enzymes, transcription regulators, transporters, kinases, peptidases, transmembrane receptors, cytokines, ion channels, and growth factors. Many of these proteins are involved in pathways associated with antiviral response, antigen presentation, and immune cell activation. Several immune response pathway proteins were significantly activated during infection, e.g., interferon signaling, T cell receptor signaling, IL-8 signaling, and Th1 signaling. The altered protein levels were linked to predicted activation of immune response in HSerC, which was predicted to suppress ZIKV infection. ZIKV infection also affected the levels of critical regulators of gluconeogenesis and glycolysis pathways such as phosphoglycerate mutase, phosphoglycerate kinase, and enolase. Interestingly, many significantly altered proteins were associated with cardiac hypertrophy, which may induce heart failure in infected patients. In summary, our research contributes to a better understanding of ZIKV replication dynamics and infection in Sertoli cells.


Subject(s)
Semen/virology , Sertoli Cells/immunology , Virus Replication , Zika Virus Infection/immunology , Carbohydrate Metabolism/immunology , Cardiovascular Diseases/immunology , Disease Transmission, Infectious , Humans , Male , Protein Processing, Post-Translational , Proteomics , RNA, Viral/genetics , Sertoli Cells/virology , Zika Virus/isolation & purification , Zika Virus Infection/transmission
17.
Biochem Biophys Res Commun ; 404(3): 829-33, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21172306

ABSTRACT

The major problem of the application of endothelial cells (ECs) in transplantation is the lack of proliferation and their immunogenicity. In this study, we co-cultured ECs with Sertoli cells to monitor whether Sertoli cells can influence the proliferation and immunogenicity of co-cultured ECs. Sertoli cells were isolated from adult testicular tissue. ECs were divided into the control group and the experimental group, which included three sub-groups co-cultured with 1 × 10(3), 1 × 10(4) or 1 × 10(5) cell/ml of Sertoli cells. The growth and proliferation of ECs were observed microscopically, and the expression of vascular endothelial growth factor (VEGF) receptor-2 (KDR) was examined by Western blotting. In another experiment, ECs were divided into the control group, the single culture group and the co-culture group with the optimal concentration of Sertoli cells. After INF-γ and TNF-α were added to the culture medium, MHC II antigen expression was detected by immunofluorescence staining and western blotting; interleukin (IL)-6, IL-8 and soluble intercellular adhesion molecule (sICAM) were measured in the culture medium by ELISA. We demonstrated that 1 × 10(4) cell/ml Sertoli cells promoted the proliferation of co-cultured ECs more dramatically than that in other groups (P<0.05). Western blotting showed that 1 × 10(4) cell/ml of the Sertoli cells was most effective in the up-regulation of KDR expression in the co-cultured ECs (P<0.05). Sertoli cells can effectively suppress INF-γ-induced MHC II antigen expression in co-cultured ECs compared with single culture group (P<0.05). TNF-α induced the expression of IL-6, IL-8 and sICAM in ECs. When co-cultured with Sertoli cells, their expressions were significantly lower than in the EC single culture group (P<0.05). ECs co-cultured with Sertoli cells also did not significantly increase the stimulation index of spleen lymphocytes compared to the single culture group (P<0.05). Our results suggested that co-culturing with Sertoli cells can significantly promote the proliferation of ECs, accelerate post-transplant angiogenesis, while reduce EC immunogenicity and stimulus to lymphocytes.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/immunology , Sertoli Cells/immunology , Adult , Biomarkers/analysis , Cell Proliferation , Coculture Techniques/methods , Endothelial Cells/transplantation , Histocompatibility Antigens Class II/immunology , Humans , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Interleukin-8/metabolism , Male , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor Receptor-2/analysis
18.
Clin Exp Immunol ; 165(1): 104-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21501149

ABSTRACT

Pig xenografts represent an alternative source of organs for transplantation. Immunosuppression can prevent rejection, but involves high risk and cost. New anti-rejection techniques have been developed; however, results have not been successful. Few studies have reported xenoantibody levels in xenotransplanted patients with diabetes and no patients have reported a clinical correlation. We analysed anti-galactose (Gal) and other anti-pig antibody (APA) levels in xenotransplanted patients with type 1 diabetes and the relation to the clinical outcome. Twenty-three patients with type 1 diabetes were transplanted with porcine islets inside a device without immunosuppression. Demographic characteristics, insulin dose and xenoantibody levels at different periods were recorded. Anti-Gal and anti-pig antibodies were measured through indirect enzyme-linked immunosorbent assay (ELISA) and haemolytic anti-pig antibody assay. More than 50% were female; the mean current age, current diabetes duration, diabetes duration at xenotransplantation and time post-transplantation were: 20·8, 11, 5·5 and 5·7 years, respectively. Insulin doses remained with a mean reduction greater than 33% in more than 50% of the patients. The lowest anti-Gal antibody levels were related to the highest insulin dose reductions. This relationship could be explained by the device, Sertoli cells and accommodation process.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Galactose/metabolism , Insulin/metabolism , Islets of Langerhans Transplantation , Adolescent , Animals , Animals, Newborn , Antibodies, Heterophile/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/physiopathology , Equipment and Supplies , Female , Follow-Up Studies , Galactose/immunology , Graft Survival/immunology , Humans , Immune Tolerance , Islets of Langerhans Transplantation/instrumentation , Islets of Langerhans Transplantation/methods , Male , Recovery of Function , Sertoli Cells/immunology , Sertoli Cells/metabolism , Sertoli Cells/pathology , Swine , Transplantation, Heterologous , Young Adult
19.
Microsc Microanal ; 17(6): 1002-14, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22047748

ABSTRACT

Transplantation of spermatogonial stem cells (SSCs), the male germline stem cells, in experimental animal models has been successfully used to study mechanisms involved in SSC self-renewal and to restore fertility. However, there are still many challenges associated with understanding the recipient immune response for SSCs use in clinical therapies. Here, we have undertaken a detailed structural study of macrophages elicited by SSCs transplantation in mice using both high-resolution light microscopy (HRLM) and transmission electron microscopy (TEM). We demonstrate that SSCs transplantation elicits a rapid and potent recruitment of macrophages into the seminiferous epithelium (SE). Infiltrating macrophages were derived from differentiation of peritubular monocyte-like cells into typical activated macrophages, which actively migrate through the SE, accumulate in the tubule lumen, and direct phagocytosis of differentiating germ cells and spermatozoa. Quantitative TEM analyses revealed increased formation of lipid bodies (LBs), organelles recognized as intracellular platforms for synthesis of inflammatory mediators and key markers of macrophage activation, within both infiltrating macrophages and Sertoli cells. LBs significantly increased in number and size in parallel to the augmented macrophage migration during different times post-transplantation. Our findings suggest that LBs may be involved with immunomodulatory mechanisms regulating the seminiferous tubule niche after SSC transplantation.


Subject(s)
Microscopy, Electron, Transmission/methods , Seminiferous Epithelium/ultrastructure , Seminiferous Tubules/ultrastructure , Sertoli Cells/ultrastructure , Spermatogonia/ultrastructure , Stem Cell Transplantation/methods , Stem Cells/immunology , Animals , Cell Count , Cell Differentiation , Cell Movement , Cell Proliferation , Cytokines/biosynthesis , Macrophages/cytology , Macrophages/immunology , Macrophages/ultrastructure , Male , Mice , Monocytes/cytology , Monocytes/immunology , Monocytes/ultrastructure , Organelles/immunology , Organelles/ultrastructure , Phagocytosis/immunology , Seminiferous Epithelium/immunology , Seminiferous Tubules/immunology , Sertoli Cells/immunology , Spermatogenesis , Spermatogonia/cytology , Spermatogonia/immunology , Spermatogonia/transplantation , Stem Cells/cytology , Time Factors
20.
Front Immunol ; 12: 582858, 2021.
Article in English | MEDLINE | ID: mdl-33679734

ABSTRACT

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.


Subject(s)
Blood-Testis Barrier/immunology , Escherichia coli Infections/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Animals , Blood-Testis Barrier/metabolism , Cells, Cultured , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Orchitis/immunology , Orchitis/metabolism , Orchitis/microbiology , Rats, Sprague-Dawley , Sertoli Cells/immunology , Sertoli Cells/metabolism , Sertoli Cells/microbiology , Spermatogenesis/immunology , Testis/immunology , Testis/metabolism , Tight Junction Proteins/immunology , Tight Junction Proteins/metabolism , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/physiology
SELECTION OF CITATIONS
SEARCH DETAIL