1
|
Vinay L, Hess RA, Belleannée C. Human efferent ductules and epididymis display unique cell lineages with motile and primary cilia. Andrology 2025; 13:894-910. [PMID: 39212979 PMCID: PMC12006893 DOI: 10.1111/andr.13732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Previous research has illustrated the role of cilia as mechanical and sensory antennae in various organs within the mammalian male reproductive system across different developmental stages. Despite their significance in both organ development and homeostasis, primary cilia in the human male reproductive excurrent duct have been overlooked due to limited access to human specimens. OBJECTIVE This study aimed to characterize the unique cellular composition of human efferent and epididymal ducts, with a focus on their association with primary cilia. MATERIALS AND METHODS Human efferent ductules/epididymides from five donors aged 32-47 years, were obtained through our local organ transplant program. Cell lineage specificity and primary cilia features were examined by immunofluorescent staining and confocal microscopy in the efferent ductules and the distinct segments of the epididymis. RESULTS The epithelium of the human efferent duct exhibited estrogen receptor-positive cells with primary cilia, FoxJ1-positive multiciliated cells with numerous motile cilia, and non-ciliated intraepithelial immune cells. Notably, intraluminal macrophages, identified by CD163/CD68 positivity, were observed to engage in sperm phagocytosis. In all three segments of the human epididymis, primary cilia were found on the surface of principal and basal cells. DISCUSSION AND CONCLUSIONS Our research indicates that the human efferent ductules create a distinct environment, characterized by the presence of two types of ciliated cells that are in contact with immune cells. The discovery of sensory primary cilia exposed on the surface of reabsorptive cells in the efferent ductules, as well as on basal and principal cells in the epididymis, lays the foundation for complementary functional studies. This research uncovers novel characteristics exclusive to human efferent ductules and epididymides, providing a basis for exploring innovative approaches to male contraception and infertility treatment.
Collapse
Affiliation(s)
- Ludovic Vinay
- Faculty of MedicineDepartment of ObstetricsGynecology and ReproductionUniversité Laval, CHU de Québec Research Center (CHUL)Quebec CityCanada
- Centre de recherche en ReproductionDéveloppement et Santé IntergénérationnelleQuebec CityCanada
| | - Rex A. Hess
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Clémence Belleannée
- Faculty of MedicineDepartment of ObstetricsGynecology and ReproductionUniversité Laval, CHU de Québec Research Center (CHUL)Quebec CityCanada
- Centre de recherche en ReproductionDéveloppement et Santé IntergénérationnelleQuebec CityCanada
| |
Collapse
|
2
|
Wang Y, Hong Y. Genetic insights into the immunological basis of male infertility: a translational perspective. F&S SCIENCE 2025; 6:130-140. [PMID: 39988235 DOI: 10.1016/j.xfss.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
OBJECTIVE To elaborate the causal relationships between specific immunocyte phenotypes and male infertility. DESIGN Mendelian randomization using genome-wide association study data. SUBJECTS Large cohorts of European ancestry. EXPOSURE 731 immunocyte phenotypes or male infertility. MAIN OUTCOMES MEASURES Genetic variants were used as instrumental variables to infer causality, minimizing confounding and bias. The causal associations were assessed using the inverse variance-weighted (IVW) method for primary analysis, and the findings were validated using MR-Egger, Weighted Median, Simple Mode, and Weighted Mode approaches. Additional sensitivity analyses were performed to validate the robustness of the findings. RESULTS Our analysis identified significant causal associations between specific immunocyte phenotypes and male infertility. Phenotypes such as naive-mature B cell %lymphocyte (odds ratio [OR] = 1.257) and IgD- CD38dim %B cell (OR = 1.100) were positively associated with increased infertility risk, whereas phenotypes like CD39+ CD8br %T cell (OR = 0.856) and B cells activator of the TNF-α family receptor (BAFF-R) on transitional (OR = 0.833) were negatively associated, suggesting a protective effect. Additionally, reverse MR analysis revealed that male infertility might causally affect certain immunocyte phenotypes, including CD14- CD16+ monocyte %monocyte (OR = 1.049). CONCLUSION This study provides robust evidence for the causal role of specific immunocyte phenotypes in male infertility and highlights the bidirectional relationship between immune function and reproductive health. These findings provide new insights into the immunological factors contributing to male infertility and suggest potential biomarkers and therapeutic targets for future research and clinical interventions.
Collapse
Affiliation(s)
- Yi Wang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanggang Hong
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Rodriguez AR, Babcock RL, Guimarães JPT, Kaur G, Dufour JM. Immune Regulation in the Testis and Epididymis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:25-47. [PMID: 40301251 DOI: 10.1007/978-3-031-82990-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Immune regulation within the male reproductive tract is necessary for the protection of the spermatogenic cells from a detrimental immune response. This is done by the production of immunomodulatory factors, sequestration of the spermatogenic cells behind the blood-testis barrier (BTB) and blood-epididymal barrier (BEB), and controlled presentation of germ cell antigens. At the same time, bacteria and viruses can take advantage of this unique environment, inducing inflammation and infecting the male reproductive tract, resulting in histological damage, germ cell loss, and potentially leading to infertility. An antimicrobial response is important to counter this affliction that if not properly controlled can lead to germ cell autoimmunity or provide a haven for pathogens. Therefore, the immunomodulatory environment within the testis and epididymis is intrinsically important to maintain this property.
Collapse
Affiliation(s)
- Alexis R Rodriguez
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Rachel L Babcock
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | | | - Gurvinder Kaur
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Jannette M Dufour
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
4
|
Qu H, Liu Q, Zheng D, Ni Y, Xiao X. A Comprehensive Bibliometric Analysis of Orchitis Research from 1980 to 2023. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:207-243. [PMID: 40301259 DOI: 10.1007/978-3-031-82990-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Orchitis, an inflammation of the testes, presents significant implications for male fertility and has been a focal area of scientific inquiry over the past four decades. This study employs a comprehensive bibliometric analysis to assess the progression of global research on orchitis from 1980 to 2023. Drawing from a dataset of 1586 publications indexed in the Web of Science Core Collection, we uncover emerging patterns, collaborations, and pivotal works that have shaped the field. The United States, China, and Germany emerge as leading contributors, while the Journal of Urology stands out as a primary publishing avenue. The results highlight the increasing recognition of autoimmune responses, alongside infectious agents, as key contributors to orchitis. Moreover, molecules such as TNF-α, IL-6, and IFN-γ are identified as central to the disease's pathology. The dynamic interplay of testosterone and regulatory T cells is underscored as a determinant of the testicular immune milieu. Notably, disruptions in the blood-testis barrier (BTB) and germ cell apoptosis emerge as pivotal consequences of the condition. This analysis underscores the expansive and multidisciplinary nature of orchitis research, revealing a consistent growth in collaborative endeavors. In summary, our findings catalog the evolution of orchitis research, providing a consolidated perspective on past achievements and signposting future research avenues. Such insights are instrumental for researchers aiming to navigate the complexities of orchitis and its multifaceted impact on male reproductive health.
Collapse
Affiliation(s)
- Haoyang Qu
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Qiubei Liu
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Dongwang Zheng
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Ya Ni
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
- Zhejiang Provincial Laboratory of Experimental Animal's and Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China
| | - Xiang Xiao
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China.
- Zhejiang Provincial Laboratory of Experimental Animal's and Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China.
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
5
|
Elizagaray ML, Barrachina F, Avenatti MC, Bastepe I, Chen A, Odriozola A, Ukairo O, Ros VD, Ottino K, Subiran N, Battistone MA. Chronic inflammation drives epididymal tertiary lymphoid structure formation and autoimmune fertility disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623224. [PMID: 39605691 PMCID: PMC11601424 DOI: 10.1101/2024.11.12.623224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The incomplete understanding of epididymal mucosal immunity is a significant contributing factor to the classification of many male infertility cases as idiopathic. Conditions that disrupt the immune balance in the male reproductive tract, such as vasectomy and infections, can expose sperm to the immune system, leading to increased production of anti-sperm antibodies (ASAs) and subsequent reproductive challenges. Regulatory T cells (Tregs) regulate inflammation and maintain sperm tolerance. In a murine model, we demonstrated that disrupting sperm immunotolerance induces chronic autoimmune responses characterized by antibody production targeting sperm and reproductive tissue autoantigens and unique tissue-specific immune cell signatures in the epididymis and testis. Such inflammatory features impair sperm function, contribute to epididymal damage, and drive sustained male subfertility. Tertiary lymphoid structures (TLSs) were formed within the epididymis after Treg depletion, defined by clusters of heterogenous B and T cells, fibroblasts, and endothelial cells. These ectopic structures perpetuate inflammation and lower the activation threshold for future immune threats. Similar isotypes of autoantibodies were detected in the seminal plasma of infertile patients, suggesting shared mechanistic pathways between mice and humans. Overall, we provide an in-depth understanding of the diverse B- and T-cell dynamics and TLS formation during epididymitis to develop precision-targeted therapies for infertility and chronic inflammation. Additionally, this immunological characterization of the epididymal microenvironment has the potential to identify novel targets for the development of male contraceptives. One Sentence Summary Understanding the epididymal immune cell landscape dynamics aids in developing targeted therapies for infertility and contraception.
Collapse
|
6
|
Mukherjee AG, Gopalakrishnan AV. Anti-sperm Antibodies as an Increasing Threat to Male Fertility: Immunological Insights, Diagnostic and Therapeutic Strategies. Reprod Sci 2024; 31:3303-3322. [PMID: 38831152 DOI: 10.1007/s43032-024-01610-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
It is a fact that sperm possess antigenic properties. Substantial scientific research suggests that specific antibodies that attach to sperm antigens can induce infertility in both humans and other species. Antisperm antibodies (ASA) represent a significant etiology of infertility in humans, leading to immunoinfertility. The association between ASA and infertility is multifaceted. The observation of sperm agglutination, although not conclusive for the diagnosis of immunological infertility, may suggest the presence of ASA. Nevertheless, ASA may also manifest in the lack of any sperm agglutination. Managing ASA from an andrological perspective depends on the underlying cause and the specific approaches healthcare professionals adopt. The precise etiology of male infertility resulting from ASA remains unclear. Current research has examined the impact of ASA and its prevalence among infertile males to understand the relationship between ASA and changes in semen parameters. However, the findings have been inconclusive. Numerous techniques have been documented for the management of immunoinfertility. This review examines the importance of ASA in the context of infertility, encompassing the postulated mechanisms underlying the development of ASA, the various assays employed for detecting them, and the available treatments.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
7
|
Li H, Wang XR, Hu YF, Xiong YW, Zhu HL, Huang YC, Wang H. Advances in immunology of male reproductive toxicity induced by common environmental pollutants. ENVIRONMENT INTERNATIONAL 2024; 190:108898. [PMID: 39047547 DOI: 10.1016/j.envint.2024.108898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Humans are exposed to an ever-increasing number of environmental toxicants, some of which have gradually been identified as major risk factors for male reproductive health, even associated with male infertility. Male infertility is usually due to the reproductive system damage, which may be influenced by the exposure to contaminants such as heavy metals, plasticizers, along with genetics and lifestyle. Testicular immune microenvironment (TIM) is important in maintaining normal physiological functions of the testis, whether disturbed TIM after exposure to environmental toxicants could induce reproductive toxicity remains to be explored. Therefore, the current review aims to contribute to the further understanding of exposure and male infertility by characterizing environmental exposures and the effect on TIM. We first summarized the male reproductive toxicity phenotypes induced by common environmental pollutants. Contaminants including heavy metals and plastic additives and fine particulate matter (PM2.5), have been repetitively associated with male infertility, whereas emerging contaminants such as perfluoroalkyl substances and micro(nano)plastics have also been found to disrupt TIM and lead to male reproductive toxicity. We further reviewed the importance of TIM and its homeostasis in maintaining the normal physiological functions of the testis. Most importantly, we discussed the advances in immunology of male reproductive toxicity induced by metals and metalloids, plastic additives, persistent organic pollutants (POPs), micro(nano)plastic and PM2.5 to suggest the importance of reproductive immunotoxicology in the future study of environmental toxicants, but also contribute to the development of effective prevention and treatment strategies for mitigating adverse effects of environmental pollutants on human health.
Collapse
Affiliation(s)
- Hao Li
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230000, China
| | - Xin-Run Wang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230000, China
| | - Yi-Fan Hu
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230000, China
| | - Yong-Wei Xiong
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230000, China
| | - Hua-Long Zhu
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230000, China
| | - Yi-Chao Huang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, 230000, China.
| | - Hua Wang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, 230000, China.
| |
Collapse
|
8
|
Myers C, Atkins GR, Villarreal J, Sutton RB, Cornwall GA. The mouse epididymal amyloid matrix is a mammalian counterpart of a bacterial biofilm. iScience 2024; 27:110152. [PMID: 38974467 PMCID: PMC11225826 DOI: 10.1016/j.isci.2024.110152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/14/2024] [Accepted: 05/28/2024] [Indexed: 07/09/2024] Open
Abstract
The mouse epididymis is a long tubule connecting the testis to the vas deferens. Its primary functions are to mature spermatozoa into motile and fertile cells and to protect them from pathogens that ascend the male tract. We previously demonstrated that a functional extracellular amyloid matrix surrounds spermatozoa in the epididymal lumen and has host defense functions, properties not unlike that of an extracellular biofilm that encloses and protects a bacterial community. Here we show the epididymal amyloid matrix also structurally resembles a biofilm by containing eDNA, eRNA, and mucin-like polysaccharides. Further these structural components exhibit comparable behaviors and perform functions such as their counterparts in bacterial biofilms. Our studies suggest that nature has used the ancient building blocks of bacterial biofilms to form an analogous structure that nurtures and protects the mammalian male germline.
Collapse
Affiliation(s)
- Caitlyn Myers
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Georgia Rae Atkins
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Johanna Villarreal
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - R. Bryan Sutton
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Gail A. Cornwall
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
9
|
Xie S, Qin R, Zeng W, Li J, Lai Y. Pseudopregnant mice generated from Piwil1 deficiency sterile mice. PLoS One 2024; 19:e0296414. [PMID: 38771805 PMCID: PMC11108164 DOI: 10.1371/journal.pone.0296414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/12/2023] [Indexed: 05/23/2024] Open
Abstract
Vasectomized mice play a key role in the production of transgenic mice. However, vasectomy can cause great physical and psychological suffering to mice. Therefore, there is an urgent need to find a suitable replacement for vasectomized mice in the production of transgenic mice. In this study, we generated C57BL/6J mice (Piwil1 D633A-INS99, Piwil1mt/mt) with a 99-base insertion in the Miwi (Piwil1) gene using CRISPR/Cas9 technology and showed that Piwil1mt/+ heterozygous mice were normally fertile and that homozygous Piwil1mt/mt males were sterile and females were fertile. Transplantation of normal fertilized eggs into wild pseudopregnant females following mating with Piwil1mt/mt males produced no Piwil1mt/mt genotype offspring, and the number of offspring did not differ significantly from that of pseudopregnant mice following mating and breeding with ligated males. The CRISPR‒Cas9 system is available for generating Miwi-modified mice, and provides a powerful resource to replace ligated males in assisted reproduction research.
Collapse
Affiliation(s)
- Shuoshuo Xie
- Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Department of Medical Genetics, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ruixin Qin
- Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Department of Medical Genetics, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wentao Zeng
- Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Department of Medical Genetics, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Laboratory Animal Center, Animal Core facility, Key Laboratory of Model Animal, Nanjing, Jiangsu Province, China
| | - Jianmin Li
- Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Department of Medical Genetics, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Laboratory Animal Center, Animal Core facility, Key Laboratory of Model Animal, Nanjing, Jiangsu Province, China
| | - Yana Lai
- Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Department of Medical Genetics, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Laboratory Animal Center, Animal Core facility, Key Laboratory of Model Animal, Nanjing, Jiangsu Province, China
| |
Collapse
|
10
|
Yi J, Zhang H, Bao F, Chen Z, Zhong Y, Ye T, Chen X, Qian J, Tian M, Zhu M, Peng Z, Pan Z, Li J, Hu Z, Shen W, Xu J, Zhang X, Cai Y, Wu M, Liu H, Zhou J, Ouyang H. A pathological joint-liver axis mediated by matrikine-activated CD4 + T cells. Signal Transduct Target Ther 2024; 9:109. [PMID: 38714712 PMCID: PMC11076293 DOI: 10.1038/s41392-024-01819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/08/2024] [Accepted: 03/27/2024] [Indexed: 05/10/2024] Open
Abstract
The knee joint has long been considered a closed system. The pathological effects of joint diseases on distant organs have not been investigated. Herein, our clinical data showed that post-traumatic joint damage, combined with joint bleeding (hemarthrosis), exhibits a worse liver function compared with healthy control. With mouse model, hemarthrosis induces both cartilage degeneration and remote liver damage. Next, we found that hemarthrosis induces the upregulation in ratio and differentiation towards Th17 cells of CD4+ T cells in peripheral blood and spleen. Deletion of CD4+ T cells reverses hemarthrosis-induced liver damage. Degeneration of cartilage matrix induced by hemarthrosis upregulates serological type II collagen (COL II), which activates CD4+ T cells. Systemic application of a COL II antibody blocks the activation. Furthermore, bulk RNAseq and single-cell qPCR analysis revealed that the cartilage Akt pathway is inhibited by blood treatment. Intra-articular application of Akt activator blocks the cartilage degeneration and thus protects against the liver impairment in mouse and pig models. Taken together, our study revealed a pathological joint-liver axis mediated by matrikine-activated CD4+ T cells, which refreshes the organ-crosstalk axis and provides a new treatment target for hemarthrosis-related disease. Intra-articular bleeding induces cartilage degradation through down-reulation of cartilage Akt pathway. During this process, the soluble COL II released from the damaged cartilage can activate peripheral CD4+ T cells, differention into Th17 cells and secretion of IL-17, which consequently induces liver impairment. Intra-articular application of sc79 (inhibitor of Akt pathway) can prevent the cartilage damage as well as its peripheral influences.
Collapse
Affiliation(s)
- Junzhi Yi
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Zhang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
| | - Fangyuan Bao
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhichu Chen
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuliang Zhong
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianning Ye
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuri Chen
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyi Qian
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengya Tian
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Zhu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, The Key Laboratory of Cancer Molecular Cell Biology of Zhejiang Province, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Zhi Peng
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongyou Pan
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianyou Li
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Zihao Hu
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiliang Shen
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Jiaqi Xu
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianzhu Zhang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Youzhi Cai
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengjie Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
| | - Hua Liu
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Jing Zhou
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| | - Hongwei Ouyang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
11
|
Barrachina F, Ottino K, Elizagaray ML, Gervasi MG, Tu LJ, Markoulaki S, Spallanzani RG, Capen D, Brown D, Battistone MA. Regulatory T cells play a crucial role in maintaining sperm tolerance and male fertility. Proc Natl Acad Sci U S A 2023; 120:e2306797120. [PMID: 37676910 PMCID: PMC10500189 DOI: 10.1073/pnas.2306797120] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/31/2023] [Indexed: 09/09/2023] Open
Abstract
Regulatory T cells (Tregs) modulate tissue homeostatic processes and immune responses. Understanding tissue-Treg biology will contribute to developing precision-targeting treatment strategies. Here, we show that Tregs maintain the tolerogenic state of the testis and epididymis, where sperm are produced and mature. We found that Treg depletion induces severe autoimmune orchitis and epididymitis, manifested by an exacerbated immune cell infiltration [CD4 T cells, monocytes, and mononuclear phagocytes (MPs)] and the development of antisperm antibodies (ASA). In Treg-depleted mice, MPs increased projections toward the epididymal lumen as well as invading the lumen. ASA-bound sperm enhance sperm agglutination and might facilitate sperm phagocytosis. Tolerance breakdown impaired epididymal epithelial function and altered extracellular vesicle cargo, both of which play crucial roles in the acquisition of sperm fertilizing ability and subsequent embryo development. The affected mice had reduced sperm number and motility and severe fertility defects. Deciphering these immunoregulatory mechanisms may help to design new strategies to treat male infertility, as well as to identify potential targets for immunocontraception.
Collapse
Affiliation(s)
- Ferran Barrachina
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Kiera Ottino
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Maia Lina Elizagaray
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Maria Gracia Gervasi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA01003
- Genetically Engineered Models Center, Whitehead Institute of Biomedical Research, Cambridge, MA02142
| | - Leona J. Tu
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Styliani Markoulaki
- Genetically Engineered Models Center, Whitehead Institute of Biomedical Research, Cambridge, MA02142
| | - Raul G. Spallanzani
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA02115
| | - Diane Capen
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Dennis Brown
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Maria Agustina Battistone
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| |
Collapse
|
12
|
Wanjari UR, Gopalakrishnan AV. A review on immunological aspects in male reproduction: An immune cells and cytokines. J Reprod Immunol 2023; 158:103984. [PMID: 37390629 DOI: 10.1016/j.jri.2023.103984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/17/2023] [Accepted: 06/25/2023] [Indexed: 07/02/2023]
Abstract
The male reproductive system, particularly the male gamete, offers a unique barrier to the immune system. The growing germ cells in the testis need to be shielded from autoimmune damage. Hence the testis has to establish and sustain an immune-privileged milieu. Sertoli cells create this safe space, protected by the blood-testis barrier. Cytokines are a type of immune reaction that can positively and negatively affect male reproductive health. Inflammation, disease, and obesity are just a few physiological conditions for which cytokines mediate signals. They interact with steroidogenesis, shaping the adrenals and testes to produce the hormones needed for survival. In particular pathological condition, including autoimmune disorders, contains high levels of the same cytokines in semen that play an essential role in the immunomodulation of the male gonad. This review focuses on understanding the immunological role of cytokines in the control and development of male reproduction. Also, in maintaining male reproductive health and diseases linked with their aberrant function in the testis.
Collapse
Affiliation(s)
- Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
13
|
Doroftei B, Ilie OD, Maftei R, Scripcariu IS, Armeanu T, Stoian IL, Ilea C. A Narrative Review Discussing Vasectomy-Related Impact upon the Status of Oxidative Stress and Inflammation Biomarkers and Semen Microbiota. J Clin Med 2023; 12:jcm12072671. [PMID: 37048754 PMCID: PMC10095584 DOI: 10.3390/jcm12072671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Male contraceptive approaches besides tubal sterilization involve vasectomy and represent the method of choice among midlife men in developing countries thanks to many advantages. However, the subsidiary consequences of this intervention are insufficiently explored since the involved mechanisms may offer insight into a much more complex picture. Methods: Thus, in this manuscript, we aimed to reunite all available data by searching three separate academic database(s) (PubMed, Web of Knowledge, and Scopus) published in the past two decades by covering the interval 2000–2023 and using a predefined set of keywords and strings involving “oxidative stress” (OS), “inflammation”, and “semen microbiota” in combination with “humans”, “rats”, and “mice”. Results: By following all evidence that fits in the pre-, post-, and vasectomy reversal (VR) stages, we identified a total of n = 210 studies from which only n = 21 were finally included following two procedures of eligibility evaluation. Conclusions: The topic surrounding this intricate landscape has created debate since the current evidence is contradictory, limited, or does not exist. Starting from this consideration, we argue that further research is mandatory to decipher how a vasectomy might disturb homeostasis.
Collapse
Affiliation(s)
- Bogdan Doroftei
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street no 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street no 34, 700038 Iasi, Romania
- Origyn Fertility Center, Palace Street, no 3C, 700032 Iasi, Romania
| | - Ovidiu-Dumitru Ilie
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue no 20A, 700505 Iasi, Romania
| | - Radu Maftei
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street no 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street no 34, 700038 Iasi, Romania
- Origyn Fertility Center, Palace Street, no 3C, 700032 Iasi, Romania
| | - Ioana-Sadyie Scripcariu
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street no 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street no 34, 700038 Iasi, Romania
| | - Theodora Armeanu
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street no 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street no 34, 700038 Iasi, Romania
- Origyn Fertility Center, Palace Street, no 3C, 700032 Iasi, Romania
| | - Irina-Liviana Stoian
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street no 16, 700115 Iasi, Romania
| | - Ciprian Ilea
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street no 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street no 34, 700038 Iasi, Romania
| |
Collapse
|
14
|
Zhou M, Xie J, Kawase O, Nishikawa Y, Ji S, Zhu S, Cao S, Xuan X. Characterization of anti-erythrocyte and anti-platelet antibodies in hemolytic anemia and thrombocytopenia induced by Plasmodium spp. and Babesiaspp. infection in mice. Front Cell Infect Microbiol 2023; 13:1143138. [PMID: 37124034 PMCID: PMC10140361 DOI: 10.3389/fcimb.2023.1143138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/20/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Malaria and Babesiosis are acute zoonotic disease that caused by infection with the parasite in the phylum Apicomplexa. Severe anemia and thrombocytopenia are the most common hematological complication of malaria and babesiosis. However, the mechanisms involved have not been elucidated, and only a few researches focus on the possible role of anti-erythrocyte and anti-platelet antibodies. Methods In this study, the Plasmodium yoelii, P. chabaudi, Babesia microti and B. rodhaini infected SCID and ICR mice. The parasitemia, survival rate, platelet count, anti-platelet antibodies, and the level of IFN-γ and interleukin (IL) -10 was tested after infection. Furthermore, the P. yoelii, P. chabaudi, B. rodhaini and B. microti infected ICR mice were treated with artesunate and diminaze, the development of the anti-erythrocyte and anti-platelet antibodies in chronic stage were examined. At last, the murine red blood cell and platelet membrane proteins probed with auto-antibodies induced by P. yoelii, P. chabaudi, B. rodhaini, and B. microti infection were characterized by proteomic analysis. Results and discussion The high anti-platelet and anti-erythrocyte antibodies were detected in ICR mice after P. yoelii, P. chabaudi, B. rodhaini, and B. microti infection. Actin of murine erythrocyte and platelet is a common auto-antigen in Plasmodium and Babesia spp. infected mice. Our findings indicate that anti-erythrocyte and anti-platelet autoantibodies contribute to thrombocytopenia and anemia associated with Plasmodium spp. and Babesia spp. infection. This study will help to understand the mechanisms of malaria and babesiosis-related thrombocytopenia and hemolytic anemia.
Collapse
Affiliation(s)
- Mo Zhou
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Jiangsu Key Laboratory for High-tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, China
- Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, China
| | - Jun Xie
- Jiangsu Key Laboratory for High-tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, China
- Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, China
| | - Osamu Kawase
- Department of Biology, Premedical Sciences, Dokkyo Medical University, Tochigi, Japan
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Shengwei Ji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, China
- Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, China
| | - Shinuo Cao
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Jiangsu Key Laboratory for High-tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, China
- Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, China
- *Correspondence: Shinuo Cao, ; Xuenan Xuan,
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- *Correspondence: Shinuo Cao, ; Xuenan Xuan,
| |
Collapse
|
15
|
Myers C, Hastert MC, Cornwall GA. Host defense functions of the epididymal amyloid matrix. Mol Hum Reprod 2022; 28:6823549. [PMID: 36367296 PMCID: PMC9709822 DOI: 10.1093/molehr/gaac038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
The epididymal lumen is an immunologically distinct environment. It maintains tolerance for the naturally antigenic spermatozoa to allow their maturation into functional cells while simultaneously defending against pathogens that can ascend the male tract and cause infertility. We previously demonstrated that a nonpathological amyloid matrix that includes several cystatin-related epididymal spermatogenic (CRES) subgroup family members is distributed throughout the mouse epididymal lumen but its function was unknown. Here, we reveal a role for the epididymal amyloid matrix in host defense and demonstrate that the CRES amyloids and CD-1 mouse epididymal amyloid matrix exhibit potent antimicrobial activity against bacterial strains that commonly cause epididymal infections in men. We show the CRES and epididymal amyloids use several defense mechanisms including bacterial trapping, disruption of bacterial membranes and promotion of unique bacterial ghost-like structures. Remarkably, these antimicrobial actions varied depending on the bacterial strain indicating CRES amyloids and the epididymal amyloids elicit strain-specific host defense responses. We also demonstrate that the CRES monomer and immature assemblies of the epididymal amyloid transitioned into advanced structures in the presence of bacteria, suggesting their amyloid-forming/shape-shifting properties allows for a rapid reaction to a pathogen and provides an inherent plasticity in their host defense response. Together, our studies reveal new mechanistic insight into how the male reproductive tract defends against pathogens. Future studies using a mouse model for human epididymitis are needed to establish the epididymal amyloid responses to pathogens in vivo. Broadly, our studies provide an example of why nature has maintained the amyloid fold throughout evolution.
Collapse
Affiliation(s)
- Caitlyn Myers
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | | | - Gail A Cornwall
- Correspondence address. Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; E-mail:
| |
Collapse
|
16
|
Wang F, Zhang J, Wang Y, Chen Y, Han D. Viral tropism for the testis and sexual transmission. Front Immunol 2022; 13:1040172. [PMID: 36439102 PMCID: PMC9682072 DOI: 10.3389/fimmu.2022.1040172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 10/17/2023] Open
Abstract
The mammalian testis adopts an immune privileged environment to protect male germ cells from adverse autoimmune reaction. The testicular immune privileged status can be also hijacked by various microbial pathogens as a sanctuary to escape systemic immune surveillance. In particular, several viruses have a tropism for the testis. To overcome the immune privileged status and mount an effective local defense against invading viruses, testicular cells are well equipped with innate antiviral machinery. However, several viruses may persist an elongated duration in the testis and disrupt the local immune homeostasis, thereby impairing testicular functions and male fertility. Moreover, the viruses in the testis, as well as other organs of the male reproductive system, can shed to the semen, thus allowing sexual transmission to partners. Viral infection in the testis, which can impair male fertility and lead to sexual transmission, is a serious concern in research on known and on new emerging viruses. To provide references for our scientific peers, this article reviews research achievements and suggests future research focuses in the field.
Collapse
Affiliation(s)
| | | | | | - Yongmei Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Daishu Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Malko D, Elmzzahi T, Beyer M. Implications of regulatory T cells in non-lymphoid tissue physiology and pathophysiology. Front Immunol 2022; 13:954798. [PMID: 35936011 PMCID: PMC9354719 DOI: 10.3389/fimmu.2022.954798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/29/2022] [Indexed: 11/26/2022] Open
Abstract
Treg cells have been initially described as gatekeepers for the control of autoimmunity, as they can actively suppress the activity of other immune cells. However, their role goes beyond this as Treg cells further control immune responses during infections and tumor development. Furthermore, Treg cells can acquire additional properties for e.g., the control of tissue homeostasis. This is instructed by a specific differentiation program and the acquisition of effector properties unique to Treg cells in non-lymphoid tissues. These tissue Treg cells can further adapt to their tissue environment and acquire distinct functional properties through specific transcription factors activated by a combination of tissue derived factors, including tissue-specific antigens and cytokines. In this review, we will focus on recent findings extending our current understanding of the role and differentiation of these tissue Treg cells. As such we will highlight the importance of tissue Treg cells for tissue maintenance, regeneration, and repair in adipose tissue, muscle, CNS, liver, kidney, reproductive organs, and the lung.
Collapse
Affiliation(s)
- Darya Malko
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Tarek Elmzzahi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Marc Beyer
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Platform foR SinglE Cell GenomIcS and Epigenomics (PRECISE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and University of Bonn, Bonn, Germany
| |
Collapse
|
18
|
Roberts EK, Tardif S, Wright EA, Platt RN, Bradley RD, Hardy DM. Rapid divergence of a gamete recognition gene promoted macroevolution of Eutheria. Genome Biol 2022; 23:155. [PMID: 35821049 PMCID: PMC9275260 DOI: 10.1186/s13059-022-02721-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Speciation genes contribute disproportionately to species divergence, but few examples exist, especially in vertebrates. Here we test whether Zan, which encodes the sperm acrosomal protein zonadhesin that mediates species-specific adhesion to the egg's zona pellucida, is a speciation gene in placental mammals. RESULTS Genomic ontogeny reveals that Zan arose by repurposing of a stem vertebrate gene that was lost in multiple lineages but retained in Eutheria on acquiring a function in egg recognition. A 112-species Zan sequence phylogeny, representing 17 of 19 placental Orders, resolves all species into monophyletic groups corresponding to recognized Orders and Suborders, with <5% unsupported nodes. Three other rapidly evolving germ cell genes (Adam2, Zp2, and Prm1), a paralogous somatic cell gene (TectA), and a mitochondrial gene commonly used for phylogenetic analyses (Cytb) all yield trees with poorer resolution than the Zan tree and inferior topologies relative to a widely accepted mammalian supertree. Zan divergence by intense positive selection produces dramatic species differences in the protein's properties, with ordinal divergence rates generally reflecting species richness of placental Orders consistent with expectations for a speciation gene that acts across a wide range of taxa. Furthermore, Zan's combined phylogenetic utility and divergence exceeds those of all other genes known to have evolved in Eutheria by positive selection, including the only other mammalian speciation gene, Prdm9. CONCLUSIONS Species-specific egg recognition conferred by Zan's functional divergence served as a mode of prezygotic reproductive isolation that promoted the extraordinary adaptive radiation and success of Eutheria.
Collapse
Affiliation(s)
- Emma K. Roberts
- Department of Biological Sciences, Texas Tech University, Lubbock, TX USA
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX USA
| | - Steve Tardif
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX USA
- Reproductive Biology Division, JangoBio, Fitchburg, WI USA
| | - Emily A. Wright
- Department of Biological Sciences, Texas Tech University, Lubbock, TX USA
| | - Roy N. Platt
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, San Antonio, TX USA
| | - Robert D. Bradley
- Department of Biological Sciences, Texas Tech University, Lubbock, TX USA
- Natural Science Research Laboratory, Museum of Texas Tech University, Lubbock, TX USA
| | - Daniel M. Hardy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX USA
| |
Collapse
|
19
|
O'Donnell L, Smith LB, Rebourcet D. Sperm-specific proteins: new implications for diagnostic development and cancer immunotherapy. Curr Opin Cell Biol 2022; 77:102104. [PMID: 35671587 DOI: 10.1016/j.ceb.2022.102104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 11/03/2022]
Abstract
Spermatozoa are comprised of many unique proteins not expressed elsewhere. Sperm-specific proteins are first expressed at puberty, after the development of immune tolerance to self-antigens, and have been assumed to remain confined inside the seminiferous tubules, protected from immune cell recognition by various mechanisms of testicular immune privilege. However, new data has shown that sperm-specific proteins are released by the tubules into the surrounding interstitial fluid; from here they can contact immune cells, potentially promote immune tolerance, and enter the circulation. These new findings have clinical implications for diagnostics and therapeutics targeted at a specific class of proteins known as cancer-testis antigens (CTA), the opportunity to identify new communication pathways in the testis, and to discover new ways to monitor testis function.
Collapse
Affiliation(s)
- Liza O'Donnell
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, 3168, Victoria, Australia; Monash University, Clayton, 3168, Victoria, Australia.
| | - Lee B Smith
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; Griffith University, Parklands Drive, Southport, 4222, Queensland, Australia
| | - Diane Rebourcet
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
20
|
Harakal J, Qiao H, Wheeler K, Rival C, Paul AGA, Hardy DM, Cheng CY, Goldberg E, Tung KSK. Exposed and Sequestered Antigens in Testes and Their Protection by Regulatory T Cell-Dependent Systemic Tolerance. Front Immunol 2022; 13:809247. [PMID: 35693780 PMCID: PMC9179417 DOI: 10.3389/fimmu.2022.809247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/07/2022] [Indexed: 12/03/2022] Open
Abstract
Continuous exposure of tissue antigen (Ag) to the autoantigen-specific regulatory T cells (Treg) is required to maintain Treg-dependent systemic tolerance. Thus, testis autoantigens, previously considered as sequestered, may not be protected by systemic tolerance. We now document that the complete testis antigen sequestration is not valid. The haploid sperm Ag lactate dehydrogenase 3 (LDH3) is continuously exposed and not sequestered. It enters the residual body (RB) to egress from the seminiferous tubules and interact with circulating antibody (Ab). Some LDH3 also remains inside the sperm cytoplasmic droplets (CD). Treg-depletion in the DEREG mice that express diphtheria toxin receptor on the Foxp3 promoter results in spontaneous experimental autoimmune orchitis (EAO) and Ab to LDH3. Unlike the wild-type male mice, mice deficient in LDH3 (wild-type female or LDH3 NULL males) respond vigorously to LDH3 immunization. However, partial Treg depletion elevated the wild-type male LDH3 responses to the level of normal females. In contrast to LDH3, zonadhesin (ZAN) in the sperm acrosome displays properties of a sequestered Ag. However, when ZAN and other sperm Ag are exposed by vasectomy, they rapidly induce testis Ag-specific tolerance, which is terminated by partial Treg-depletion, leading to bilateral EAO and ZAN Ab response. We conclude that some testis/sperm Ag are normally exposed because of the unique testicular anatomy and physiology. The exposed Ag: 1) maintain normal Treg-dependent systemic tolerance, and 2) are pathogenic and serve as target Ag to initiate EAO. Unexpectedly, the sequestered Ags, normally non-tolerogenic, can orchestrate de novo Treg-dependent, systemic tolerance when exposed in vasectomy.
Collapse
Affiliation(s)
- Jessica Harakal
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, University of Virginia, Charlottesville, VA, United States
- Bierne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Hui Qiao
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
- Bierne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Karen Wheeler
- Department of Microbiology, University of Virginia, Charlottesville, VA, United States
- Bierne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Claudia Rival
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
- Bierne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Alberta G. A. Paul
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
- Bierne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Daniel M. Hardy
- Cell Biology and Biochemistry Department, Texas Tech University Health Science Center (HSC), Lubbock, TX, United States
| | - C. Yan Cheng
- Center for Biomedical Research, Population Council, New York, NY, United States
| | - Erwin Goldberg
- Molecular Biochemistry Department, Northwestern University, Evanstan, IL, United States
| | - Kenneth S. K. Tung
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, University of Virginia, Charlottesville, VA, United States
- Bierne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- *Correspondence: Kenneth S. K. Tung,
| |
Collapse
|
21
|
Sharpe MA, Baskin DS, Jenson AV, Baskin AM. Hijacking Sexual Immuno-Privilege in GBM-An Immuno-Evasion Strategy. Int J Mol Sci 2021; 22:10983. [PMID: 34681642 PMCID: PMC8536168 DOI: 10.3390/ijms222010983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 01/12/2023] Open
Abstract
Regulatory T-cells (Tregs) are immunosuppressive T-cells, which arrest immune responses to 'Self' tissues. Some immunosuppressive Tregs that recognize seminal epitopes suppress immune responses to the proteins in semen, in both men and women. We postulated that GBMs express reproductive-associated proteins to manipulate reproductive Tregs and to gain immune privilege. We analyzed four GBM transcriptome databases representing ≈900 tumors for hypoxia-responsive Tregs, steroidogenic pathways, and sperm/testicular and placenta-specific genes, stratifying tumors by expression. In silico analysis suggested that the presence of reproductive-associated Tregs in GBM tumors was associated with worse patient outcomes. These tumors have an androgenic signature, express male-specific antigens, and attract reproductive-associated Related Orphan Receptor C (RORC)-Treg immunosuppressive cells. GBM patient sera were interrogated for the presence of anti-sperm/testicular antibodies, along with age-matched controls, utilizing monkey testicle sections. GBM patient serum contained anti-sperm/testicular antibodies at levels > six-fold that of controls. Myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are associated with estrogenic tumors which appear to mimic placental tissue. We demonstrate that RORC-Tregs drive poor patient outcome, and Treg infiltration correlates strongly with androgen levels. Androgens support GBM expression of sperm/testicular proteins allowing Tregs from the patient's reproductive system to infiltrate the tumor. In contrast, estrogen appears responsible for MDSC/TAM immunosuppression.
Collapse
MESH Headings
- Androgens/metabolism
- Brain Neoplasms/immunology
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Databases, Factual
- Estrogens/metabolism
- Female
- Glioblastoma/immunology
- Glioblastoma/mortality
- Glioblastoma/pathology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kaplan-Meier Estimate
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Microglia/immunology
- Microglia/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Microenvironment
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
Collapse
Affiliation(s)
- Martyn A. Sharpe
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| | - David S. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Amanda V. Jenson
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| | - Alexandra M. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| |
Collapse
|
22
|
Dutta S, Sandhu N, Sengupta P, Alves MG, Henkel R, Agarwal A. Somatic-Immune Cells Crosstalk In-The-Making of Testicular Immune Privilege. Reprod Sci 2021; 29:2707-2718. [PMID: 34580844 DOI: 10.1007/s43032-021-00721-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/22/2021] [Indexed: 11/27/2022]
Abstract
Immunological infertility contributes significantly to the etiology of idiopathic male infertility. Shielding the spermatogenic cells from systemic immune responses is fundamental to secure normal production of spermatozoa. The body's immune system is tuned with the host self-components since the early postnatal period, while sperm first develops during puberty, thus rendering spermatogenic proteins as 'non-self' or 'antigenic.' Development of antibodies to these antigens elicits autoimmune responses affecting sperm motility, functions, and fertility. Therefore, the testes need to establish a specialized immune-privileged microenvironment to protect the allogenic germ cells by orchestration of various testicular cells and resident immune cells. This is achieved through sequestration of antigenic germ cells by blood-testis barrier and actions of various endocrine, paracrine, immune-suppressive, and immunomodulatory mechanisms. The various mechanisms are very complex and need conceptual integration to disclose the exact physiological scenario, and to facilitate detection and management of immunogenic infertility caused by disruption of testicular immune regulation. The present review aims to (a) discuss the components of testicular immune privilege; (b) explain testicular somatic and immune cell interactions in establishing and maintaining the testicular immune micro-environment; and (c) illustrate the integration of multiple mechanisms involved in the control of immune privilege of the testis.
Collapse
Affiliation(s)
- Sulagna Dutta
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom, Selangor , Malaysia
| | - Narpal Sandhu
- Molecular and Cellular Biology, University of California, Berkeley, CA, USA
| | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, Selangor , Malaysia
| | - Marco G Alves
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Ralf Henkel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Medical Bioscience, University of the Western Cape, Bellville, South Africa
- LogixX Pharma, Theale, Berkshire, UK
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
23
|
Fang Y, Su Y, Xu J, Hu Z, Zhao K, Liu C, Zhang H. Varicocele-Mediated Male Infertility: From the Perspective of Testicular Immunity and Inflammation. Front Immunol 2021; 12:729539. [PMID: 34531872 PMCID: PMC8438154 DOI: 10.3389/fimmu.2021.729539] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/16/2021] [Indexed: 01/14/2023] Open
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.
Collapse
Affiliation(s)
- Yiwei Fang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufang Su
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Hu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Warren BD, Ahn SH, Brittain KS, Nanjappa MK, Wang H, Wang J, Blanco G, Sanchez G, Fan Y, Petroff BK, Cooke PS, Petroff MG. Multiple Lesions Contribute to Infertility in Males Lacking Autoimmune Regulator. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1592-1609. [PMID: 34126085 PMCID: PMC8420865 DOI: 10.1016/j.ajpath.2021.05.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 04/21/2021] [Accepted: 05/18/2021] [Indexed: 11/15/2022]
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.
Collapse
Affiliation(s)
- Bryce D Warren
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Soo H Ahn
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Kathryn S Brittain
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan; Cell and Molecular Biology Graduate Program, College of Natural Sciences, Michigan State University, East Lansing, Michigan
| | - Manjunatha K Nanjappa
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Hao Wang
- Department of Computational Mathematics, Science and Engineering, College of Engineering, Michigan State University, East Lansing, Michigan
| | - Jianrong Wang
- Department of Computational Mathematics, Science and Engineering, College of Engineering, Michigan State University, East Lansing, Michigan
| | - Gustavo Blanco
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Gladis Sanchez
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Yong Fan
- Institute of Cellular Therapeutics, Alleghany Health Network, Pittsburgh, Pennsylvania
| | - Brian K Petroff
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Paul S Cooke
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Margaret G Petroff
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas; Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan; Cell and Molecular Biology Graduate Program, College of Natural Sciences, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
25
|
Hill MA, Kwon JH, Gerry B, Hardy WA, Walkowiak OA, Kavarana MN, Nadig SN, Rajab TK. Immune Privilege of Heart Valves. Front Immunol 2021; 12:731361. [PMID: 34447390 PMCID: PMC8383064 DOI: 10.3389/fimmu.2021.731361] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/22/2021] [Indexed: 01/22/2023] Open
Abstract
Immune privilege is an evolutionary adaptation that protects vital tissues with limited regenerative capacity from collateral damage by the immune response. Classical examples include the anterior chamber of the eye and the brain. More recently, the placenta, testes and articular cartilage were found to have similar immune privilege. What all of these tissues have in common is their vital function for evolutionary fitness and a limited regenerative capacity. Immune privilege is clinically relevant, because corneal transplantation and meniscal transplantation do not require immunosuppression. The heart valves also serve a vital function and have limited regenerative capacity after damage. Moreover, experimental and clinical evidence from heart valve transplantation suggests that the heart valves are spared from alloimmune injury. Here we review this evidence and propose the concept of heart valves as immune privileged sites. This concept has important clinical implications for heart valve transplantation.
Collapse
Affiliation(s)
- Morgan Ashley Hill
- College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jennie H Kwon
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Brielle Gerry
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - William A Hardy
- College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Olivia Agata Walkowiak
- College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Minoo N Kavarana
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Satish N Nadig
- Division of Transplant Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - T Konrad Rajab
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
26
|
O'Donnell L, Rebourcet D, Dagley LF, Sgaier R, Infusini G, O'Shaughnessy PJ, Chalmel F, Fietz D, Weidner W, Legrand JMD, Hobbs RM, McLachlan RI, Webb AI, Pilatz A, Diemer T, Smith LB, Stanton PG. Sperm proteins and cancer-testis antigens are released by the seminiferous tubules in mice and men. FASEB J 2021; 35:e21397. [PMID: 33565176 PMCID: PMC7898903 DOI: 10.1096/fj.202002484r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023]
Abstract
Sperm develop from puberty in the seminiferous tubules, inside the blood-testis barrier to prevent their recognition as "non-self" by the immune system, and it is widely assumed that human sperm-specific proteins cannot access the circulatory or immune systems. Sperm-specific proteins aberrantly expressed in cancer, known as cancer-testis antigens (CTAs), are often pursued as cancer biomarkers and therapeutic targets based on the assumption they are neoantigens absent from the circulation in healthy men. Here, we identify a wide range of germ cell-derived and sperm-specific proteins, including multiple CTAs, that are selectively deposited by the Sertoli cells of the adult mouse and human seminiferous tubules into testicular interstitial fluid (TIF) that is "outside" the blood-testis barrier. From TIF, the proteins can access the circulatory- and immune systems. Disruption of spermatogenesis decreases the abundance of these proteins in mouse TIF, and a sperm-specific CTA is significantly decreased in TIF from infertile men, suggesting that exposure of certain CTAs to the immune system could depend on fertility status. The results provide a rationale for the development of blood-based tests useful in the management of male infertility and indicate CTA candidates for cancer immunotherapy and biomarker development that could show sex-specific and male-fertility-related responses.
Collapse
Affiliation(s)
- Liza O'Donnell
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia.,Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Diane Rebourcet
- Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Laura F Dagley
- Walter and Eliza Hall Institute, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Raouda Sgaier
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia.,Department of Urology, Pediatric Urology and Andrology, Medical Faculty, Justus-Liebig-University Giessen, UKGM GmbH, Giessen, Germany
| | - Giuseppe Infusini
- Walter and Eliza Hall Institute, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Peter J O'Shaughnessy
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Frederic Chalmel
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, University Rennes, Rennes, France
| | - Daniela Fietz
- Institute for Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Wolfgang Weidner
- Department of Urology, Pediatric Urology and Andrology, Medical Faculty, Justus-Liebig-University Giessen, UKGM GmbH, Giessen, Germany
| | - Julien M D Legrand
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Robin M Hobbs
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Robert I McLachlan
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Andrew I Webb
- Walter and Eliza Hall Institute, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Adrian Pilatz
- Department of Urology, Pediatric Urology and Andrology, Medical Faculty, Justus-Liebig-University Giessen, UKGM GmbH, Giessen, Germany
| | - Thorsten Diemer
- Department of Urology, Pediatric Urology and Andrology, Medical Faculty, Justus-Liebig-University Giessen, UKGM GmbH, Giessen, Germany
| | - Lee B Smith
- Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia.,MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Peter G Stanton
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
27
|
Ye L, Huang W, Liu S, Cai S, Hong L, Xiao W, Thiele K, Zeng Y, Song M, Diao L. Impacts of Immunometabolism on Male Reproduction. Front Immunol 2021; 12:658432. [PMID: 34367130 PMCID: PMC8334851 DOI: 10.3389/fimmu.2021.658432] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
The physiological process of male reproduction relies on the orchestration of neuroendocrine, immune, and energy metabolism. Spermatogenesis is controlled by the hypothalamic-pituitary-testicular (HPT) axis, which modulates the production of gonadal steroid hormones in the testes. The immune cells and cytokines in testes provide a protective microenvironment for the development and maturation of germ cells. The metabolic cellular responses and processes in testes provide energy production and biosynthetic precursors to regulate germ cell development and control testicular immunity and inflammation. The metabolism of immune cells is crucial for both inflammatory and anti-inflammatory responses, which supposes to affect the spermatogenesis in testes. In this review, the role of immunometabolism in male reproduction will be highlighted. Obesity, metabolic dysfunction, such as type 2 diabetes mellitus, are well documented to impact male fertility; thus, their impacts on the immune cells distributed in testes will also be discussed. Finally, the potential significance of the medicine targeting the specific metabolic intermediates or immune metabolism checkpoints to improve male reproduction will also be reassessed.
Collapse
Affiliation(s)
- Lijun Ye
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Wensi Huang
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Su Liu
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Songchen Cai
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Ling Hong
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Weiqiang Xiao
- Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Kristin Thiele
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yong Zeng
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Mingzhe Song
- Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Lianghui Diao
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| |
Collapse
|
28
|
Mediterranean Pine Vole, Microtus duodecimcostatus: A Paradigm of an Opportunistic Breeder. Animals (Basel) 2021; 11:ani11061639. [PMID: 34205873 PMCID: PMC8228771 DOI: 10.3390/ani11061639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/23/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In temperate zones of the Earth, some mammalian species reproduce seasonally whereas others do it continuously. Other species are summer breeders in the north and winter breeders in the south. Thus, the reproductive pattern seems not to be a species-specific but a population-specific trait. We investigated the reproduction pattern of the Mediterranean pine vole, Microtus duodecimcostatus, in the area around the city of Granada in Southern Spain, and found that individuals living in wastelands reproduce seasonally whereas those living in close poplar plantations (just 8 km apart) reproduce throughout the year, as did voles captured in wastelands and kept in captivity. These animals represent thus a paradigm of an opportunistic breeder as particular individuals stop breeding or not, depending on the environmental conditions they face at any moment. Sexually inactive male voles undergo complete testis inactivation and their sperm production is halted. The immune system in active testes is depressed, a phenomenon known as “immune privilege” that protect germ cells from autoimmune attack. We studied gene activity in active and inactive testes and our results indicate that such an immune privilege is lost in inactive testes, suggesting an important role for this process during testis regression. Abstract Most mammalian species of the temperate zones of the Earth reproduce seasonally, existing a non-breeding period in which the gonads of both sexes undergo functional regression. It is widely accepted that photoperiod is the principal environmental cue controlling these seasonal changes, although several exceptions have been described in other mammalian species in which breeding depends on cues such as food or water availability. We studied the circannual reproductive cycle in males of the Mediterranean pine vole, Microtus duodecimcostatus, in the Southeastern Iberian Peninsula. Morphological, hormonal, functional, molecular and transcriptomic analyses were performed. As reported for populations of other species from the same geographic area, male voles captured in wastelands underwent seasonal testis regression in summer whereas, surprisingly, those living either in close poplar plantations or in our animal house reproduced throughout the year, showing that it is the microenvironment of a particular vole subpopulation what determines its reproductive status and that these animals are pure opportunistic, photoperiod-independent breeders. In addition, we show that several molecular pathways, including MAPK, are deregulated and that the testicular “immune privilege” is lost in the inactive testes, providing novel mechanisms linking seasonal testosterone reduction and testis regression.
Collapse
|
29
|
Gong J, Zeng Q, Yu D, Duan YG. T Lymphocytes and Testicular Immunity: A New Insight into Immune Regulation in Testes. Int J Mol Sci 2020; 22:ijms22010057. [PMID: 33374605 PMCID: PMC7793097 DOI: 10.3390/ijms22010057] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Jialei Gong
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Qunxiong Zeng
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
30
|
Pleuger C, Silva EJR, Pilatz A, Bhushan S, Meinhardt A. Differential Immune Response to Infection and Acute Inflammation Along the Epididymis. Front Immunol 2020; 11:599594. [PMID: 33329594 PMCID: PMC7729520 DOI: 10.3389/fimmu.2020.599594] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022] Open
Abstract
The epididymis is a tubular structure connecting the vas deferens to the testis. This organ consists of three main regions—caput, corpus, and cauda—that face opposing immunological tasks. A means of combating invading pathogens is required in the distally located cauda, where there is a risk of ascending bacterial infections originating from the urethra. Meanwhile, immune tolerance is necessary at the caput, where spermatozoa with immunogenic neo-antigens originate from the testis. Consistently, when challenged with live bacteria or inflammatory stimuli, the cauda elicits a much stronger immune response and inflammatory-inflicted damage than the caput. At the cellular level, a role for diverse and strategically positioned mononuclear phagocytes is emerging. At the mechanistic level, differential expression of immunoprotective and immunomodulatory mediators has been detected between the three main regions of the epididymis. In this review, we summarize the current state of knowledge about region-specific immunological characteristics and unveil possible underlying mechanisms on cellular and molecular levels. Improved understanding of the different immunological microenvironments is the basis for an improved therapy and counseling of patients with epididymal infections.
Collapse
Affiliation(s)
- Christiane Pleuger
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Erick José Ramo Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Adrian Pilatz
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University Giessen, Giessen, Germany.,Department of Urology, Pediatric Urology and Andrology, University Hospital, Justus-Liebig-University Giessen, Giessen, Germany
| | - Sudhanshu Bhushan
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Andreas Meinhardt
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
31
|
Stocks BT, Mirabal JR, Payne K, Lipshultz LI. It Is Time We Rethink Our Approach to Enhancing Access to Male Infertility Care. CURRENT SEXUAL HEALTH REPORTS 2020. [DOI: 10.1007/s11930-020-00275-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
32
|
Lustig L, Guazzone VA, Theas MS, Pleuger C, Jacobo P, Pérez CV, Meinhardt A, Fijak M. Pathomechanisms of Autoimmune Based Testicular Inflammation. Front Immunol 2020; 11:583135. [PMID: 33101310 PMCID: PMC7546798 DOI: 10.3389/fimmu.2020.583135] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022] Open
Abstract
Infection and inflammation of the male reproductive tract are relevant causes of infertility. Inflammatory damage occurs in the special immunosuppressive microenvironment of the testis, a hallmark termed testicular immune privilege, which allows tolerance to neo-antigens from developing germ cells appearing at puberty, long after the establishment of systemic immune tolerance. Experimental autoimmune orchitis (EAO) is a well-established rodent model of chronic testicular inflammation and organ specific autoimmunity that offers a valuable in vivo tool to investigate the pathological and molecular mechanisms leading to the breakdown of the testicular immune privilege. The disease is characterized by the infiltration of the interstitium by immune cells (mainly macrophages, dendritic cells, and T cells), formation of autoantibodies against testicular antigens, production of pro-inflammatory mediators such as NO, MCP1, TNFα, IL6, or activins and dysregulation of steroidogenesis with reduced levels of serum testosterone. EAO leads to sloughing of germ cells, atrophic seminiferous tubules and fibrotic remodeling, parameters all found similarly to changes in human biopsies from infertile patients with inflammatory infiltrates. Interestingly, testosterone supplementation during the course of EAO leads to expansion of the regulatory T cell population and inhibition of disease development. Knowledge of EAO pathogenesis aims to contribute to a better understanding of human testicular autoimmune disease as an essential prerequisite for improved diagnosis and treatment.
Collapse
Affiliation(s)
- Livia Lustig
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas (INBIOMED), Consejo Nacional de Investigaciones Científicas y Tècnicas (CONICET), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Vanesa A Guazzone
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas (INBIOMED), Consejo Nacional de Investigaciones Científicas y Tècnicas (CONICET), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - María S Theas
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas (INBIOMED), Consejo Nacional de Investigaciones Científicas y Tècnicas (CONICET), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Christiane Pleuger
- Department of Anatomy and Cell Biology, Justus-Liebig University Giessen, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Patricia Jacobo
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas (INBIOMED), Consejo Nacional de Investigaciones Científicas y Tècnicas (CONICET), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Cecilia V Pérez
- Instituto de Investigaciones Biomédicas (INBIOMED), Consejo Nacional de Investigaciones Científicas y Tècnicas (CONICET), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig University Giessen, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus-Liebig University Giessen, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
33
|
Bhushan S, Theas MS, Guazzone VA, Jacobo P, Wang M, Fijak M, Meinhardt A, Lustig L. Immune Cell Subtypes and Their Function in the Testis. Front Immunol 2020; 11:583304. [PMID: 33101311 PMCID: PMC7554629 DOI: 10.3389/fimmu.2020.583304] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022] Open
Abstract
Immunoregulation in the testis is characterized by a balance between immuno-suppression (or immune privilege) and the ability to react to infections and inflammation. In this review, we analyze the phenotypes of the various immune cell subtypes present in the testis, and how their functions change between homeostatic and inflammatory conditions. Starting with testicular macrophages, we explore how this heterogeneous population is shaped by the testicular microenvironment to ensure immune privilege. We then describe how dendritic cells exhibit a tolerogenic status under normal conditions, but proliferate, mature and then stimulate effector T-cell expansion under inflammatory conditions. Finally, we outline the two T-cell populations in the testis: CD4+/CD8+ αβ T cells and CD4+/CD8+ Foxp3+ regulatory T cells and describe the distribution and function of mast cells. All these cells help modulate innate immunity and regulate the immune response. By improving our understanding of immune cell behavior in the testis under normal and inflammatory conditions, we will be better placed to evaluate testis impairment due to immune mechanisms in affected patients.
Collapse
Affiliation(s)
- Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus-Liebig University Giessen, Giessen, Germany.,Hessian Center of Reproductive Medicine, Justus-Leibig-University Giessen, Giessen, Germany
| | - María S Theas
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Vanesa A Guazzone
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Patricia Jacobo
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Ming Wang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus-Liebig University Giessen, Giessen, Germany.,Hessian Center of Reproductive Medicine, Justus-Leibig-University Giessen, Giessen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig University Giessen, Giessen, Germany.,Hessian Center of Reproductive Medicine, Justus-Leibig-University Giessen, Giessen, Germany
| | - Livia Lustig
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|
34
|
Voisin A, Saez F, Drevet JR, Guiton R. The epididymal immune balance: a key to preserving male fertility. Asian J Androl 2020; 21:531-539. [PMID: 30924450 PMCID: PMC6859654 DOI: 10.4103/aja.aja_11_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Up to 15% of male infertility has an immunological origin, either due to repetitive infections or to autoimmune responses mainly affecting the epididymis, prostate, and testis. Clinical observations and epidemiological data clearly contradict the idea that the testis confers immune protection to the whole male genital tract. As a consequence, the epididymis, in which posttesticular spermatozoa mature and are stored, has raised some interest in recent years when it comes to its immune mechanisms. Indeed, sperm cells are produced at puberty, long after the establishment of self-tolerance, and they possess unique surface proteins that cannot be recognized as self. These are potential targets of the immune system, with the risk of inducing autoantibodies and consequently male infertility. Epididymal immunity is based on a finely tuned equilibrium between efficient immune responses to pathogens and strong tolerance to sperm cells. These processes rely on incompletely described molecules and cell types. This review compiles recent studies focusing on the immune cell types populating the epididymis, and proposes hypothetical models of the organization of epididymal immunity with a special emphasis on the immune response, while also discussing important aspects of the epididymal immune regulation such as tolerance and tumour control.
Collapse
Affiliation(s)
- Allison Voisin
- Team Mechanisms of Posttesticular Infertility, GReD Laboratory, CNRS UMR 6293 - INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand 63001, France
| | - Fabrice Saez
- Team Mechanisms of Posttesticular Infertility, GReD Laboratory, CNRS UMR 6293 - INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand 63001, France
| | - Joël R Drevet
- Team Mechanisms of Posttesticular Infertility, GReD Laboratory, CNRS UMR 6293 - INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand 63001, France
| | - Rachel Guiton
- Team Mechanisms of Posttesticular Infertility, GReD Laboratory, CNRS UMR 6293 - INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand 63001, France
| |
Collapse
|
35
|
Gualdoni GS, Jacobo PV, Sobarzo CM, Pérez CV, Matzkin ME, Höcht C, Frungieri MB, Hill M, Anegon I, Lustig L, Guazzone VA. Role of indoleamine 2,3-dioxygenase in testicular immune-privilege. Sci Rep 2019; 9:15919. [PMID: 31685866 PMCID: PMC6828782 DOI: 10.1038/s41598-019-52192-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 10/12/2019] [Indexed: 02/07/2023] Open
Abstract
Male meiotic germ cell including the spermatozoa represent a great challenge to the immune system, as they appear long after the establishment of normal immune tolerance mechanisms. The capacity of the testes to tolerate autoantigenic germ cells as well as survival of allogeneic organ engrafted in the testicular interstitium have led to consider the testis an immunologically privileged site. Disruption of this immune privilege following trauma, tumor, or autoimmune orchitis often results in male infertility. Strong evidence indicates that indoleamine 2,3-dioxygenase (IDO) has been implicated in fetal and allograft tolerance, tumor immune resistance, and regulation of autoimmune diseases. IDO and tryptophan 2,3-dioxygenase (TDO) catalyze the same rate-limiting step of tryptophan metabolism along a common pathway, which leads to tryptophan starvation and generation of catabolites collectively known as kynurenines. However, the relevance of tryptophan metabolism in testis pathophysiology has not yet been explored. Here we assessed the in vivo role of IDO/TDO in experimental autoimmune orchitis (EAO), a model of autoimmune testicular inflammation and immunologically impaired spermatogenesis. EAO was induced in adult Wistar rats with testicular homogenate and adjuvants. Control (C) rats injected with saline and adjuvants and normal untreated rats (N) were also studied. mRNA expression of IDO decreased in whole testes and in isolated Sertoli cells during EAO. TDO and IDO localization and level of expression in the testis were analyzed by immunostaining and Western blot. TDO is expressed in granulomas from EAO rats, and similar protein levels were observed in N, C, and EAO groups. IDO was detected in mononuclear and endothelial cells and reduced IDO expression was detected in EAO group compared to N and C rats. This phenomenon was concomitant with a significant reduction of IDO activity in EAO testis measured by tryptophan and kynurenine concentrations (HPLC). Finally, in vivo inhibition of IDO with 1-methyl-tryptophan increased severity of the disease, demonstrating down regulation of IDO-based tolerance when testicular immune regulation was disrupted. We present evidence that an IDO-based mechanism is involved in testicular immune privilege.
Collapse
Affiliation(s)
- Gisela S Gualdoni
- Universidad de Buenos Aires (UBA), Facultad de Medicina, Departamento de Biología Celular e Histología/Unidad Académica II., Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - Patricia V Jacobo
- Universidad de Buenos Aires (UBA), Facultad de Medicina, Departamento de Biología Celular e Histología/Unidad Académica II., Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - Cristian M Sobarzo
- Universidad de Buenos Aires (UBA), Facultad de Medicina, Departamento de Biología Celular e Histología/Unidad Académica II., Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - Cecilia V Pérez
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - María E Matzkin
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Ciudad Autónoma de Buenos Aires, C1428ADN, Argentina
| | - Christian Höcht
- Cátedra de Farmacología. Facultad de Farmacia y Bioquímica, UBA, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - Mónica B Frungieri
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Ciudad Autónoma de Buenos Aires, C1428ADN, Argentina
| | - Marcelo Hill
- Laboratory of Immunoregulation and Inflammation, Institut Pasteur de Montevideo, 11400, Montevideo, Uruguay.,Immunobiology Department, Faculty of Medicine, University of the Republic, 11800, Montevideo, Uruguay
| | - Ignacio Anegon
- Inserm, Université de Nantes, Centre de Recherche en Transplantation et Immunologie, Nantes, France, INSERM UMR 1064, France
| | - Livia Lustig
- Universidad de Buenos Aires (UBA), Facultad de Medicina, Departamento de Biología Celular e Histología/Unidad Académica II., Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina
| | - Vanesa A Guazzone
- Universidad de Buenos Aires (UBA), Facultad de Medicina, Departamento de Biología Celular e Histología/Unidad Académica II., Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Ciudad Autónoma de Buenos Aires, C1121ABG, Argentina.
| |
Collapse
|
36
|
Wang LL, Li ZH, Duan YG, Yuan SQ, Mor G, Liao AH. Identification of programmed cell death 1 and its ligand in the testicular tissue of mice. Am J Reprod Immunol 2018; 81:e13079. [PMID: 30578744 DOI: 10.1111/aji.13079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/23/2018] [Accepted: 12/07/2018] [Indexed: 12/18/2022] Open
Abstract
PROBLEM This study aims to determine the expression and localization of programmed cell death 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) in the testes of mice at different developmental stages. METHOD OF STUDY By means of RT-qPCR, Western blot and immunofluorescence, the expression and localization of PD-1 and PD-L1 were detected in the testicular tissues of mice at different postnatal times: P7, P14, P21, P28, P35, and adulthood. Meanwhile, the level of soluble PD-L1 (sPD-L1) was evaluated by ELISA in the testicular interstitial fluid (IF) of the adult mice, culture supernatants of TM4 cell lines (Sertoli cells lines), and primary Sertoli cells at P14. RESULTS Pd-1 mRNA levels were unexpectedly low. From P7 to P21, there was limited PD-1 protein detected while PD-1 was evident at P28 and afterward at significantly higher levels than at P14 and P21 (P < 0.05). Despite being found in the interstitial area at P7, P14, and P21, PD-1 was also detected in the germ cells of the seminiferous tubules after P28. Pd-l1 mRNA exhibited age-related changes, peaking at P21, while PD-L1 protein was constitutively expressed at any stage, specifically localized in the nucleus of Sertoli cells. Moreover, the level of sPD-L1 in IF was significantly higher than that in the culture supernatants of both TM4 and primary Sertoli cells at P14. CONCLUSIONS PD-1 and PD-L1 were present in the testicular tissue of adult mice. The expression and localization of PD-1 fluctuated with age, and PD-1 was mainly localized to advanced germ cells, suggesting that it may play a role in spermiogenesis. PD-L1 was constitutively expressed in the nucleus of Sertoli cells, which could secrete sPD-L1 into the testicular interstitial space and thus may be involved in testicular immune privilege.
Collapse
Affiliation(s)
- Li-Ling Wang
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Hui Li
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong - ShenZhen Hospital, Guangdong, China
| | - Shui-Qiao Yuan
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gil Mor
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Reproductive Immunology Unit, Department of Obstetrics Gynecology and Reproductive Science, Yale University School of Medicine, New Haven, Connecticut
| | - Ai-Hua Liao
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Jacobo P. The role of regulatory T Cells in autoimmune orchitis. Andrologia 2018; 50:e13092. [DOI: 10.1111/and.13092] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/06/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Patricia Jacobo
- Departmental and Institutional Affiliation, Instituto de Investigaciones Biomédicas, UBA-CONICET, Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| |
Collapse
|
38
|
Ethylene dimethane sulfonate (EDS) ablation of Leydig cells in adult rat depletes testosterone resulting in epididymal sperm granuloma: Testosterone replacement prevents granuloma formation. Reprod Biol 2018; 19:89-99. [PMID: 30528522 DOI: 10.1016/j.repbio.2018.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/13/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022]
Abstract
Sperm granuloma may develop in the epididymis following vasectomy or chemical insults. Inflammation due to sperm granuloma causes abdominal and scrotal pain. Prolonged and persistent inflammation in the epididymis due to sperm granuloma may lead to infertility. Extravasation of germ cells into the interstitium of epididymis following damage of the epididymal epithelium is one of the primary reasons for sperm granuloma-associated pathology. Since testosterone is vital for the maintenance of epididymal epithelium, we investigated the pathology of sperm granuloma and its relationship with testosterone. Adult rats were treated with a Leydig cell-specific toxicant ethylene dimethane sulfonate (EDS) to eliminate testosterone. At 7 days post-EDS, disrupted epididymal epithelium and sperm granuloma were observed in the caput epididymis. Sperm granuloma and caput were collagen-filled indicating fibrosis. Numerous round apoptotic cells were localized inside the caput lumen and dispersed through the sperm granuloma. Tnp1 (round spermatid marker) was significantly higher in the epididymis of the EDS-treated group compared to controls suggesting the apoptotic cells were round spermatids. Increases in CD68+ macrophages and T cells (CD4 and CD8) support an inflammatory immune infiltration in post-EDS epididymis. However, testosterone replacement following EDS prevented the sperm granuloma-associated pathology. We suggest that the immune response in the sperm granuloma may be due to the increased numbers of apoptotic round spermatids or other testicular tissue components that may be released, in addition to the regression of epididymal epithelium due to testosterone loss. Thus, testosterone replacement prevents EDS-induced sperm granuloma and ameliorates sperm granuloma-associated pathology.
Collapse
|
39
|
Pierucci-Alves F, Midura-Kiela MT, Fleming SD, Schultz BD, Kiela PR. Transforming Growth Factor Beta Signaling in Dendritic Cells Is Required for Immunotolerance to Sperm in the Epididymis. Front Immunol 2018; 9:1882. [PMID: 30166986 PMCID: PMC6105693 DOI: 10.3389/fimmu.2018.01882] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 07/30/2018] [Indexed: 01/16/2023] Open
Abstract
The epididymis exhibits a less restrictive physical blood–tissue barrier than the testis and, while numerous immunosuppressive factors have been identified in the latter, no mechanisms for epididymal immunotolerance have been identified to date. Therefore, data are currently insufficient to explain how the immune system tolerates the extremely large load of novel antigens expressed on sperm, which become present in the male body after puberty, i.e., long after central tolerance was established. This study tested the hypothesis that transforming growth factor beta (TGFβ) signaling in dendritic cells (DCs) is required for immunotolerance to sperm located in the epididymis, and that male mice lacking TGFβ signaling in DCs would develop severe epididymal inflammation. To test this, we employed adult Tgfbr2ΔDC males, which exhibit a significant reduction of Tgfbr2 expression and TGFβ signaling in DCs, as reported previously. Results show that Tgfbr2ΔDC males exhibit sperm-specific immune response and severe epididymal leukocytosis. This phenotype is consistent with epididymal loss of immunotolerance to sperm and suggests that TGFβ signaling in DCs is a factor required for a non-inflammatory steady state in the epididymis, and therefore for male tract homeostasis and function.
Collapse
Affiliation(s)
| | | | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Bruce D Schultz
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| | - Pawel R Kiela
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Immunobiology, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
40
|
Fijak M, Pilatz A, Hedger MP, Nicolas N, Bhushan S, Michel V, Tung KSK, Schuppe HC, Meinhardt A. Infectious, inflammatory and 'autoimmune' male factor infertility: how do rodent models inform clinical practice? Hum Reprod Update 2018; 24:416-441. [PMID: 29648649 PMCID: PMC6016649 DOI: 10.1093/humupd/dmy009] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/02/2018] [Accepted: 03/10/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Infection and inflammation of the reproductive tract are significant causes of male factor infertility. Ascending infections caused by sexually transmitted bacteria or urinary tract pathogens represent the most frequent aetiology of epididymo-orchitis, but viral, haematogenous dissemination is also a contributory factor. Limitations in adequate diagnosis and therapy reflect an obvious need for further understanding of human epididymal and testicular immunopathologies and their contribution to infertility. A major obstacle for advancing our knowledge is the limited access to suitable tissue samples. Similarly, the key events in the inflammatory or autoimmune pathologies affecting human male fertility are poorly amenable to close examination. Moreover, the disease processes generally have occurred long before the patient attends the clinic for fertility assessment. In this regard, data obtained from experimental animal models and respective comparative analyses have shown promise to overcome these restrictions in humans. OBJECTIVE AND RATIONALE This narrative review will focus on male fertility disturbances caused by infection and inflammation, and the usefulness of the most frequently applied animal models to study these conditions. SEARCH METHODS An extensive search in Medline database was performed without restrictions until January 2018 using the following search terms: 'infection' and/or 'inflammation' and 'testis' and/or 'epididymis', 'infection' and/or 'inflammation' and 'male genital tract', 'male infertility', 'orchitis', 'epididymitis', 'experimental autoimmune' and 'orchitis' or 'epididymitis' or 'epididymo-orchitis', antisperm antibodies', 'vasectomy'. In addition to that, reference lists of primary and review articles were reviewed for additional publications independently by each author. Selected articles were verified by each two separate authors and discrepancies discussed within the team. OUTCOMES There is clear evidence that models mimicking testicular and/or epididymal inflammation and infection have been instructive in a better understanding of the mechanisms of disease initiation and progression. In this regard, rodent models of acute bacterial epididymitis best reflect the clinical situation in terms of mimicking the infection pathway, pathogens selected and the damage, such as fibrotic transformation, observed. Similarly, animal models of acute testicular and epididymal inflammation using lipopolysaccharides show impairment of reproduction, endocrine function and histological tissue architecture, also seen in men. Autoimmune responses can be studied in models of experimental autoimmune orchitis (EAO) and vasectomy. In particular, the early stages of EAO development showing inflammatory responses in the form of peritubular lymphocytic infiltrates, thickening of the lamina propria of affected tubules, production of autoantibodies against testicular antigens or secretion of pro-inflammatory mediators, replicate observations in testicular sperm extraction samples of patients with 'mixed atrophy' of spermatogenesis. Vasectomy, in the form of sperm antibodies and chronic inflammation, can also be studied in animal models, providing valuable insights into the human response. WIDER IMPLICATIONS This is the first comprehensive review of rodent models of both infectious and autoimmune disease of testis/epididymis, and their clinical implications, i.e. their importance in understanding male infertility related to infectious and non-infectious/autoimmune disease of the reproductive organs.
Collapse
Affiliation(s)
- Monika Fijak
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
| | - Adrian Pilatz
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University of Giessen, Germany
| | - Mark P Hedger
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, Australia
| | - Nour Nicolas
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, Australia
| | - Sudhanshu Bhushan
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
| | - Vera Michel
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
| | - Kenneth S K Tung
- Departments of Pathology and Microbiology, Beirne Carter Center for Immunology Research, University of Virginia, 345 Crispell Drive, Charlottesville, VA, USA
| | - Hans-Christian Schuppe
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University of Giessen, Germany
| | - Andreas Meinhardt
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, Australia
| |
Collapse
|
41
|
Kawase O, Jimbo M. Detection of sperm-reactive antibodies in wild sika deer and identification of the sperm antigens. J Vet Med Sci 2018; 80:802-809. [PMID: 29553063 PMCID: PMC5989026 DOI: 10.1292/jvms.17-0660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Antisperm antibodies potentially inhibit sperm functions causing the sterility in humans and experimentally treated animals. However, there is no information about antisperm antibodies
emerging spontaneously in wildlife. In this study, we searched for the sperm-reactive antibodies, spontaneously produced in wild sika deer (Cervus nippon), and identified
the sperm antigens. We collected 529 fecal masses of sika deer in Japanese cities, from which we extracted the mucosal antibodies to test them for reactivities to deer sperm proteins by
ELISA. Two of the extracts contained IgAs that were highly reactive to the sperm proteins. The molecular weights of the active IgAs, partially purified by DEAE-sephadex A-50, were estimated
at more than 100 kDa, suggesting that the IgAs evaded drastic digestion in the gastrointestinal tract. Two-dimensional electrophoresis and immunoblotting detected three major antigens, and
the following LC-MS/MS analysis identified them as alpha-enolase, phosphoglycerate kinase 2 and acrosin-binding protein. The antibodies were cross-reactive to a recombinant human
acrosin-binding protein. To our knowledge, this is the first research to find that the sperm-reactive antibodies are produced spontaneously in wildlife and they recognize a common antigen
found in humans.
Collapse
Affiliation(s)
- Osamu Kawase
- Department of Biology, Premedical Sciences, Dokkyo Medical University, Mibu-machi, Shimotsuga-gun, Tochigi 321-0293, Japan
| | - Mitsuru Jimbo
- Department of Marine Biosciences, School of Marine Biosciences, Kitasato University, Kitasato, Sagamihara, Kanagawa 252-0373, Japan
| |
Collapse
|
42
|
Cormier N, McGlone JJ, Leszyk J, Hardy DM. Immunocontraceptive target repertoire defined by systematic identification of sperm membrane alloantigens in a single species. PLoS One 2018; 13:e0190891. [PMID: 29342175 PMCID: PMC5771590 DOI: 10.1371/journal.pone.0190891] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 12/21/2017] [Indexed: 12/13/2022] Open
Abstract
Sperm competence in animal fertilization requires the collective activities of numerous sperm-specific proteins that are typically alloimmunogenic in females. Consequently, sperm membrane alloantigens are potential targets for contraceptives that act by blocking the proteins' functions in gamete interactions. Here we used a targeted proteomics approach to identify the major alloantigens in swine sperm membranes and lipid rafts, and thereby systematically defined the repertoire of these sperm-specific proteins in a single species. Gilts with high alloantibody reactivity to proteins in sperm membranes or lipid rafts produced fewer offspring (73% decrease) than adjuvant-only or nonimmune control animals. Alloantisera recognized more than 20 potentially unique sperm membrane proteins and five sperm lipid raft proteins resolved on two-dimensional immunoblots with or without prior enrichment by anion exchange chromatography. Dominant sperm membrane alloantigens identified by mass spectrometry included the ADAMs fertilin α, fertilin ß, and cyritestin. Less abundant alloantigens included ATP synthase F1 β subunit, myo-inositol monophosphatase-1, and zymogen granule membrane glycoprotein-2. Immunodominant sperm lipid raft alloantigens included SAMP14, lymphocyte antigen 6K, and the epididymal sperm protein E12. Of the fifteen unique membrane alloantigens identified, eleven were known sperm-specific proteins with uncertain functions in fertilization, and four were not previously suspected to exist as sperm-specific isoforms. De novo sequences of tryptic peptides from sperm membrane alloantigen "M6" displayed no evident homology to known proteins, so is a newly discovered sperm-specific gene product in swine. We conclude that alloimmunizing gilts with sperm membranes or lipid rafts evokes formation of antibodies to a relatively small number of dominant alloantigens that include known and novel sperm-specific proteins with possible functions in fertilization and potential utility as targets for immunocontraception.
Collapse
Affiliation(s)
- Nathaly Cormier
- Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - John J. McGlone
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, Texas, United States of America
| | - John Leszyk
- Proteomic and Mass Spectrometry Facility and Department of Biochemistry & Pharmacology, University of Massachusetts Medical School, Shrewsbury, Massachusetts, United States of America
| | - Daniel M. Hardy
- Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
- * E-mail:
| |
Collapse
|
43
|
Wang LL, Li ZH, Hu XH, Muyayalo KP, Zhang YH, Liao AH. The roles of the PD-1/PD-L1 pathway at immunologically privileged sites. Am J Reprod Immunol 2017; 78. [PMID: 28585775 DOI: 10.1111/aji.12710] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/20/2017] [Indexed: 12/14/2022] Open
Abstract
The PD-1/PD-L1 pathway plays a vital role in the maintenance of peripheral tolerance, promoting the development and function of regulatory T cells, and maintaining the quiescence of autoreactive T cells. Abnormalities in this inhibitory pathway are involved in the pathogenesis of some disorders such as tumours, autoimmune diseases, pregnancy complications, and transplantation rejection. Immune privilege represents a special immunological condition, where foreign antigens can be tolerated and do not elicit an immune response. The anterior chamber of the eye, central nervous system, testis, pregnant uterus, and hair follicles are all regarded as immune-privileged sites in humans. Numerous studies show that the PD-1/PD-L1 pathway contributes to the maintenance of the immune-privileged microenvironment. In this review, we will mainly focus on the roles of the PD-1/PD-L1 pathway in the anterior chamber of the eye, brain and testis, as well as further investigations in testis.
Collapse
Affiliation(s)
- Li-Ling Wang
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Hui Li
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Hui Hu
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kahinho P Muyayalo
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong-Hong Zhang
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ai-Hua Liao
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Mono-(2-ethylhexyl) phthalate-induced Sertoli cell injury stimulates the production of pro-inflammatory cytokines in Fischer 344 rats. Reprod Toxicol 2017; 69:150-158. [PMID: 28238932 DOI: 10.1016/j.reprotox.2017.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/06/2017] [Accepted: 02/21/2017] [Indexed: 11/22/2022]
Abstract
Exposure of rodents to the Sertoli cell (SC) toxicant mono-(2-ethylhexyl) phthalate (MEHP) has been reported to trigger an infiltration of macrophages into the testis in an age- and species-dependent manner. Here we challenge the hypothesis that the peripubertal rat-specific infiltration of macrophages after MEHP exposure is due, in part, to an increase in SC-specific inflammatory cytokine expression. To rule out that germ cell(GC) apoptosis itself is responsible for macrophage recruitment, rats were exposed to a direct GC toxicant, methoxyacetic acid (MAA), but no infiltration of macrophages was observed. Next, mRNA levels of inflammatory cytokines were evaluated after MEHP exposure. IL-1α, IL-6, and MCP-1 expression were increased in vivo and correlated with macrophage infiltration in a species-specific manner. Additionally, IL-6 and MCP-1 expression was increased in SC-GC co-cultures and ASC-17D SCs. These results indicate that MEHP-injury in pubertal rats specifically stimulates secretion of pro-inflammatory cytokines and alters the immune microenvironment.
Collapse
|
45
|
Tung KSK, Harakal J, Qiao H, Rival C, Li JCH, Paul AGA, Wheeler K, Pramoonjago P, Grafer CM, Sun W, Sampson RD, Wong EWP, Reddi PP, Deshmukh US, Hardy DM, Tang H, Cheng CY, Goldberg E. Egress of sperm autoantigen from seminiferous tubules maintains systemic tolerance. J Clin Invest 2017; 127:1046-1060. [PMID: 28218625 DOI: 10.1172/jci89927] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/21/2016] [Indexed: 12/29/2022] Open
Abstract
Autoimmune responses to meiotic germ cell antigens (MGCA) that are expressed on sperm and testis occur in human infertility and after vasectomy. Many MGCA are also expressed as cancer/testis antigens (CTA) in human cancers, but the tolerance status of MGCA has not been investigated. MGCA are considered to be uniformly immunogenic and nontolerogenic, and the prevailing view posits that MGCA are sequestered behind the Sertoli cell barrier in seminiferous tubules. Here, we have shown that only some murine MGCA are sequestered. Nonsequestered MCGA (NS-MGCA) egressed from normal tubules, as evidenced by their ability to interact with systemically injected antibodies and form localized immune complexes outside the Sertoli cell barrier. NS-MGCA derived from cell fragments that were discarded by spermatids during spermiation. They egressed as cargo in residual bodies and maintained Treg-dependent physiological tolerance. In contrast, sequestered MGCA (S-MGCA) were undetectable in residual bodies and were nontolerogenic. Unlike postvasectomy autoantibodies, which have been shown to mainly target S-MGCA, autoantibodies produced by normal mice with transient Treg depletion that developed autoimmune orchitis exclusively targeted NS-MGCA. We conclude that spermiation, a physiological checkpoint in spermatogenesis, determines the egress and tolerogenicity of MGCA. Our findings will affect target antigen selection in testis and sperm autoimmunity and the immune responses to CTA in male cancer patients.
Collapse
|
46
|
Terayama H, Hirai S, Naito M, Qu N, Katagiri C, Nagahori K, Hayashi S, Sasaki H, Moriya S, Hiramoto M, Miyazawa K, Hatayama N, Li ZL, Sakabe K, Matsushita M, Itoh M. Specific autoantigens identified by sera obtained from mice that are immunized with testicular germ cells alone. Sci Rep 2016; 6:35599. [PMID: 27752123 PMCID: PMC5067510 DOI: 10.1038/srep35599] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 10/04/2016] [Indexed: 02/08/2023] Open
Abstract
There are various autoimmunogenic antigens (AIs) in testicular germ cells (TGCs) recognized as foreign by the body's immune system. However, there is little information of TGC-specific AIs being available. The aim of this study is to identify TGC-specific AIs. We have previously established that immunization using viable syngeneic TGC can also induce murine experimental autoimmune orchitis (EAO) without using any adjuvant. This study is to identify TGC-specific AIs by TGC liquid chromatography-tandem mass spectrometry analysis, followed by two-dimensional gel electrophoresis that reacted with serum IgG from EAO mice. In this study, we identified 11 TGC-specific AIs that reacted with serum from EAO mice. Real-time RT-PCR analysis showed that the mRNA expressions of seven TGC-specific AIs were significantly higher in only mature testis compared to other organs. Moreover, the recombinant proteins of identified 10 (except unnamed protein) TGC-specific AIs were created by using human embryonic kidney 293 (HEK293) cells and these antigencities were reconfirmed by Western blot using EAO serum reaction. These results indicated Atp6v1a, Hsc70t, Fbp1 and Dazap1 were candidates for TGC-specific AIs. Identification of these AIs will facilitate new approaches for understanding infertility and cancer pathogenesis and may provide a basis for the development of novel therapies.
Collapse
Affiliation(s)
- Hayato Terayama
- Department of Anatomy, Division of Basic Medical Science, Tokai University School of Medicine, Kanagawa, Japan.,Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Shuichi Hirai
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan.,Department of Anatomy, Aichi Medical University, Aichi, Japan
| | - Munekazu Naito
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan.,Department of Anatomy, Aichi Medical University, Aichi, Japan
| | - Ning Qu
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Chiaki Katagiri
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kenta Nagahori
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Shogo Hayashi
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Hiraku Sasaki
- Department of Health Science, School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Shota Moriya
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Masaki Hiramoto
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Naoyuki Hatayama
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan.,Department of Anatomy, Aichi Medical University, Aichi, Japan
| | - Zhong-Lian Li
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Kou Sakabe
- Department of Anatomy, Division of Basic Medical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Masayuki Matsushita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Masahiro Itoh
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
47
|
Oh YS, Na EJ, Gye MC. Effects of bilateral vasectomy on the interleukin 1 system in mouse epididymis. Am J Reprod Immunol 2016; 76:235-42. [PMID: 27476761 DOI: 10.1111/aji.12548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/13/2016] [Indexed: 11/26/2022] Open
Abstract
PROBLEM Regional difference in the inflammatory response in vasectomized (VAX) epididymis remains unraveled. METHOD OF STUDY Epididymal expression of interleukin 1α (IL1α), IL1β, IL1 receptor antagonist (IL1ra), IL1 receptor 1 (IL1R1), IL6, IL10, tumor necrosis factor α (TNF-α), and transforming growth factor-β1 (TGF-β1) was examined in bilateral VAX mice by real-time RT-PCR and immunohistochemistry. RESULTS IL1α, IL1β, IL1ra, and IL1R1 were expressed in the epididymal epithelia of normal mice. Following VAX, IL1R1 mRNA was not changed, but TNF-α, IL10, IL1α, IL1β, and IL1ra mRNA were significantly upregulated in whole epididymis, whereas IL6 and TGF-β1 mRNA were significantly increased in corpus and cauda but not in caput epididymis. Consistently, IL1α, IL1β, and IL1ra immunoreactivities were visibly increased in epididymis following VAX. CONCLUSION Following VAX, IL1α, IL1β, IL1ra, IL10, and TNF-α may mediate immune reaction in whole epididymis, whereas IL6 and TGF-β1 may mediate regionally different immune response primarily in the lower part of epididymis.
Collapse
Affiliation(s)
- Yeong Seok Oh
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Eun Ju Na
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Myung Chan Gye
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| |
Collapse
|
48
|
Schagdarsurengin U, Western P, Steger K, Meinhardt A. Developmental origins of male subfertility: role of infection, inflammation, and environmental factors. Semin Immunopathol 2016; 38:765-781. [PMID: 27315198 DOI: 10.1007/s00281-016-0576-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/06/2016] [Indexed: 12/28/2022]
Abstract
Male gamete development begins with the specification of primordial cells in the epiblast of the early embryo and is not complete until spermatozoa mature in the epididymis of adult males. This protracted developmental process involves extensive alteration of the paternal germline epigenome. Initially, epigenetic reprogramming in fetal germ cells results in removal of most DNA methylation, including parent-specific epigenetic information. The germ cells then establish sex-specific epigenetic information through de novo methylation and undergo spermatogenesis. Chromatin in haploid germ cells is repackaged into protamines during spermiogenesis, providing further widespread epigenetic reorganization. Finally, after fertilization, epigenetic reprogramming in the preimplantation embryo is necessary for regaining totipotency. These events provide substantial windows during which epigenetic errors either may be corrected or may occur in the germline. There is now increasing evidence that environmental factors such as exposure to toxicants, the parents' and individual's diet, and even infectious and inflammatory events in the male reproductive tract may influence epigenetic reprogramming. This, together with other damage inflicted on the germline chromatin, may result in negative consequences for fertility and health. Large epidemiological birth cohort studies have yielded insight into possible causative environmental factors. Together with experimental animal studies, a clearer view of environmental impacts on fetal development and their intergenerational and even transgenerational effects on reproductive health has emerged and is reviewed in this article.
Collapse
Affiliation(s)
- Undraga Schagdarsurengin
- Department of Urology, Pediatric Urology and Andrology, Section Molecular Andrology, Justus-Liebig University of Giessen, Giessen, Germany
| | - Patrick Western
- Centre for Genetic Diseases, Hudson Institute for Medical Research and Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
| | - Klaus Steger
- Department of Urology, Pediatric Urology and Andrology, Section Molecular Andrology, Justus-Liebig University of Giessen, Giessen, Germany
| | - Andreas Meinhardt
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig University of Giessen, Aulweg 123, 35385, Giessen, Germany.
| |
Collapse
|
49
|
Harakal J, Rival C, Qiao H, Tung KS. Regulatory T Cells Control Th2-Dominant Murine Autoimmune Gastritis. THE JOURNAL OF IMMUNOLOGY 2016; 197:27-41. [PMID: 27259856 DOI: 10.4049/jimmunol.1502344] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/18/2016] [Indexed: 12/12/2022]
Abstract
Pernicious anemia and gastric carcinoma are serious sequelae of autoimmune gastritis (AIG). Our study indicates that in adult C57BL/6-DEREG mice expressing a transgenic diphtheria toxin receptor under the Foxp3 promoter, transient regulatory T cell (Treg) depletion results in long-lasting AIG associated with both H(+)K(+)ATPase and intrinsic factor autoantibody responses. Although functional Tregs emerge over time during AIG occurrence, the effector T cells rapidly become less susceptible to Treg-mediated suppression. Whereas previous studies have implicated dysregulated Th1 cell responses in AIG pathogenesis, eosinophils have been detected in gastric biopsy specimens from patients with AIG. Indeed, AIG in DEREG mice is associated with strong Th2 cell responses, including dominant IgG1 autoantibodies, elevated serum IgE, increased Th2 cytokine production, and eosinophil infiltration in the stomach-draining lymph nodes. In addition, the stomachs exhibit severe mucosal and muscular hypertrophy, parietal cell loss, mucinous epithelial cell metaplasia, and massive eosinophilic inflammation. Notably, the Th2 responses and gastritis severity are significantly ameliorated in IL-4- or eosinophil-deficient mice. Furthermore, expansion of both Th2-promoting IFN regulatory factor 4(+) programmed death ligand 2(+) dendritic cells and ILT3(+) rebounded Tregs was detected after transient Treg depletion. Collectively, these data suggest that Tregs maintain physiological tolerance to clinically relevant gastric autoantigens, and Th2 responses can be a pathogenic mechanism in AIG.
Collapse
Affiliation(s)
- Jessica Harakal
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908; and
| | - Claudia Rival
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908; and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Hui Qiao
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908; and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Kenneth S Tung
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908; and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
50
|
Abstract
The onslaught of foreign antigens carried by spermatozoa into the epididymis, an organ that has not demonstrated immune privilege, a decade or more after the establishment of central immune tolerance presents a unique biological challenge. Historically, the physical confinement of spermatozoa to the epididymal tubule enforced by a tightly interwoven wall of epithelial cells was considered sufficient enough to prevent cross talk between gametes and the immune system and, ultimately, autoimmune destruction. The discovery of an intricate arrangement of mononuclear phagocytes (MPs) comprising dendritic cells and macrophages in the murine epididymis suggests that we may have underestimated the existence of a sophisticated mucosal immune system in the posttesticular environment. This review consolidates our current knowledge of the physiology of MPs in the steady state epididymis and speculates on possible interactions between auto-antigenic spermatozoa, pathogens and the immune system by drawing on what is known about the immune system in the intestinal mucosa. Ultimately, further investigation will provide valuable information regarding the origins of pathologies arising as a result of autoimmune or inflammatory responses in the epididymis, including epididymitis and infertility.
Collapse
Affiliation(s)
- Nicolas Da Silva
- Massachusetts General Hospital and Harvard Medical School, Division of Nephrology, Center for Systems Biology, Boston, Massachusetts, USA
| | | |
Collapse
|