Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 976
Filter
1.
Am J Reprod Immunol ; 91(4): e13846, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38650368

ABSTRACT

PURPOSE: Abnormal spermatozoa significantly impact reproductive health, affecting fertility rates, potentially prolonging conception time, and increasing the risk of miscarriages. This study employs Mendelian randomization to explore their potential link with immune cells, aiming to reveal their potential causal association and wider implications for reproductive health. METHODS: We conducted forward and reverse Mendelian randomization analyses to explore the potential causal connection between 731 immune cell signatures and abnormal spermatozoa. Using publicly available genetic data, we investigated various immune signatures such as median fluorescence intensities (MFI), relative cell (RC), absolute cell (AC), and morphological parameters (MP). Robustness was ensured through comprehensive sensitivity analyses assessing consistency, heterogeneity, and potential horizontal pleiotropy. The MR study produced a statistically significant p-value of .0000684, Bonferroni-corrected for the 731 exposures. RESULTS: The Mendelian randomization analysis revealed strong indications of a reciprocal relationship between immune cell pathways and sperm integrity. When examining immune cell exposure, a potential causal link with abnormal sperm was observed in 35 different types of immune cells. Conversely, the reverse Mendelian randomization results indicated that abnormal sperm might causally affect 39 types of immune cells. These outcomes suggest a potential mutual influence between alterations in immune cell functionality and the quality of spermatozoa. CONCLUSION: This study highlights the close link between immune responses and sperm development, suggesting implications for reproductive health and immune therapies. Further research may offer crucial insights into male fertility and immune disorders.


Subject(s)
Mendelian Randomization Analysis , Spermatozoa , Male , Humans , Spermatozoa/immunology , Infertility, Male/genetics , Infertility, Male/immunology
2.
Andrology ; 10(1): 105-110, 2022 01.
Article in English | MEDLINE | ID: mdl-34331520

ABSTRACT

BACKGROUND: Studies on immunological infertility after inguinal hernia correction are few and not very representative. Anti-sperm antibodies have been shown to reduce male fertility. Although the extent of infertility due to anti-sperm antibodies alone is not very clear, data indicates that about 8%-10% of infertile patients have immunological infertility DESIGN: This retrospective study includes all infertile male patients (n = 2258) who underwent mixed antiglobulin reaction tests and urologic examination from 2000 to 2020. Sperm quality (assessed by the number of spermatozoa, their motility, vitality, and normal form) was also evaluated. Among these patients, 191 had previously undergone unilateral or bilateral inguinal hernia surgery repair. The aim of the study is to evaluate if there is a higher incidence of positive mixed antiglobulin reaction test among patients undergoing inguinal hernioplasty compared to the unselected infertile population. RESULTS: Anti-sperm antibodies would seem to increase in both patients who performed general andrological surgery and groin hernia correction, respectively 3.48 (95% Confidence Interval: 1.70-7.10; p < 0.001) and 2.45 (95% Confidence Interval: 1.01-5.99; p < 0.05) times more than the unselected infertile population. CONCLUSIONS: Mixed antiglobulin reaction test could be useful in patients undergone previous scrotal surgery or hernia correction men, to avoid false unexplained infertility diagnoses and to direct the couple to assisted reproductive technology procedures. Basal evaluation of spermatozoa does not actually consider andrological surgery as an indication to autoimmunity investigation.


Subject(s)
Autoimmune Diseases/immunology , Hernia, Inguinal/surgery , Herniorrhaphy/adverse effects , Infertility, Male/immunology , Postoperative Complications/immunology , Adult , Autoantibodies/immunology , Autoimmune Diseases/epidemiology , Humans , Incidence , Infertility, Male/epidemiology , Male , Postoperative Complications/epidemiology , Postoperative Period , Retrospective Studies , Semen Analysis , Spermatozoa/immunology
3.
Front Immunol ; 12: 729539, 2021.
Article in English | MEDLINE | ID: mdl-34531872

ABSTRACT

Background: Varicocele (VC) is present in 35 - 40% of men with infertility. However, current surgical and antioxidant treatments are not completely effective. In addition to oxidative stress, it is likely that other factors such as testicular immune microenvironment disorder contribute to irreversible testicular. Evidence suggests that VC is associated with anti-sperm antibodies (ASAs), spermatogenesis and testosterone secretion abnormalities, and testicular cytokine production. Moreover, inhibition of inflammation can alleviate VC-mediated pathogenesis. The normal function of the testis depends on its immune tolerance mechanism. Testicular immune regulation is complex, and many infectious or non-infectious diseases may damage this precision system. Results: The testicular immune microenvironment is composed of common immune cells and other cells involved in testicular immunity. The former includes testicular macrophages, T cells, dendritic cells (DCs), and mast cells, whereas the latter include Leydig cells and Sertoli cells (SCs). In animal models and in patients with VC, most studies have revealed an abnormal increase in the levels of ASAs and pro-inflammatory cytokines such as interleukin (IL)-1 and tumor necrosis factor (TNF)-alpha in the seminal plasma, testicular tissue, and even peripheral blood. It is also involved in the activation of potential inflammatory pathways, such as the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing (NLRP)-3 pathway. Finally, the development of VC-mediated infertility (VMI) may be facilitated by abnormal permeability of proteins, such as claudin-11, that constitute the blood-testis barrier (BTB). Conclusions: The testicular immune response, including the production of ASAs and inflammatory factors, activation of inflammatory pathways, and destruction of the BTB may be involved in the pathogenesis of VMI it is necessary to further explore how patient outcomes can be improved through immunotherapy.


Subject(s)
Cellular Microenvironment/immunology , Fertility , Infertility, Male/immunology , Inflammation Mediators/metabolism , Orchitis/immunology , Testis/immunology , Varicocele/immunology , Animals , Humans , Immunotherapy , Infertility, Male/metabolism , Infertility, Male/physiopathology , Infertility, Male/therapy , Male , Orchitis/metabolism , Orchitis/physiopathology , Orchitis/therapy , Signal Transduction , Testis/metabolism , Testis/physiopathology , Varicocele/metabolism , Varicocele/physiopathology , Varicocele/therapy
4.
PLoS One ; 16(9): e0257097, 2021.
Article in English | MEDLINE | ID: mdl-34506561

ABSTRACT

Ceratitis capitata (medfly) is one of the most devastating crop pests worldwide. The Sterile Insect Technique (SIT) is a control method that is based on the mass rearing of males, their sterilization, and release in the field. However, the effectiveness of the technique depends on the quality of the released males and their fitness. We previously isolated and selected a probiotic bacteria (Enterobacter sp.), from wild-caught medflies, according to criteria that improved biological quality traits of reared medfly males.We firstly evaluated the impact of the irradiation on the expression of different immune and stress genes in the medfly sterile males. Expression was measured at differents time points ranging from 0 to 168 h after irradiation to capture the response of genes with distinct temporal expression patterns. Then, we supplemented the larval diet with previously isolated Enterobacter sp.strain, live and autoclaved at various concentrations to see whether the probiotic treatments affect, through their protective role, the gene expression level, and quality traits. The irradiation had significant effect on the genes attacin, cecropin, PGPR-LC, hsp23, and hsp70 level expression. The expression of attacin and PGPR-LC was up-regulated while that of cecropin was down-regulated. Hsp genes showed decreased levels between 0 and 18 h to peak at 72 h. However, the supplementation of the probiotic strain, either live or autoclaved, was statistically significant only for attacingene. However, significant interaction time x probiotic was noticed for attacin, cecropin, hsp23 and hsp70. The probiotic treatments also improved the quality control parameters like pupal weight. From this work we can conclude that a consortium of parabiotics (autoclaved probiotics) treatment will be recommended in insectaries considering both the beneficial effects on mass reared insects and its general safety for insectary workers and for environment.


Subject(s)
Ceratitis capitata/immunology , Ceratitis capitata/radiation effects , Diet , Immunity/drug effects , Infertility, Male/immunology , Pest Control, Biological , Probiotics/pharmacology , Stress, Physiological/drug effects , Animals , Body Weight/drug effects , Ceratitis capitata/genetics , Cobalt Radioisotopes , Female , Flight, Animal/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Immunity/genetics , Immunity/radiation effects , Infertility, Male/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Male , Pupa/drug effects , Statistics as Topic , Stress, Physiological/genetics
5.
Mol Biol Rep ; 48(8): 5935-5942, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34319544

ABSTRACT

BACKGROUND: Male infertility is a problem that affects 10-15% of men of reproductive age. In particular, gametogenesis is a complex process in which inflammation may play a central role through the secretion of cytokines and the expression of microRNAs. We assessed the potential role of proinflammatory cytokines (TNF-α, IL-6 and IL-1α) and microRNAs (miR-146a-5p, miR-34a-5p and miR-23a-3p) in the seminal plasma of infertile men compared to controls, evaluating their correlation with seminal and biochemical parameters. METHODS AND RESULTS: Expression of cytokines and microRNAs was analyzed by ELISA and q-PCR. Our data shows that IL-1α was significantly increased in the azoospermic group compared to controls, TNF-α mRNA was more expressed in the oligozoospermic group than controls. There were no significant differences in miRNAs expression among the three groups. The correlations between sperm parameters and inflammatory markers were evaluated, however no significance was highlighted. CONCLUSIONS: The determination of each inflammatory marker separately in the seminal plasma of subfertile men, despite some significant differences, does not have a diagnostic value in male infertility even if an assay of selective pro-inflammatory cytokines and microRNAs in the semen may improve the diagnosis of male infertility.


Subject(s)
Infertility, Male/genetics , Infertility, Male/immunology , Semen/physiology , Adult , Biomarkers/metabolism , Cytokines/metabolism , Humans , Interleukin-1alpha/metabolism , Interleukin-6/metabolism , Male , MicroRNAs/genetics , Semen/metabolism , Spermatozoa/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tunisia
6.
Am J Pathol ; 191(9): 1592-1609, 2021 09.
Article in English | MEDLINE | ID: mdl-34126085

ABSTRACT

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


Subject(s)
Infertility, Male/immunology , Polyendocrinopathies, Autoimmune/pathology , Transcription Factors/metabolism , Animals , Female , Infertility, Male/genetics , Male , Mice , Mice, Knockout , Polyendocrinopathies, Autoimmune/genetics , Transcription Factors/genetics , AIRE Protein
7.
Biomed Pharmacother ; 139: 111514, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33951576

ABSTRACT

Male immune infertility is a kind of disease that damages family life and happiness. The development of novel methods treating male immune infertility is of great significance. This study aimed to investigate the therapeutic effects of Chinese medicine Xiaokang Liuwei Dihuang decoction on immune infertility of male rats and explored the involved mechanisms. Model rats were established by lipopolysaccharide (LPS) injection. Anti-sperm antibody (AsAb) was detected by ELISA assay and testicular cell apoptosis was evaluated by TUNEL staining to verify the successful model establishment and screen suitable doses of Xiaokang Liuwei Dihuang decoction. Thirty rats were then divided into five groups (n = 6 per group): Control, LPS, Xiaokang Liuwei Dihuang decoction (15.12 g/kg, 30.24 g/kg and 45.36 g/kg). Results of HE staining showed that compared with LPS group, Xiaokang Liuwei Dihuang decoction treatments gradually improved the morphology of seminiferous tubules and elevated the number of spermatogenic cells as the doses increased. The sperm number and the levels of testosterone, luteinizing hormone (LH) and follicle stimulating hormone (FSH) in the serum of 15.12 g/kg, 30.24 g/kg and 45.36 g/kg Xiaokang Liuwei Dihuang decoction groups were much higher than those in LPS group. Results of TUNEL staining, ELISA assay and western blot showed that compared with LPS group, the testicular cell apoptosis and the levels of interleukin 1ß (IL-1ß), tumor necrosis factor α (TNF-α), AsAb, malondialdehyde (MDA) and toll-like receptor 2 (TLR2) in the testicular tissue significantly decreased in three Xiaokang Liuwei Dihuang decoction groups. Compared with LPS group, Bax expression in the 30.24 g/kg and 45.36 g/kg Xiaokang Liuwei Dihuang decoction groups was significantly down-regulated as well. In conclusion, Xiaokang Liuwei Dihuang decoction might ameliorate the immune infertility of male rats induced by LPS through regulating the levels of sex hormones, reactive oxygen species, pro-apoptotic and immune factors.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Drugs, Chinese Herbal/therapeutic use , Gonadal Steroid Hormones/metabolism , Infertility, Male/drug therapy , Infertility, Male/immunology , Reactive Oxygen Species/metabolism , Animals , Autoantibodies/analysis , Immunologic Factors/metabolism , Infertility, Male/chemically induced , Lipopolysaccharides , Male , Rats , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Seminiferous Tubules/metabolism , Sperm Count , Spermatogenesis/drug effects , Spermatozoa/immunology , Testis/cytology , Testis/drug effects
8.
J Reprod Immunol ; 145: 103318, 2021 06.
Article in English | MEDLINE | ID: mdl-33894646

ABSTRACT

Experimental autoimmune orchitis (EAO) may be used as a model to investigate immunological infertility in men. Murine EAO is induced via immunization with auto-immunogenic antigens (AIAgs) from testicular germ cells (TGCs). CD4 + T cells play a crucial role in EAO induction. However, whether AIAgs induce an immune response remains unclear. We aimed to identify self-antigens that induce EAO by screening a phage display library of random TGC peptides using IgG from EAO-induced A/J mice. Twenty TGC-specific AIAgs were detected, and G protein-coupled receptor kinase 2 interacting protein-1 (GIT1) and heat shock protein A4L (HSPA4L) were identified as candidate AIAgs that induce EAO. Immunization with GIT1 or HSPA4L, emulsified in complete Freund's adjuvant, resulted in 66 % or 100 % incidence of EAO, respectively, indicating that HSPA4L is a most potent AIAg that induces EAO in mice. These findings may expectedly help improve the diagnostic procedures and treatment of immunological infertility in men.


Subject(s)
Autoantigens/immunology , HSP70 Heat-Shock Proteins/immunology , Orchitis/immunology , Animals , Autoantigens/analysis , Biomarkers/analysis , Cell Cycle Proteins/administration & dosage , Cell Cycle Proteins/immunology , Disease Models, Animal , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , GTPase-Activating Proteins/administration & dosage , GTPase-Activating Proteins/immunology , HSP70 Heat-Shock Proteins/administration & dosage , HSP70 Heat-Shock Proteins/analysis , Humans , Infertility, Male/diagnosis , Infertility, Male/immunology , Male , Mice , Orchitis/diagnosis , Orchitis/pathology , Testis/immunology , Testis/pathology
9.
J Reprod Immunol ; 145: 103325, 2021 06.
Article in English | MEDLINE | ID: mdl-33930667

ABSTRACT

Contraceptive vaccine (CV) is a valuable, non-invasive, and alternative method for purposeful contraception. Sperm antigens are useful targets for producing CVs due to their specialized expression in sperm. In this study, a recombinant protein containing three main sperm epitopes (IZUMO1, SACA3, and PH-20) was designed and evaluated as CV to control fertility in male mice. The chimeric recombinant protein was expressed and purified in E. coli. Male mice were immunized by 100 µg purified protein and sera were collected to assess IgG antibodies. Evaluating the reproductive performance, immunized male mice mated with normal-fertile female mice and mating rate and the number of newborns was studied. Immunized mice were sacrificed and necropsy and histopathology studies were conducted. The results revealed that the designed chimeric protein stimulated the immune system of the mice effectively. The level of IgG antibody was significantly higher in vaccinated mouse rather than control mouse. Eighty percent of the vaccinated mice became infertile and in the remaining ones, the number of children decreased to 4-6 offspring instead of 10-12 in normal mice. Histopathological studies showed that no organs including heart, brain, lung, liver, kidney and intestine were damaged. However, Normal spermatogenesis has been disrupted and necrotic spermatogonia cells were reported in Seminiferous tubules. We concluded that the designed chimeric protein containing IZUMO1, SACA3, and PH-20 epitopes can stimulate the immune system and cause male contraception without any side effects.


Subject(s)
Contraception, Immunologic/methods , Infertility, Male/immunology , Recombinant Fusion Proteins/immunology , Vaccines, Contraceptive/immunology , Animals , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Disease Models, Animal , Epitopes/administration & dosage , Epitopes/genetics , Epitopes/immunology , Humans , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/immunology , Immunoglobulins/administration & dosage , Immunoglobulins/genetics , Immunoglobulins/immunology , Infertility, Male/pathology , Isoantigens/administration & dosage , Isoantigens/genetics , Isoantigens/immunology , Male , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Seminal Plasma Proteins/administration & dosage , Seminal Plasma Proteins/genetics , Seminal Plasma Proteins/immunology , Seminiferous Tubules/cytology , Seminiferous Tubules/immunology , Seminiferous Tubules/pathology , Spermatogonia/immunology , Spermatogonia/pathology , Vaccines, Contraceptive/administration & dosage , Vaccines, Contraceptive/genetics
10.
Front Immunol ; 12: 582946, 2021.
Article in English | MEDLINE | ID: mdl-33815357

ABSTRACT

The causative agent of mumps is a single-stranded, non-segmented, negative sense RNA virus belonging to the Paramyxoviridae family. Besides the classic symptom of painfully swollen parotid salivary glands (parotitis) in mumps virus (MuV)-infected men, orchitis is the most common form of extra-salivary gland inflammation. Mumps orchitis frequently occurs in young adult men, and leads to pain and swelling of the testis. The administration of MuV vaccines in children has been proven highly effective in reducing the incidence of mumps. However, a recent global outbreak of mumps and the high rate of orchitis have recently been considered as threats to male fertility. The pathogenesis of mumps orchitis remains largely unclear due to lack of systematic clinical data analysis and animal models studies. The alarming increase in the incidence of mumps orchitis and the high risk of the male fertility have thus become a major health concern. Recent studies have revealed the mechanisms by which MuV-host cells interact and MuV infection induces inflammatory responses in testicular cells. In this mini-review, we highlight advances in our knowledge of the clinical aspects and possible mechanisms of mumps orchitis.


Subject(s)
Infertility, Male/immunology , Mumps virus/immunology , Mumps/immunology , Orchitis/immunology , Host-Pathogen Interactions/immunology , Humans , Infertility, Male/complications , Infertility, Male/prevention & control , Male , Mumps/complications , Mumps/virology , Mumps Vaccine/administration & dosage , Mumps Vaccine/immunology , Mumps virus/physiology , Orchitis/complications , Orchitis/virology , Risk Factors , Vaccination/methods
11.
Am J Reprod Immunol ; 86(1): e13401, 2021 07.
Article in English | MEDLINE | ID: mdl-33576153

ABSTRACT

PROBLEM: While the testes represent an immune-privileged organ, there is evidence that systemic inflammation is accompanied by local inflammatory responses. We therefore examined whether transient systemic inflammation caused any inflammatory and functional consequences in murine testes. METHOD OF STUDY: Using a single systemic administration of Toll-like receptor (TLR) agonists [lipopolysaccharide (LPS) or peptidoglycan (PG) or polyinosinic-polycytidylic acid (polyIC)] in young adult male mice, we assessed testicular immune-inflammatory landscape and reproductive functionality. RESULTS: Our findings demonstrated a significant induction of testicular TNF-α, IL-1ß and IL-6 transcripts within 24 h of TLR agonist injection. By day 6, these cytokine levels returned to baseline. While there was no change in caudal sperm counts at early time points, eight weeks later, twofold decrease in sperm count and reduced testicular testosterone levels were evident. When these mice were subjected to mating studies, no differences in mating efficiencies or litter sizes were observed compared with controls. Nonetheless, the neonatal weights of progeny from LPS/PG/polyIC-treated sires were significantly lower than controls. Postnatal weight gain up to three weeks was also slower in the progeny of LPS/polyIC-treated sires. Placental weights at 17.5 days post-coitum were significantly lower in females mated to LPS- and polyIC-treated males. Given this likelihood of an epigenetic effect, we found lower testicular levels of histone methyltransferase enzyme, mixed-lineage leukaemia-1, in mice given LPS/PG/polyIC 8 weeks earlier. CONCLUSION: Exposure to transient systemic inflammation leads to transient local inflammation in the testes, with persistent sperm-mediated consequences for foetal development.


Subject(s)
Infertility, Male/immunology , Inflammation/immunology , Orchitis/immunology , Testis/metabolism , Thinness/immunology , Animals , Cytokines/metabolism , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Immune Privilege , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Peptidoglycan/immunology , Poly I-C/immunology , Testis/pathology
12.
Methods Mol Biol ; 2198: 255-268, 2021.
Article in English | MEDLINE | ID: mdl-32822037

ABSTRACT

Male infertility is associated with several causes affecting the paternal nucleus such as DNA lesions (breaks, deletions, mutations, ...) or numerical chromosome anomalies. More recently, male infertility has also been associated with changes in the sperm epigenome, including modification in the topology of chromatin (Olszewska et al., Chromosome Research 16:875-890, 2008; Alladin et al., Syst Biol Reprod Med 59: 146-152, 2013) ref with number 1, 2. Indeed, the positioning of chromosomes in the sperm nucleus is nonrandom and defines chromosome territories (Champroux et al., Genes (Basel) 9:501, 2018) ref with number 3 whose optimal organization determines the success of embryonic development. In this context, the study of the spatial distribution of chromosomes in sperm cells could be relevant for clinical diagnosis. We describe here a in situ fluorescence hybridization (FISH) strategy coupled with a fluorescent immunocytochemistry approach followed by confocal analysis and reconstruction (2D/3D) as a powerful tool to analyze the location of chromosomes in the sperm nucleus using the mouse sperm as a model. Already, the two-dimensional (2D) analysis of FISH and immunofluorescence data reveal the location of chromosomes as well as the different markings on the spermatic nucleus. In addition, a good 3D rendering after Imaris software processing was obtained when Z-stacks of images were acquired over a defined volume (10 µm × 13 µm × 15 µm) with a sequential scanning mode to minimize bleed-through effects and avoid overlapping wavelengths.


Subject(s)
Chromosome Positioning/immunology , Microscopy, Confocal/methods , Spermatozoa/immunology , Aneuploidy , Animals , Cell Nucleus/immunology , Chromatin , Chromosome Aberrations , Chromosome Positioning/genetics , Chromosomes/immunology , Disease Models, Animal , Fluorescent Antibody Technique/methods , In Situ Hybridization, Fluorescence/methods , Infertility, Male/immunology , Male , Mice , Spermatozoa/cytology
13.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33372158

ABSTRACT

Macrophages are the principal immune cells of the epididymis and testis, but their origins, heterogeneity, development, and maintenance are not well understood. Here, we describe distinct populations of epididymal and testicular macrophages that display an organ-specific cellular identity. Combining in vivo fate-mapping, chimeric and parabiotic mouse models with in-depth cellular analyses, we found that CD64hiMHCIIlo and CD64loMHCIIhi macrophage populations of epididymis and testis arise sequentially from yolk sac erythro-myeloid progenitors, embryonic hematopoiesis, and nascent neonatal monocytes. While monocytes were the major developmental source of both epididymal and testicular macrophages, both populations self-maintain in the steady-state independent of bone marrow hematopoietic precursors. However, after radiation-induced macrophage ablation or during infection, bone marrow-derived circulating monocytes are recruited to the epididymis and testis, giving rise to inflammatory macrophages that promote tissue damage. These results define the layered ontogeny, maintenance and inflammatory response of macrophage populations in the male reproductive organs.


Subject(s)
Infertility, Male/immunology , Macrophages/immunology , Macrophages/metabolism , Animals , Cell Differentiation , Cell Lineage , Epididymis/immunology , Epididymis/metabolism , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Testis/immunology , Testis/metabolism
14.
Front Immunol ; 11: 2115, 2020.
Article in English | MEDLINE | ID: mdl-33117332

ABSTRACT

The epididymis is an important male accessory sex organ where sperm motility and fertilization ability develop. When spermatozoa carrying foreign antigens enter the epididymis, the epididymis shows "immune privilege" to tolerate them. It is well-known that a tolerogenic environment exists in the caput epididymis, while pro-inflammatory circumstances prefer the cauda epididymis. This meticulously regulated immune environment not only protects spermatozoa from autoimmunity but also defends spermatozoa against pathogenic damage. Epididymitis is one of the common causes of male infertility. Up to 40% of patients suffer from permanent oligospermia or azoospermia. This is related to the immune characteristics of the epididymis itself. Moreover, epididymitis induced by different pathogenic microbial infections has different characteristics. This article elaborates on the distribution and immune response characteristics of epididymis immune cells, the role of epididymis epithelial cells (EECs), and the epididymis defense against different pathogenic infections (such as uropathogenic Escherichia coli, Chlamydia trachomatis, and viruses to provide therapeutic approaches for epididymitis and its subsequent fertility problems.


Subject(s)
Epididymis/immunology , Epididymitis/immunology , Spermatozoa/immunology , Activins/physiology , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Blood-Testis Barrier , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Defensins/physiology , Epididymitis/complications , Epididymitis/epidemiology , Epididymitis/microbiology , Escherichia coli Infections/immunology , Humans , Immune System/cytology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Infertility, Male/etiology , Infertility, Male/immunology , Infertility, Male/microbiology , Male , Mice , Middle Aged , TGF-beta Superfamily Proteins/physiology , Uropathogenic Escherichia coli/immunology , Virus Diseases/immunology , Young Adult
15.
J Reprod Immunol ; 142: 103213, 2020 11.
Article in English | MEDLINE | ID: mdl-33080435

ABSTRACT

COVID-19 is a present-day complex pandemic infection with unpredictable levels of morbidity and mortality in various global populations. COVID-19 is associated with the different comorbidities with its change in biological function such as causing heart dysfunction via deregulating ACE-2 receptor, gastrointestinal risk via causing vomiting, diarrhea, and abdominal pain, chronic kidney disease via proteinuria and hematuria, diabetes mellitus, liver injury via increasing ALT, AST and bilirubin level, lung injury, CNS risk, ocular risk, and cancer risk. In this, we are focused on the COVID-19 connected with male infertility. Some of the studies show that the patients of COVID-19 are associated with impaired spermatogenesis. Impaired spermatogenesis via COVID-19 decreases the level of testosterone by disturbing cytokines such as TNF-α, IL-4, IL-6, and IL-12 and further, attenuates the sperm count. COVID-19 is causing inflammation via TNF-α and interferons. IL-4 plays an eminent role in the activation of the JAK-STAT pathway and leads to the disturbing pro-inflammatory cytokine as well as further cause's male infertility. Th2 activates the IL-4 through IgG and IgE and mediates apoptosis with the triggering of STAT signaling. The activated STAT signaling augments Batf/Irf4, and the Bach2/Batf pathway. On the other hand, SARS-CoV-2 is activating the level of Th2 cells. So, we hypothesized that the augmented Th2 cells would disturb the level of IL-4, JAK-STAT signaling, Batf/Irf4, and Bach2/Batf pathway. The disturbed IL-4 decreases the level of the ACE-2 with the inflammation. This further leads to male infertility in COVID-19 patients. So, in this hypothesis, we focused on the role of IL-4 in COVID-19 patients associated with male infertility via Th2 cells and JAK-STAT signaling.


Subject(s)
COVID-19/complications , Infertility, Male/virology , Interleukin-4/immunology , Signal Transduction/immunology , Humans , Infertility, Male/immunology , Infertility, Male/physiopathology , Male , SARS-CoV-2
17.
Am J Mens Health ; 14(5): 1557988320939731, 2020.
Article in English | MEDLINE | ID: mdl-32938274

ABSTRACT

Infertility is among the most serious medical problems worldwide. Male factors contribute to 40%-50% of all infertility cases, and approximately 7% of men worldwide are affected by infertility. Spermatozoa are extremely vulnerable to oxidative insult. Oxidative stress results in axonemal damage and increased midpiece sperm morphological defects, which lead to reduced sperm motility. The aim of the study is to evaluate the association between sperm motility and the levels of selected antioxidants, cytokines, and markers of oxidative damage in the seminal plasma.The study group included 107 healthy males, who were split into two subgroups based on the percentage of motile spermatozoa after 1 hr: low motility (LM, n = 51) and high motility (HM, n = 56).The glucose-6-phosphate dehydrogenase (G6PD) activity was 52% lower in the LM group compared to that in the HM group. The level of malondialdehyde (MDA) was 12% higher in the LM group compared to that in the HM group. Similarly, the median values of interleukin (IL)-1ß, IL-10, IL-12, and tumor necrosis factor alpha (TNF-α) were higher in the LM group than those in the HM group. Results of the present study revealed that the percentage of motile spermatozoa after 1 hr correlated positively with the levels of IL-1ß, IL-10, IL-12, and TNFα.The lower motility of spermatozoa in healthy men is associated with a decreased activity of G6PD and increased levels of cytokines, which may be related to increased oxidative stress in seminal plasma that manifests as an increased level of MDA.


Subject(s)
Infertility, Male/immunology , Oxidative Stress/immunology , Sperm Motility , Adult , Biomarkers/blood , Humans , Male
18.
J Reprod Immunol ; 142: 103183, 2020 11.
Article in English | MEDLINE | ID: mdl-32853846

ABSTRACT

Bacterial infections play a disruptive and hidden role in male reproductive failure. Different kinds of bacteria are often able to interfere with reproductive function in both sexes and lead to infertility. In this study, to further evaluate the role of bacterial infections in male reproduction we provided an extensive overview of so far researches investigating the effects of bacterial infections on male fertility. We searched Medline, PubMed, Scopus and Google scholar databases to identify the potentially relevant studies on bacterial infections and their implications in male infertility. All the bacteria included in this article have negative effects on the male reproductive function; however, there is ample evidence to blame bacteria such as Escherichia coli, Chlamydia trachomatis, Ureaplasma, Mycoplasma and Staphylococcus aureus for reduced fertility and deterioration of sperm parameters. More studies are needed to clarify the molecular mechanisms by which different bacteria exert their detrimental effects on male reproductive system. Getting more insight into probable mechanisms, would significantly facilitate the production of new, advanced, and effective remedies in the future. In view of all evidence, we strongly suggest increasing awareness among people and considering screening programs for patients seeking fertility both to avoid transmission and to improve fertility outcomes among them.


Subject(s)
Bacterial Infections/complications , Genitalia, Male/microbiology , Infertility, Male/immunology , Reproductive Tract Infections/complications , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bacterial Infections/therapy , Fertility/immunology , Genitalia, Male/immunology , Humans , Infertility, Male/microbiology , Infertility, Male/prevention & control , Male , Reproductive Tract Infections/immunology , Reproductive Tract Infections/microbiology , Reproductive Tract Infections/therapy , Spermatogenesis/immunology
19.
J Reprod Immunol ; 142: 103178, 2020 11.
Article in English | MEDLINE | ID: mdl-32739646

ABSTRACT

Natural killer (NK) cells are important effector lymphocytes that play a pivotal role in the innate and adaptive immune responses to tumors and viral infection. NKT cells are a heterogeneous group of T cells that share properties with both T cells and NK cells. They display immunoregulatory properties as they facilitate the cell-mediated immune response to tumors and infectious diseases, and inhibit cell-mediated immunity associated with autoimmune diseases and allograft rejection. However, the roles of NK and NKT cells in the male reproductive tract remain largely unexplored, in particular, NKT cells, tissue distribution, and state of health or disease. Infection and inflammation of the male genital tract are thought to be the primary etiological factors of male infertility. In this review, we considered this complex and rapidly growing field. We summarize the recent findings and the characterization and roles of NK and NKT cells in the male reproductive tract, including the testis, epididymis, prostate, seminal vesicle, and semen, to enhance our understanding of the immunological mechanisms of male infertility and for the design effective vaccines for male reproductive health in the future.


Subject(s)
Infertility, Male/immunology , Killer Cells, Natural/immunology , Natural Killer T-Cells/immunology , Prostatic Neoplasms/immunology , Reproductive Tract Infections/immunology , Genitalia, Male/immunology , Genitalia, Male/pathology , Humans , Immune Privilege , Immunity, Cellular , Immunity, Innate , Infertility, Male/prevention & control , Killer Cells, Natural/metabolism , Male , Natural Killer T-Cells/metabolism , Prostatic Neoplasms/complications , Prostatic Neoplasms/pathology , Reproductive Tract Infections/complications , Reproductive Tract Infections/pathology , Semen/immunology , Spermatozoa/immunology , Tumor Microenvironment/immunology
20.
Mol Reprod Dev ; 87(9): 952-965, 2020 09.
Article in English | MEDLINE | ID: mdl-32749760

ABSTRACT

Male infertility or subfertility is frequently associated with disruption of the hypothalamic-pituitary-testis axis events, like secondary hypogonadism. However, little is known how this condition affects the proteomic composition of the epididymal fluid. In the present study, we evaluated the proteomic changes in the cauda epididymal fluid (CEF) in a swine model of secondary hypogonadism induced by anti-GnRH immunization using multidimensional protein identification technology. Seven hundred and eighteen proteins were identified in both GnRH-immunized and control groups. GnRH immunization doubled the number of proteins in the CEF, with 417 proteins being found exclusively in samples from GnRH-immunized boars. CEF from GnRH-immunized boars presented an increase in the number of proteins related to cellular and metabolic processes, with affinity to organic cyclic compounds, small molecules, and heterocyclic compounds, as well changed the enzymatic profile of the CEF. Also, a significant increase in the number of proteins associated to the ubiquitin-proteasome system was identified in CEF from GnRH-immunized animals. These results bring strong evidence of the impact of secondary hypogonadism on the epididymal environment, which is responsible for sperm maturation and storage prior ejaculation. Finally, the differently expressed proteins in the CEF are putative seminal biomarkers for testicular and epididymal disorders caused by secondary hypogonadism.


Subject(s)
Body Fluids/metabolism , Epididymis/metabolism , Hypogonadism/metabolism , Infertility, Male/metabolism , Proteome/metabolism , Animals , Antibodies/pharmacology , Body Fluids/chemistry , Body Fluids/drug effects , Contraception, Immunologic/methods , Contraception, Immunologic/veterinary , Epididymis/chemistry , Epididymis/drug effects , Gonadotropin-Releasing Hormone/immunology , Gonadotropin-Releasing Hormone/metabolism , Hypogonadism/etiology , Hypogonadism/immunology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Infertility, Male/etiology , Infertility, Male/immunology , Infertility, Male/veterinary , Male , Models, Animal , Proteome/analysis , Proteome/drug effects , Proteomics , Signal Transduction/drug effects , Swine/metabolism , Testis/drug effects , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...