Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Eur J Pharmacol ; 938: 175430, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36460131

ABSTRACT

Disruption of blood-testis barrier (BTB) was a crucial pathological feature of diabetes induced-testicular injury at early phase. Aucubin (AU), a main active component in Eucommiae Cortex, has drawn attention for its benefits against male reproductive system disease. The current study was aimed at investigating the protective role of AU and exploring the underlying mechanism in diabetic model. A murine model of type 2 diabetes mellitus (T2DM) was induced by high-fat diet (HFD) combined with streptozocin (STZ). Testicular weight index and morphology, sperm quality, integrity of BTB and protein levels were analyzed. The underlying mechanism of the protective effect of AU was further explored in Sertoli cells (SCs) cultured with high glucose (HG). Our results showed AU inhibited testicular structural destruction, restored disruption of BTB and improved abnormal spermatogenic function in diabetic mice. Consistent with in vivo results, HG induced decreased transcellular resistance and increased permeability in SCs monolayers, while AU exposure reverses this trend. Meanwhile, reduced expression of Zonula occludin-1(ZO-1) and Connexin43(Cx43) in testicular tissue diabetic mice and HG-induced SCs was prominently reversed via AU treatment. Mechanistic studies suggested a high affinity interaction between AU and c-Src protein was identified based on molecular docking, and the activation of c-Src was significantly inhibited in AU treatment. Furthermore, AU significantly increased the expression of Cx43 and ZO-1 proteins HG-induced SCs, which can be further enhanced in gene-silenced c-Src cells to some extent. Our results suggested that AU ameliorated disruption of BTB and spermatogenesis dysfunction in diabetic mice via inactivating c-Src to stabilize cell junction integrity.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Male , Mice , Animals , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/pathology , Connexin 43/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Molecular Docking Simulation , Semen/metabolism , Testis , Sertoli Cells/metabolism , Intercellular Junctions/metabolism , Dietary Supplements
2.
Inflamm Res ; 70(10-12): 1165-1175, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34436630

ABSTRACT

OBJECTIVE: Junctional proteins are the most important component of the blood-testis barrier and maintaining the integrity of this barrier is essential for spermatogenesis and male fertility. The present study elucidated the effect of SARS-CoV-2 infection on the blood-testis barrier (BTB) in patients who died from severe acute respiratory syndrome coronavirus 2 (COVID-19) complications. METHODS: In this study, lung and testis tissue was collected from autopsies of COVID-19 positive (n = 10) and negative men (n = 10) and was taken for stereology, immunocytochemistry, and RNA extraction. RESULTS: Evaluation of the lung tissue showed that the SARS-CoV-2 infection caused extensive damage to the lung tissue and also increases inflammation in testicular tissue and destruction of the testicular blood barrier. Autopsied testicular specimens of COVID-19 showed that COVID-19 infection significantly changes the spatial arrangement of testicular cells and notably decreased the number of Sertoli cells. Moreover, the immunohistochemistry results showed a significant reduction in the protein expression of occluding, claudin-11, and connexin-43 in the COVID-19 group. In addition, we also observed a remarkable enhancement in protein expression of CD68 in the testes of the COVID-19 group in comparison with the control group. Furthermore, the result showed that the expression of TNF-α, IL1ß, and IL6 was significantly increased in COVID-19 cases as well as the expression of occludin, claudin-11, and connexin-43 was decreased in COVID-19 cases. CONCLUSIONS: Overall, the present study demonstrated that SARS-CoV-2 could induce the up-regulation of the pro-inflammatory cytokine and down-regulation of junctional proteins of the BTB, which can disrupt BTB and ultimately impair spermatogenesis.


Subject(s)
Blood-Testis Barrier/pathology , COVID-19/pathology , Cytokines/metabolism , Autopsy , Claudins/metabolism , Connexin 43/metabolism , Humans , Immunohistochemistry , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lung/pathology , Male , Middle Aged , Occludin/metabolism , RNA, Viral/analysis , Sertoli Cells/pathology , Testis/pathology , Tumor Necrosis Factor-alpha/metabolism
3.
Biosci Rep ; 41(6)2021 06 25.
Article in English | MEDLINE | ID: mdl-34060622

ABSTRACT

To facilitate temperature adjustments, the testicles are located outside the body cavity. In most mammals, the temperature of the testes is lower than the body temperature to ensure the normal progression of spermatogenesis. Rising temperatures affect spermatogenesis and eventually lead to a decline in male fertility or even infertility. However, the testes are composed of different cell types, including spermatogonial stem cells (SSCs), spermatocytes, spermatozoa, Leydig cells, and Sertoli cells, which have different cellular responses to heat stress. Recent studies have shown that using different drugs can relieve heat stress-induced reproductive damage by regulating different signaling pathways. Here, we review the mechanisms by which heat stress damages different cells in testes and possible treatments.


Subject(s)
Fertility , Heat-Shock Proteins/metabolism , Heat-Shock Response , Hot Temperature/adverse effects , Infertility, Male/metabolism , Testis/metabolism , Animals , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/pathology , Fertility/drug effects , Fertility Agents, Male/therapeutic use , Heat-Shock Response/drug effects , Humans , Infertility, Male/drug therapy , Infertility, Male/pathology , Infertility, Male/physiopathology , Leydig Cells/metabolism , Leydig Cells/pathology , Male , Risk Factors , Sertoli Cells/metabolism , Sertoli Cells/pathology , Signal Transduction , Spermatocytes/metabolism , Spermatocytes/pathology , Spermatogonia/metabolism , Spermatogonia/pathology , Testis/drug effects , Testis/pathology , Testis/physiopathology
4.
Gene ; 780: 145531, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33631249

ABSTRACT

Sertoli cells are "nurturing cells'' in the seminiferous tubules of the testis which have essential roles in the development, proliferation and differentiation of germ cells. These cells also divide the seminiferous epithelium into a basal and an adluminal compartment and establish the blood-testis barrier (BTB). BTB shields haploid germ cells from recognition by the innate immune system. Moreover, after translocation of germ cells into the adluminal compartment their nutritional source is separated from the circulatory system being only supplied by the Sertoli cells. The integrity of BTB is influenced by several organic/ organometallic, hormonal and inflammatory substances. Moreover, several environmental contaminants such as BPA have hazardous effects on the integrity of BTB. In the current review, we summarize the results of studies that assessed the impact of these agents on the integrity of BTB. These studies have implications in understanding the molecular mechanism of male infertility and also in the male contraception.


Subject(s)
Blood-Testis Barrier/metabolism , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Spermatogenesis/drug effects , Animals , Blood-Testis Barrier/pathology , Humans , Male , Seminiferous Epithelium/pathology , Sertoli Cells/pathology
5.
Development ; 148(1)2021 01 13.
Article in English | MEDLINE | ID: mdl-33441379

ABSTRACT

Cryptorchidism is the most common urologic birth defect in men and is a predisposing factor of male infertility and testicular cancer, yet the etiology remains largely unknown. E2F1 microdeletions and microduplications contribute to cryptorchidism, infertility and testicular tumors. Although E2f1 deletion or overexpression in mice causes spermatogenic failure, the mechanism by which E2f1 influences testicular function is unknown. This investigation revealed that E2f1-null mice develop cryptorchidism with severe gubernacular defects and progressive loss of germ cells resulting in infertility and, in rare cases, testicular tumors. It was hypothesized that germ cell depletion resulted from an increase in WNT4 levels. To test this hypothesis, the phenotype of a double-null mouse model lacking both Wnt4 and E2f1 in germ cells was analyzed. Double-null mice are fertile. This finding indicates that germ cell maintenance is dependent on E2f1 repression of Wnt4, supporting a role for Wnt4 in germ cell survival. In the future, modulation of WNT4 expression in men with cryptorchidism and spermatogenic failure due to E2F1 copy number variations may provide a novel approach to improve their spermatogenesis and perhaps their fertility potential after orchidopexy.


Subject(s)
E2F1 Transcription Factor/metabolism , Spermatogenesis , Testis/metabolism , Wnt4 Protein/metabolism , Aging/pathology , Animals , Animals, Newborn , Blood-Testis Barrier/pathology , Cell Cycle/genetics , Cryptorchidism/genetics , Cryptorchidism/pathology , E2F1 Transcription Factor/deficiency , Fertility , Gene Expression Regulation , Male , Mice, Inbred C57BL , Models, Biological , Signal Transduction , Spermatozoa/metabolism , Testis/pathology
6.
Article in English | MEDLINE | ID: mdl-33006953

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS- CoV-2) that causes COVID-19 infections penetrates body cells by binding to angiotensin-converting enzyme-2 (ACE2) receptors. Evidence shows that SARS-CoV-2 can also affect the urogenital tract. Hence, it should be given serious attention when treating COVID-19-infected male patients of reproductive age group. Other viruses like HIV, mumps, papilloma and Epstein-Barr can induce viral orchitis, germ cell apoptosis, inflammation and germ cell destruction with attending infertility and tumors. The blood-testis barrier (BTB) and blood-epididymis barrier (BEB) are essential physical barricades in the male reproductive tract located between the blood vessel and seminiferous tubules in the testes. Despite the significant role of these barriers in male reproductive function, studies have shown that a wide range of viruses can still penetrate the barriers and induce testicular dysfunctions. Therefore, this mini-review highlights the role of ACE2 receptors in promoting SARS-CoV-2-induced blood-testis/epididymal barrier infiltration and testicular dysfunction.


Subject(s)
Blood-Testis Barrier/enzymology , Blood-Testis Barrier/pathology , Coronavirus Infections/enzymology , Coronavirus Infections/pathology , Infertility, Male/etiology , Infertility, Male/pathology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/enzymology , Pneumonia, Viral/pathology , Angiotensin-Converting Enzyme 2 , COVID-19 , Humans , Infertility, Male/enzymology , Male , Pandemics , Testis/metabolism
7.
Toxicology ; 438: 152463, 2020 05 30.
Article in English | MEDLINE | ID: mdl-32294493

ABSTRACT

Methyl parathion (Me-Pa) is an extremely toxic organophosphorus pesticide still used in developing countries. It has been associated with decreased sperm function and fertility and with oxidative and DNA damage. The blood-testis barrier (BTB) is a structure formed by tight junction (TJ) proteins in Sertoli cells and has a critical role in spermatogenesis. We assessed the effect of repeated doses of Me-Pa (3-12 mg/kg/day for 5 days, i.p.) on sperm quality, lipid oxidation, DNA integrity, and BTB permeability in adult male mice and explored oxidation as a mechanism of toxicity. Me-Pa caused dose-dependent effects on sperm quality, lipoperoxidation, and DNA integrity. Testis histology results showed the disruption of spermatogenesis progression and atrophy of seminiferous tubules. The pesticide opened the BTB, as evidenced by the presence of a biotin tracer in the adluminal compartment of the seminiferous tubules. This effect was not observed after 45 days of exposure when a spermatogenic cycle had completed. The coadministration of the antioxidant α-tocopherol (50 mg/kg/day for 5 days, oral) prevented the effects of Me-Pa on sperm quality, DNA and the BTB, indicating the importance of oxidative stress in the damage generated by Me-Pa. As evidenced by immunochemistry, no changes were found in the localization of the TJ proteins of the BTB, although oxidation (carbonylation) of total proteins in testis homogenates was detected. Our results show that Me-Pa disturbs the BTB and that oxidation is involved in the observed toxic effects on sperm cells.


Subject(s)
Blood-Testis Barrier/drug effects , Capillary Permeability/drug effects , Cholinesterase Inhibitors/toxicity , DNA Damage , Methyl Parathion/toxicity , Oxidative Stress/drug effects , Pesticides/toxicity , Spermatozoa/drug effects , Acetylcholinesterase/metabolism , Animals , Antioxidants/pharmacology , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/pathology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Lipid Peroxidation/drug effects , Male , Mice, Inbred ICR , Protein Carbonylation/drug effects , Spermatogenesis/drug effects , Spermatozoa/metabolism , Spermatozoa/pathology
8.
Andrology ; 8(5): 1398-1408, 2020 09.
Article in English | MEDLINE | ID: mdl-32196966

ABSTRACT

BACKGROUND: Cryptorchidism is known to impair spermatogenesis. The blood-testis barrier (BTB) becomes defined in seminiferous tubules around puberty and provides a suitable environment for germ cells. Little is known about the BTB in undescended testes (UDT). OBJECTIVES: To determine the role of BTB during puberty in UDT using a non-surgical cryptorchid rat model. MATERIAL AND METHODS: Unilateral cryptorchid male rats were intraperitoneally injected with non-steroidal antiandrogen during intrauterine development; the testes were harvested at 4, 5, and 6 weeks after birth. Testicular histology, expression levels of the BTB proteins (claudin-11, occludin, zonula occludens-1), and apoptotic cells were evaluated by immunohistochemistry, Western blotting, and TUNEL assay. The functionality of the BTB was investigated by electron microscopy using the lanthanum tracer method. RESULTS: The testicular histology of undescended testes 6 weeks after birth showed maturation arrest at the spermatocyte level. The BTB protein distributions were altered in the UDT, with a noticeable difference in claudin-11(CLDN11) localization from 4 to 5 weeks after birth between control and UDT samples. BTB protein levels were similar. More apoptotic germ cells were detected in the adluminal compartment of tubules in the UDT than in the control testes. Electron microscopy showed that the lanthanum tracer was limited to the BTB of control testes, whereas it penetrated the BTB of UDT. DISCUSSION: Here, loss of normal BTB function and impaired spermatogenesis were observed in UDT during puberty. CLDN11 is a pivotal tight junction protein belonging to the BTB. Tight junctions are considered as essential for normal spermatogenesis, and abnormal CLDN11 organization may cause UDT-associated male infertility. CONCLUSION: CLDN11 disorganization within the BTB may cause spermatogenic impairment, possibly by limiting the BTB function.


Subject(s)
Blood-Testis Barrier/pathology , Claudins/metabolism , Cryptorchidism/pathology , Cryptorchidism/physiopathology , Sexual Maturation/physiology , Animals , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/physiopathology , Cryptorchidism/metabolism , Male , Rats , Rats, Sprague-Dawley , Spermatogenesis/physiology , Tight Junctions/metabolism , Tight Junctions/pathology
9.
Andrologia ; 52(3): e13508, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31957918

ABSTRACT

Testicular injury is the primary pathogenesis of diabetes-induced male infertility. Dioscorea zingiberensis (DZ), a traditional Chinese medicine (TCM) including saponins, flavonoids and cellulose, is used to treat diseases in the reproductive system. But the protective effects of DZ on diabetes-induced testicular injury remain poorly understood. In this study, the therapeutic effects of chronic oral DZ treatment on testis impairment in a diabetic mouse model were explored by assessing sperm morphology, blood-testes barrier (BTB) integrity and testicular histological examination. Our results showed that DZ significantly reversed BTB disruption, testicular tissue injury and abnormal sperm morphology in diabetic mice. Interestingly, diabetes-induced disruption of the BTB was associated with a decrease in the tight junction (TJ) protein zonula occludens-1 (ZO-1). Dioscorea zingiberensis effectively increased ZO-1 expression in testis tissue to restore the integrity of the BTB. Moreover, DZ treatment significantly reduced hyperglycaemia-induced increases in malondialdehyde (MDA) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) levels. Further mechanistic studies revealed that DZ substantially enhanced the expression of Nrf2, NOQ1 and HO-1, which indicated that DZ exerts potential antioxidant effects against testicular tissue damage via the activation of Nrf2. In conclusion, the protective effects of DZ rely on repairing the integrity of the BTB and on reducing oxidative stress damage by mediating ZO-1 and Nrf2. The study contributes to discovering the DZ possible mechanism of action.


Subject(s)
Blood-Testis Barrier/drug effects , Diabetes Mellitus, Experimental/complications , Dioscorea/chemistry , Infertility, Male/prevention & control , Plant Extracts/pharmacology , Animals , Blood-Testis Barrier/pathology , Diabetes Mellitus, Experimental/chemically induced , Diet, High-Fat/adverse effects , Disease Models, Animal , Ethanol/chemistry , Humans , Infertility, Male/etiology , Male , Mice , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Streptozocin/toxicity , Tight Junctions , Up-Regulation/drug effects , Zonula Occludens-1 Protein/metabolism
10.
Reprod Sci ; 27(7): 1443-1454, 2020 07.
Article in English | MEDLINE | ID: mdl-31994000

ABSTRACT

Sertoli cells are important for spermatogenesis not only by directly interacting with germ line cells in the seminiferous epithelium but also by constituting the blood-testis barrier (BTB) structure to create a favorable environment for spermatogenesis. Blind sterile (bs) male mice are infertile, with excessive germ cell apoptosis and spermatogenesis arrest. TBC1D20 (TBC1 domain family member 20) deficiency has been identified as the causative mutation in bs mice. However, whether TBC1D20 loss of function also impairs BTB integrity, which further contributes to the failed spermatogenesis of bs male mice, remains unclear. In the present study, biotin tracer assay and transmission electron microscopy showed severely disrupted BTB integrity in bs testes. Compared to the wild-type Sertoli cells, BTB components of cultured bs Sertoli cells in vitro was perturbed with downregulation of E-cadherin, ZO-1, ß-catenin, and Claudin 11. The obvious rearrangement of F-actin indicated disrupted epithelial-mesenchymal balance in TBC1D20-deficient Sertoli cells. The ability of bs Sertoli cells to maintain the clone formation of spermatogonia stem cells was also obviously limited. Furthermore, the decreasing of SOX9 (sex-determining region Y box 9) and WT1 (Wilms' tumor 1) and increasing of vimentin in bs Sertoli cells indicated that TBC1D20 loss of function attenuated the differentiation progression of bs Sertoli cells. In summary, TBC1D20 loss of function impedes the maturation of adult Sertoli cells and resulted in impaired BTB integrity, which is further implicated in the infertile phenotype of bs male mice.


Subject(s)
Blood-Testis Barrier/metabolism , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , rab1 GTP-Binding Proteins/drug effects , Animals , Blood-Testis Barrier/pathology , Cells, Cultured , Coculture Techniques , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice , Mice, Transgenic , Seminiferous Epithelium/pathology , Sertoli Cells/pathology , Testis/metabolism , Testis/pathology , rab1 GTP-Binding Proteins/genetics
11.
Sci Rep ; 9(1): 15221, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31645625

ABSTRACT

Radiation-induced abscopal effect (RIAE) may influence radiotherapy efficiency. However, it is unknown whether RIAE triggers abnormal genetic consequence. We present a novel evidence that, when mice were given fractionated irradiation on right thorax, the ultrastructure of blood-testis barrier was damaged in company with apoptosis induction in testes, and the sperm number and vitality were drastically decreased so that both the fertility and the survival of their offspring were reduced. Protein microarray assay and hormone detection showed that some cytokines especially TNF-α, TGF-ß and estradiol in the serum of irradiated mice increased to higher levels in consistent with abscopal damage, and this conditioned serum had toxic effect on TM4 cells in vitro. When the mice were fed with cimetidine, the above abscopal responses were significantly attenuated. This study demonstrates in the first time that the thoracic irradiation (Th-IR) induces structural and functional damage in the distal testes and further cause fertility decline of irradiated male mice, which may have important implications in the strategy development of radiotherapy in avoiding abnormal genetic consequence.


Subject(s)
Blood-Testis Barrier/pathology , Infertility, Male/etiology , Radiotherapy/adverse effects , Testis/pathology , Thorax/radiation effects , Animals , Apoptosis , Cytokines/blood , Dose Fractionation, Radiation , Fertility , Infertility, Male/pathology , Male , Mice , Mice, Inbred C57BL , Sperm Count
12.
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
13.
FASEB J ; 33(11): 12528-12540, 2019 11.
Article in English | MEDLINE | ID: mdl-31450968

ABSTRACT

Mumps virus (MuV) has high tropism to the testis and may lead to male infertility. Sertoli cells are the major targets of MuV infection. However, the mechanisms by which MuV infection impairs male fertility and Sertoli cell function remain unclear. The present study elucidated the effect of MuV infection on the blood-testis barrier (BTB). The transepithelial electrical resistance of MuV-infected mouse Sertoli cells was monitored, and the expression of major proteins of the BTB was examined. We demonstrated that MuV infection disrupted the BTB by reducing the levels of occludin and zonula occludens 1. Sertoli cells derived from Tlr2-/- and Tnfa-/- mice were analyzed for mediating MuV-induced impairment. TLR2-mediated TNF-α production by Sertoli cells in response to MuV infection impaired BTB integrity. MuV-impaired BTB was not observed in Tlr2-/- and Tnfa-/- Sertoli cells. Moreover, an inhibitor of TNF-α, pomalidomide, prevents the disruption of BTB in response to MuV infection. FITC-labeled biotin tracing assay confirmed that BTB permeability and spermatogenesis were transiently impaired by MuV infection in vivo. These findings suggest that the disruption of the BTB could be one of the mechanisms underlying MuV-impaired male fertility, in which TNF-α could play a critical role.-Wu, H., Jiang, X., Gao, Y., Liu, W., Wang, F., Gong, M., Chen, R., Yu, X., Zhang, W., Gao, B., Song, C., Han, D. Mumps virus infection disrupts blood-testis barrier through the induction of TNF-α in Sertoli cells.


Subject(s)
Blood-Testis Barrier/metabolism , Mumps virus/metabolism , Mumps/metabolism , Sertoli Cells/metabolism , Spermatogenesis , Tumor Necrosis Factor-alpha/metabolism , Animals , Blood-Testis Barrier/pathology , Blood-Testis Barrier/virology , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Infertility, Male/virology , Male , Mice , Mice, Knockout , Mumps/genetics , Mumps/pathology , Mumps virus/genetics , Sertoli Cells/pathology , Sertoli Cells/virology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
14.
Andrologia ; 51(6): e13285, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31006889

ABSTRACT

The present study was designed to investigate the therapeutic effect of bone marrow MSC-derived factors on gonadotropic toxicity induced by busulfan in vivo. The conditioned media (CM) was obtained from MSCs in serum-free incubation for 48 hr and concentrated ~25-fold by ultrafiltration. The CM of HEK 293 cells was treated as control (293-CM). MSC-CM was injected into busulfan mice via caudal veins after 1 day of busulfan treatment for 2 weeks (200 µl per dose/twice weekly). Compared to the 293-CM group, testicular injury was delayed in MSC-CM group, including reduced vacuolations of cells in the basal compartment of the seminiferous epithelium and detachment of cells from basement membrane. Apoptotic spermatogenic cells were significantly decreased in MSC-CM group (p ï¼œ 0.05). Interesting N-cadherin,ICAM-1 and P-cadherin expressions significantly increased in MSC-CM group, while occludin, ZO-1 and connexin 43 expressions showed no difference among MSC-CM, 293-CM and busulfan groups. Present results suggest MSC-secreted factors protect spermatogenesis impairment after busulfan treatment by reducing the apoptosis of spermatogenic cells and enhancing intercellular adhesion molecule expressions.


Subject(s)
Blood-Testis Barrier/drug effects , Busulfan/toxicity , Culture Media, Conditioned/pharmacology , Infertility, Male/drug therapy , Mesenchymal Stem Cells/metabolism , Animals , Apoptosis/drug effects , Blood-Testis Barrier/cytology , Blood-Testis Barrier/pathology , Cadherins/metabolism , Cell Adhesion/drug effects , Culture Media, Conditioned/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Infertility, Male/chemically induced , Infertility, Male/pathology , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Spermatogenesis/drug effects
15.
Contraception ; 99(6): 350-356, 2019 06.
Article in English | MEDLINE | ID: mdl-30763581

ABSTRACT

OBJECTIVE: The bioavailability of the non-hormonal male contraceptive adjudin is low in rats due to the blood-testis barrier (BTB). This study was designed to examine if F5-peptide, an endogenously produced reversible BTB modifier, could enhance the bioavailability of adjudin to affect spermatogenesis and provide a contraceptive effect in rats while reducing systemic toxicity. STUDY DESIGN: We overexpressed F5-peptide in adult male rats (n=10 rats; with 3 or 4 rats for each of the three different experiments noted in the three regimens) by intratesticular injection of a mammalian expression vector pCI-neo (pCI-neo/F5-peptide) vs. empty vector alone (pCI-neo/Ctrl) to be followed by treatment with adjudin by oral gavage at a dose of 10 or 20 mg/kg. The status of spermatogenesis was assessed by histological analysis and dual-labeled immunofluorescence analysis on Day 16. To assess fertility, we allowed treated males (n=3-4 rats) to mate with mature female rats (n=3-4) individually, and assessed the number of pups on Days 23, 36 and 82 to assess fertility and reversibility. RESULTS: All 4 treated rats overexpressed with F5-peptide and low-dose adjudin were infertile by Day 36, and half of these rats were fertile by Day 82, illustrating reversibility. However, overexpression of F5-peptide alone (or low-dose adjudin alone) had no effects on fertility in n=3 rats. These findings were consistent with the histology data that illustrated the BTB modifier F5-peptide promoted the action of adjudin to induce germ cell exfoliation, mediated by changes in cytoskeletal organization of F-actin and microtubules across the epithelium, thereby reducing the systemic toxicity of adjudin. CONCLUSION: In this proof-of-concept study, it was shown that overexpression of the F5-peptide prior to administration of adjudin to rats at a low (and ineffective dose by itself) was found to induce reversible male infertility. IMPLICATIONS: Overexpression of F5-peptide, an endogenously produced biomolecule in the testis known to induce BTB remodeling, enhanced the contraceptive effect of adjudin in rats, supporting proof of concept studies of BTB disrupters in men.


Subject(s)
Blood-Testis Barrier/metabolism , Hydrazines/pharmacology , Indazoles/pharmacology , Microtubules/metabolism , Peptide Fragments/metabolism , Sertoli Cells/metabolism , Spermatogenesis , Animals , Blood-Testis Barrier/pathology , Female , Laminin/genetics , Laminin/metabolism , Male , Microtubules/pathology , Peptide Fragments/genetics , Proof of Concept Study , Rats , Rats, Sprague-Dawley , Sertoli Cells/pathology , Transfection
16.
Histol Histopathol ; 34(5): 525-535, 2019 May.
Article in English | MEDLINE | ID: mdl-30403279

ABSTRACT

Impairment of blood-testis barrier integrity can be observed during inflammation, infection, trauma and experimental autoimmune orchitis, which is inducible in rodents. In the present study, an initially fertile two-year-old Beagle dog was presented with a decline in total sperm number resulting in azoospermia within five months, verified by twice-monthly semen analyses. The dog was clinically healthy with bilateral small testes and showed normal thyroid function. Bacterial cultures of semen were negative and serum biochemical analyses showed no abnormal findings. To determine causes of azoospermia, the dog was castrated. Histological examinations of hematoxylin-eosin stained testicular sections revealed impaired spermatogenesis, seminiferous tubules with spermatogenic arrest or Sertoli-cell-only syndrome as well as focal interstitial and even intratubular lymphocytic infiltrations. Germ cell sloughing, apoptosis and giant cells were also observed in some tubules. Subsequent immunostainings of smooth-muscle-actin, claudin3, claudin11 and connexin43 demonstrated, for the first time, a mechanical and functional disruption of the tubular wall and alterations of blood-testis barrier proteins in these tubules. Presence of claudin3 and claudin11 in canine testis was confirmed using RT-PCR and sequencing and/ or Western-blot analyses. All findings suggested a possible spontaneous autoimmune orchitis to be the underlying cause for the observed azoospermia.


Subject(s)
Autoimmune Diseases/veterinary , Dog Diseases/immunology , Dog Diseases/pathology , Orchitis/veterinary , Animals , Blood-Testis Barrier/pathology , Dogs , Male
17.
Toxicol Lett ; 295: 277-287, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-29981920

ABSTRACT

There are reports of fluorochloridone (FLC)-induced male reproductive toxicity, but the underlying toxicological mechanisms remain unknown. In this study, we looked at how FLC exposure affected the integrity of the blood-testis barrier (BTB) and the Sertoli cell barrier and studied the molecular mechanisms. Male rats received gavage administration of FLC (30 mg/kg/d) for 14 consecutive days with sample collection at the 7th and 14th day; and primary cultured Sertoli cells were treated with 0-10 µM FLC in vitro for 24 h. Our in vivo findings revealed that FLC exposure caused time-dependent testicular injuries, sperm quality decrease as well as adverse changes in BTB integrity, F-actin organization, and expressions of claudin-11 and Arp3. In Sertoli cells isolated from FLC-treated rat testis, Sertoli cell barrier tightness was increased. In Sertoli cells in vitro exposed to FLC, abnormal changes in the barrier permeability were also observed, and the protein expressions of occludin, claudin-11, ZO-1, connexin-43, and Arp3 were significantly decreased in a dose- and time-dependent manner. Furthermore, the FLC-induced adverse changes in Sertoli cell barrier and F-actin were partly alleviated by the induction of Arp3 overexpression. In conclusion, our findings revealed that FLC perturbed BTB/Sertoli cell barrier function through Arp3-mediated F-actin disorganization.


Subject(s)
Actin Cytoskeleton/drug effects , Actin-Related Protein 3/metabolism , Actins/metabolism , Air Pollutants, Occupational/toxicity , Blood-Testis Barrier/drug effects , Pyrrolidinones/toxicity , Reproduction/drug effects , Sertoli Cells/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Actin-Related Protein 3/genetics , Animals , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/pathology , Cells, Cultured , Dose-Response Relationship, Drug , Male , Permeability , Rats, Sprague-Dawley , Risk Assessment , Sertoli Cells/metabolism , Sertoli Cells/pathology , Signal Transduction/drug effects , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/pathology , Time Factors
18.
BMC Dev Biol ; 18(1): 15, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29940839

ABSTRACT

BACKGROUND: Diabetes and hypothyroidism produce adverse effects on body weight and sexual maturity by inhibiting body growth and metabolism. The occurrence of diabetes is always accompanied with thyroid dysfunction. Thus, it is important to take hypo- or hyper-thyroidism into consideration when exploring the adverse effects caused by diabetes. Previous reports have found hypothyroidism inhibits testicular growth by delaying Sertoli cell differentiation and proliferation. Hence, by establishing a mouse model of diabetes combined with hypothyroidism, we provided evidence that poly glandular autoimmune syndrome affected testicular development and spermatogenesis. RESULTS: we mimicked polyglandular deficiency syndrome in both immature and prepubertal mice by induction of diabetes and hypothyroidism, which caused decreases in serum concentrations of testosterone and insulin like growth factor 1 (IGF-1). Such reduction of growth factor resulted in inhibition of testicular and epididymal development. Moreover, expressions of Claudin-11 were observed between Sertoli cells and disrupted in the testes of syndrome group mice. We also found reduced sperm count and motility in prepubertal mice. CONCLUSIONS: This mimicry of the diabetes and thyroid dysfunction, will be helpful to better understand the reasons for male infertility in diabetic-cum-hypothyroid patients.


Subject(s)
Claudins/metabolism , Diabetes Mellitus/metabolism , Hypothyroidism/metabolism , Seminiferous Tubules/metabolism , Spermatogenesis , Animals , Blood Glucose/metabolism , Blood-Testis Barrier/pathology , Body Weight , Diabetes Mellitus/blood , Diabetes Mellitus/pathology , Epididymis/pathology , Female , Hypothyroidism/blood , Hypothyroidism/pathology , Insulin-Like Growth Factor I/metabolism , Male , Methimazole/administration & dosage , Mice, Inbred ICR , Organ Size , Sperm Motility , Streptozocin/administration & dosage , Testosterone/blood
19.
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
20.
Reproduction ; 154(3): 293-305, 2017 09.
Article in English | MEDLINE | ID: mdl-28667125

ABSTRACT

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.


Subject(s)
Apoptosis , Autoimmune Diseases/physiopathology , Disease Models, Animal , Follistatin/blood , Orchitis/physiopathology , Testis/metabolism , Up-Regulation , Activins/antagonists & inhibitors , Activins/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Biomarkers/blood , Biomarkers/metabolism , Blood-Testis Barrier/immunology , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/pathology , Blood-Testis Barrier/physiopathology , Disease Progression , Fibrosis , Follistatin/administration & dosage , Follistatin/genetics , Follistatin/metabolism , Gene Expression Regulation , Gene Transfer Techniques , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Orchitis/immunology , Orchitis/metabolism , Orchitis/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/metabolism , Testis/immunology , Testis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...