1
|
Sadek A, Khramtsova Y, Yushkov B. Mast Cells as a Component of Spermatogonial Stem Cells' Microenvironment. Int J Mol Sci 2024; 25:13177. [PMID: 39684887 DOI: 10.3390/ijms252313177] [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: 10/09/2024] [Revised: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
The formation of mature spermatozoa originates from spermatogonial stem cells (SSCs) located near the basement membrane of the seminiferous tubules. This developmental process, known as spermatogenesis, is tightly regulated to ensure continuous sperm production. A critical aspect of this regulation is the balance between SSC differentiation and self-renewal, which is directed by various factors guiding SSCs in either of these two directions. The SSC niche, defined functionally rather than anatomically, includes all factors necessary for SSC maintenance. These factors are produced by cells surrounding the SSC niche, collectively creating the microenvironment of the seminiferous tubules. Coordination between the cells in this microenvironment is essential for the proper function of the SSC niche and successful spermatogenesis. Testicular mast cells (MCs) significantly influence the regulation of this niche, as they contain various biologically active substances that regulate a wide range of physiological processes and contribute to different pathological conditions affecting fertility. This review explores the effects of testicular MCs on SSCs, their role in regulating spermatogenesis under normal and pathological conditions, and their interactions with other components of the testicular microenvironment, with a focus on their potentially critical impact on spermatogenesis and male fertility.
Collapse
Affiliation(s)
- Ali Sadek
- Department of Biology and Fundamental Medicine, Ural Federal University Named After the First President of Russia B. N. Yeltsin, 620002 Ekaterinburg, Russia
- Central Experimental Laboratory of Biotechnology, Institute of Medical Cell Technologies of the Sverdlovsk Region, 620026 Ekaterinburg, Russia
| | - Yulia Khramtsova
- Laboratory of Immunophysiology and Immunopharmacology, Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, 620049 Ekaterinburg, Russia
| | - Boris Yushkov
- Laboratory of Immunophysiology and Immunopharmacology, Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, 620049 Ekaterinburg, Russia
| |
Collapse
|
2
|
Bazzano MV, Köninger A, Solano ME. Beyond defence: Immune architects of ovarian health and disease. Semin Immunopathol 2024; 46:11. [PMID: 39134914 PMCID: PMC11319434 DOI: 10.1007/s00281-024-01021-w] [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: 03/14/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024]
Abstract
Throughout the individual's reproductive period of life the ovary undergoes continues changes, including cyclic processes of cell death, tissue regeneration, proliferation, and vascularization. Tissue-resident leucocytes particularly macrophages, play a crucial role in shaping ovarian function and maintaining homeostasis. Macrophages crucially promote angiogenesis in the follicles and corpora lutea, thereby supporting steroidogenesis. Recent research on macrophage origins and early tissue seeding has unveiled significant insights into their role in early organogenesis, e.g. in the testis. Here, we review evidence about the prenatal ovarian seeding of leucocytes, primarily macrophages with angiogenic profiles, and its connection to gametogenesis. In the prenatal ovary, germ cells proliferate, form cysts, and undergo changes that, following waves of apoptosis, give rice to the oocytes contained in primordial follicles. These follicles constitute the ovarian reserve that lasts throughout the female's reproductive life. Simultaneously, yolk-sac-derived primitive macrophages colonizing the early ovary are gradually replaced or outnumbered by monocyte-derived fetal macrophages. However, the cues indicating how macrophage colonization and follicle assembly are related are elusive. Macrophages may contribute to organogenesis by promoting early vasculogenesis. Whether macrophages contribute to ovarian lymphangiogenesis or innervation is still unknown. Ovarian organogenesis and gametogenesis are vulnerable to prenatal insults, potentially programming dysfunction in later life, as observed in polycystic ovary syndrome. Experimental and, more sparsely, epidemiological evidence suggest that adverse stimuli during pregnancy can program defective folliculogenesis or a diminished follicle reserve in the offspring. While the ovary is highly sensitive to inflammation, the involvement of local immune responses in programming ovarian health and disease remains to be thoroughly investigated.
Collapse
Affiliation(s)
- Maria Victoria Bazzano
- Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053, Regensburg, Germany
| | - Angela Köninger
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049, Regensburg, Germany
| | - Maria Emilia Solano
- Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053, Regensburg, Germany.
| |
Collapse
|
3
|
Singh M, Brooks A, Toofan P, McLuckie K. Selection of appropriate non-clinical animal models to ensure translatability of novel AAV-gene therapies to the clinic. Gene Ther 2024; 31:56-63. [PMID: 37612361 DOI: 10.1038/s41434-023-00417-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/19/2023] [Accepted: 08/04/2023] [Indexed: 08/25/2023]
Abstract
Gene Therapy Medicinal Products consist of a recombinant nucleic acid intended for the modulation or manipulation of a genetic sequence. A single administration of a novel gene therapy has the potential to be curative, with a durable long-term benefit to patients. Adeno-associated viral vectors have become the viral vector of choice for in vivo delivery of therapeutic transgenes as they are mildly immunogenic, can effectively transduce a variety of human tissues and cells, and have low levels of genomic integration. Central to the effective translation of data generated in discovery studies to the clinic is the selection of appropriate animal species for pivotal non-clinical studies. This review aims to support the selection of appropriate animal models for non-clinical studies to advance the development of novel adeno-associated virus gene therapies.
Collapse
Affiliation(s)
- Mark Singh
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, UK.
| | - Andrew Brooks
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, UK
| | - Parto Toofan
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, UK
| | - Keith McLuckie
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, UK
| |
Collapse
|
4
|
Zhang J, Song H, Fan X, He S, Yin W, Peng Z, Zhai X, Yang K, Gong H, Wang Z, Ping Y, Zhang S, Li RK, Xie J. Optimizing human endometrial mesenchymal stem cells for maximal induction of angiogenesis. Mol Cell Biochem 2022; 478:1191-1204. [PMID: 36266491 DOI: 10.1007/s11010-022-04572-4] [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: 03/21/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022]
Abstract
Human endometrial mesenchymal stem cells (hEMSCs) have been shown to promote neo-vascularization; however, its angiogenic function lessens with age. To determine the optimal conditions for maximizing hEMSC angiogenic capacity, we examined the effects of serial passaging on hEMSC activity. hEMSCs were cultured from passages (P) 3, 6, 9, and 12, and analyzed for proliferation, migration, differentiation and senescence, as well as their capacity to induce angiogenesis. The results showed that hEMSC proliferation and migration significantly decreased after P12. Furthermore, hEMSC differentiation into adipogenic and osteogenic lineages, as well as their proangiogenic capacity, gradually decreased from P9-12, while senescence only occurred after P12. Evaluation of angiogenic-related protein levels showed that both transforming growth factor β2 and Tie-2 was significantly reduced in hEMSCs at P12, compared to P3, possibly serving as the basis behind their lowered angiogenic capacity. Furthermore, in vivo angiogenesis evaluation with Matrigel plug assay showed that the optimal hEMSC to HUVEC ratio, for maximizing vessel formation, was 1:4. This study showed that hEMSC passaging was associated with lowered cellular functioning, bringing them closer to a senescent phenotype, especially after P12, thereby defining the optimal time period for cultivating fully functional hEMSCs for therapeutic applications.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China
| | - Huifang Song
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China
- Department of Anatomy, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xuemei Fan
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China
- Department of Endocrinology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, The Third Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Sheng He
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China
- Department of Radiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wenjuan Yin
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China
| | - Zexu Peng
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China
| | - Xiaoyan Zhai
- Department of Anatomy, Shanxi University of Chinese Medicine, Yuci, Taiyuan, 030001, Shanxi, China
| | - Kun Yang
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China
- Department of Endocrinology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, The Third Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Hui Gong
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China
| | - Zhijun Wang
- Xi'an International Medical Center Plastic Surgery Hospital, Xi'an, 710068, Shaanxi, China
| | - Yi Ping
- Department of Gynecology and Obstetrics, The Second Hospital of Shanxi Medical, University, Taiyuan, 030001, Shanxi, China
| | - Sanyuan Zhang
- Department of Gynecology and Obstetrics, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Ren-Ke Li
- Toronto General Hospital Research Institute, University Health Network, 101 College Street, Toronto, ON, M5G 1L7, Canada.
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, The Laboratory of Stem Cell Regenerative Medicine Research, Shanxi Key Laboratory of Birth, Defect and Cell Regeneration, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
5
|
Nguyen TT, Hulme J, Tran HD, Vo TK, Vo GV. The potential impact of COVID-19 on male reproductive health. J Endocrinol Invest 2022; 45:1483-1495. [PMID: 35181849 PMCID: PMC8856879 DOI: 10.1007/s40618-022-01764-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
The SARS-CoV-2 virus continues to overwhelm health care systems impairing human to human social and economic interactions. Invasion or damage to the male reproductive system is one of the documented outcomes of viral infection. Existing studies have reported that SARS-CoV-2 may contribute to this loss in relation to inflammatory responses and the formation of cytokine storms in COVID-19 patients. Although direct infection of the testes and entry of SARS-CoV-2 into semen as well as subsequent consequences on the male reproductive system need to be studied more systematically, warnings from two organising ASRM and SART for prospective parents when infected with SARS-CoV-2 should be considered. In the context of an increasingly complex pandemic, this review provides preliminary examples of the potential impact of COVID-19 on male reproductive health and guidance for prospective parents currently infected with or recovering from SARS-CoV-2.
Collapse
Affiliation(s)
- T T Nguyen
- Faculty of Pharmacy, HUTECH University, Ho Chi Minh City, 700000, Vietnam
| | - J Hulme
- Department of BioNano Technology, Gachon University, Seongnam-si, 461-701, Republic of Korea.
| | - H D Tran
- Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University-Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam
- Vietnam National University-Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam
| | - T K Vo
- Ministry of Culture, Sports and Tourism, Vietnam Sports Hospital, Hanoi, 100000, Vietnam
- Department of Sports Medicine, University of Medicine and Pharmacy (VNU-UMP), Vietnam National University Hanoi, Hanoi, 100000, Vietnam
| | - G V Vo
- Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University-Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
- Department of Biomedical Engineering, School of Medicine, Vietnam National University-Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
- Vietnam National University-Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
6
|
Reproductive immune microenvironment. J Reprod Immunol 2022; 152:103654. [PMID: 35728349 DOI: 10.1016/j.jri.2022.103654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/16/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022]
Abstract
About 10 %-12 % of couples in the world suffer from infertility, and immunological factors are being paid more and more attention. Attempts to induce peripheral immune tolerance in pregnant women by injecting husband cells have been widely promoted, but ultimately proved unsuccessful. Over the past two decades, our understanding of how the immune system is involved in gametogenesis and embryonic development, especially in early pregnancy, has undergone a major shift, going from the periphery to the local area of reproductive tissue. However, a holistic overview of immune responses in reproductive organs and tissues is currently lacking. Here, we further highlight the importance of regional immunity research for understanding reproductive health by reviewing the research mileage of the testis, ovary, and uterine immune microenvironment. We propose the concept of "reproductive immune microenvironment (RIM)" by summarizing the common features and basic functions of the tissue microenvironment in which immune cells reside, including the interstitial space of the testis, the ovarian stroma and the endometrium. The establishment of the concept of RIM not only focuses on the comprehensive description of the immune response in reproductive tissues, but also provides a macroscopic perspective for a deeper understanding of the immune etiology of reproductive system-related diseases.
Collapse
|
7
|
Routy JP, Dupuy FP, Lin J, Isnard S. More than a Gender Issue: Testis as a Distinctive HIV Reservoir and Its Implication for Viral Eradication. Methods Mol Biol 2022; 2407:173-186. [PMID: 34985665 DOI: 10.1007/978-1-0716-1871-4_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Early establishment of HIV reservoir represents the main impediment to an HIV cure. Mainly composed of infected memory CD4 T-cells and macrophages, HIV reservoirs are found in several organs including lymph nodes, gut, and testes. In men, and as seen in brain and eyes, testes represent a distinctive organ characterized by an immune privilege, allowing the tolerance of spermatozoa which only develop after puberty, long after the establishment of systemic immunity. The immune privilege of testes relies on a strict testis-blood barrier, and a local immunosuppressive environment. Testes has been described as reservoir for several viruses including Ebola, Zika, and HIV. Indeed, HIV reservoirs were detected in tested viremic and virally suppressed donor taking antiretroviral therapy (ART). Herein, we discuss the distinctive environment found in human testes and describe a validated method allowing the characterization and quantification of HIV-infected CD4 T-cells in human testes. Using mechanical and enzymatic treatment, cells can be extracted from human testis samples. Characterization of those cells can be performed by flow cytometry and HIV reservoir quantification performed by nested qPCR after flow cytometry sorting.
Collapse
Affiliation(s)
- Jean-Pierre Routy
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada.
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada.
- Division of Hematology, McGill University Health Centre, Montreal, QC, Canada.
| | - Franck P Dupuy
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - John Lin
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Stéphane Isnard
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
- CIHR Canadian HIV Trials Network (CTN), Vancouver, BC, Canada
| |
Collapse
|
8
|
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
|
9
|
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: 16] [Impact Index Per Article: 4.0] [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
|
10
|
Washburn RL, Hibler T, Thompson LA, Kaur G, Dufour JM. Therapeutic application of Sertoli cells for treatment of various diseases. Semin Cell Dev Biol 2021; 121:10-23. [PMID: 33910764 DOI: 10.1016/j.semcdb.2021.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022]
Abstract
Sertoli cells (SCs) are immune privileged cells found in the testis that function to immunologically protect maturing germ cells from immune destruction. This immune protection is due to the blood-testis-barrier, which prevents infiltration of cytotoxic immune cells and antibodies, and SC production of immunomodulatory factors, that favor a tolerogenic environment. The ability of SCs to create an immune privileged environment has led to the exploration of their potential use in the treatment of various diseases. SCs have been utilized to create a tolerogenic ectopic microenvironment, to protect co-grafted cells, and to deliver therapeutic proteins through gene therapy. To date, numerous studies have reported the potential use of SCs for the treatment of diabetes, neurodegenerative disorders, and restoration of spermatogenesis. Additionally, SCs have been investigated as a delivery vehicle for therapeutic products to treat other diseases like Laron syndrome, muscular dystrophy, and infections. This review will provide an overview of these therapeutic applications.
Collapse
Affiliation(s)
- Rachel L Washburn
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Taylor Hibler
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Lea Ann Thompson
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Gurvinder Kaur
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Jannette M Dufour
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
11
|
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: 14] [Impact Index Per Article: 2.8] [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
|
12
|
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: 46] [Impact Index Per Article: 9.2] [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
|
13
|
Bertolini M, McElwee K, Gilhar A, Bulfone‐Paus S, Paus R. Hair follicle immune privilege and its collapse in alopecia areata. Exp Dermatol 2020; 29:703-725. [DOI: 10.1111/exd.14155] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/18/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Affiliation(s)
| | - Kevin McElwee
- Monasterium Laboratory Münster Germany
- Centre for Skin Sciences University of Bradford Bradford UK
- Department of Dermatology and Skin Science University of British Columbia Vancouver British Columbia Canada
| | - Amos Gilhar
- Laboratory for Skin Research Rappaport Faculty of Medicine Technion‐Israel Institute of Technology Haifa Israel
| | - Silvia Bulfone‐Paus
- Monasterium Laboratory Münster Germany
- Centre for Dermatology Research University of Manchester and NIHR Manchester Biomedical Research Centre Manchester UK
| | - Ralf Paus
- Monasterium Laboratory Münster Germany
- Centre for Dermatology Research University of Manchester and NIHR Manchester Biomedical Research Centre Manchester UK
- Dr. Philip Frost Department of Dermatology & Cutaneous Surgery University of Miami Miller School of Medicine Miami FL USA
| |
Collapse
|
14
|
Batra V, Dagar K, Nayak S, Kumaresan A, Kumar R, Datta TK. A Higher Abundance of O-Linked Glycans Confers a Selective Advantage to High Fertile Buffalo Spermatozoa for Immune-Evasion From Neutrophils. Front Immunol 2020; 11:1928. [PMID: 32983120 PMCID: PMC7483552 DOI: 10.3389/fimmu.2020.01928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/17/2020] [Indexed: 12/26/2022] Open
Abstract
The glycans on the plasma membrane of cells manifest as the glycocalyx, which serves as an information-rich frontier that is directly in contact with its immediate milieu. The glycoconjugates (GCs) that adorn most of the mammalian cells are also abundant in gametes, especially the spermatozoa where they perform unique reproduction-specific functions e.g., inter-cellular recognition and communication. This study aimed to implicate the sperm glycosylation pattern as one of the factors responsible for low conception rates observed in buffalo bulls. We hypothesized that a differential abundance of glycans exists on the spermatozoa from bulls of contrasting fertilizing abilities endowing them with differential immune evasion abilities. Therefore, we investigated the role of glycan abundance in the phagocytosis and NETosis rates exhibited by female neutrophils (PMNs) upon exposure to such spermatozoa. Our results indicated that the spermatozoa from high fertile (HF) bulls possessed a higher abundance of O-linked glycans e.g., galactosyl (β-1,3)N-acetylgalactosamine and N-linked glycans like [GlcNAc]1-3, N-acetylglucosamine than the low fertile (LF) bull spermatozoa. This differential glycomic endowment appeared to affect the spermiophagy and NETosis rates exhibited by the female neutrophil cells (PMNs). The mean percentage of phagocytizing PMNs was significantly different (P < 0.0001) for HF and LF bulls, 28.44 and 59.59%, respectively. Furthermore, any introduced perturbations in the inherent sperm glycan arrangements promoted phagocytosis by PMNs. For example, after in vitro capacitation the mean phagocytosis rate (MPR) rate in spermatozoa from HF bulls significantly increased to 66.49% (P < 0.01). Likewise, the MPR increased to 70.63% (p < 0.01) after O-glycosidase & α2-3,6,8,9 Neuraminidase A treatment of spermatozoa from HF bulls. Moreover, the percentage of PMNs forming neutrophil extracellular traps (NETs) was significantly higher, 41.47% when exposed to spermatozoa from LF bulls vis-à-vis the spermatozoa from HF bulls, 15.46% (P < 0.0001). This is a pioneer report specifically demonstrating the role of O-linked glycans in the immune responses mounted against spermatozoa. Nevertheless, further studies are warranted to provide the measures to diagnose the sub-fertile phenotype thus preventing the losses incurred by incorrect selection of morphologically normal sperm in the AI/IVF reproduction techniques.
Collapse
Affiliation(s)
- Vipul Batra
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Komal Dagar
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Samiksha Nayak
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Arumugam Kumaresan
- Theriogenelogy Laboratory, SRS of National Dairy Research Institute, Bengaluru, India
| | - Rakesh Kumar
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Tirtha K Datta
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| |
Collapse
|
15
|
Le Tortorec A, Matusali G, Mahé D, Aubry F, Mazaud-Guittot S, Houzet L, Dejucq-Rainsford N. From Ancient to Emerging Infections: The Odyssey of Viruses in the Male Genital Tract. Physiol Rev 2020; 100:1349-1414. [PMID: 32031468 DOI: 10.1152/physrev.00021.2019] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The male genital tract (MGT) is the target of a number of viral infections that can have deleterious consequences at the individual, offspring, and population levels. These consequences include infertility, cancers of male organs, transmission to the embryo/fetal development abnormalities, and sexual dissemination of major viral pathogens such as human immunodeficiency virus (HIV) and hepatitis B virus. Lately, two emerging viruses, Zika and Ebola, have additionally revealed that the human MGT can constitute a reservoir for viruses cleared from peripheral circulation by the immune system, leading to their sexual transmission by cured men. This represents a concern for future epidemics and further underlines the need for a better understanding of the interplay between viruses and the MGT. We review here how viruses, from ancient viruses that integrated the germline during evolution through old viruses (e.g., papillomaviruses originating from Neanderthals) and more modern sexually transmitted infections (e.g., simian zoonotic HIV) to emerging viruses (e.g., Ebola and Zika) take advantage of genital tract colonization for horizontal dissemination, viral persistence, vertical transmission, and endogenization. The MGT immune responses to viruses and the impact of these infections are discussed. We summarize the latest data regarding the sources of viruses in semen and the complex role of this body fluid in sexual transmission. Finally, we introduce key animal findings that are relevant for our understanding of viral infection and persistence in the human MGT and suggest future research directions.
Collapse
Affiliation(s)
- Anna Le Tortorec
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Giulia Matusali
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Dominique Mahé
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Florence Aubry
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Séverine Mazaud-Guittot
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Laurent Houzet
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| |
Collapse
|
16
|
Solano ME, Arck PC. Steroids, Pregnancy and Fetal Development. Front Immunol 2020; 10:3017. [PMID: 32038609 PMCID: PMC6987319 DOI: 10.3389/fimmu.2019.03017] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022] Open
Abstract
Maternal glucocorticoids critically rise during pregnancy reaching up to a 20-fold increase of mid-pregnancy concentrations. Concurrently, another steroid hormone, progesterone, increases. Progesterone, which shows structural similarities to glucocorticoids, can bind the intracellular glucocorticoid receptor, although with lower affinity. Progesterone is essential for the establishment and continuation of pregnancy and it is generally acknowledged to promote maternal immune tolerance to fetal alloantigens through a wealth of immunomodulatory mechanisms. Despite the potent immunomodulatory capacity of glucocorticoids, little is known about their role during pregnancy. Here we aim to compare general aspects of glucocorticoids and progesterone during pregnancy, including shared common steroidogenic pathways, plasma transporters, regulatory pathways, expression of receptors, and mechanisms of action in immune cells. It was recently acknowledged that progesterone receptors are not ubiquitously expressed on immune cells and that pivotal features of progesterone induced- maternal immune adaptations to pregnancy are mediated via the glucocorticoid receptor, including e.g., T regulatory cells expansion. We hypothesize that a tight equilibrium between progesterone and glucocorticoids is critically required and recapitulate evidence supporting that their disequilibrium underlie pregnancy complications. Such a disequilibrium can occur, e.g., after maternal stress perception, which triggers the release of glucocorticoids and impair progesterone secretion, resulting in intrauterine inflammation. These endocrine misbalance might be interconnected, as increase in glucocorticoid synthesis, e.g., upon stress, may occur in detriment of progesterone steroidogenesis, by depleting the common precursor pregnenolone. Abundant literature supports that progesterone deficiency underlies pregnancy complications in which immune tolerance is challenged. In these settings, it is largely yet undefined if and how glucocorticoids are affected. However, although progesterone immunomodulation during pregnancy appear to be chiefly mediated glucocorticoid receptors, excess glucocorticoids cannot compensate by progesterone deficiency, indicating that additional und still undercover mechanisms are at play.
Collapse
Affiliation(s)
- Maria Emilia Solano
- Department for Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Clara Arck
- Department for Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
17
|
Archana SS, Selvaraju S, Binsila BK, Arangasamy A, Krawetz SA. Immune regulatory molecules as modifiers of semen and fertility: A review. Mol Reprod Dev 2019; 86:1485-1504. [DOI: 10.1002/mrd.23263] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 08/22/2019] [Indexed: 12/16/2022]
Affiliation(s)
- S. Siddalingappa Archana
- Reproductive Physiology Laboratory, Animal Physiology DivisionICAR‐National Institute of Animal Nutrition and Physiology Bengaluru India
- Department of BiochemistryJain University Bengaluru India
| | - Sellappan Selvaraju
- Reproductive Physiology Laboratory, Animal Physiology DivisionICAR‐National Institute of Animal Nutrition and Physiology Bengaluru India
| | - B. Krishnan Binsila
- Reproductive Physiology Laboratory, Animal Physiology DivisionICAR‐National Institute of Animal Nutrition and Physiology Bengaluru India
| | - Arunachalam Arangasamy
- Reproductive Physiology Laboratory, Animal Physiology DivisionICAR‐National Institute of Animal Nutrition and Physiology Bengaluru India
| | - Stephen A. Krawetz
- Department of Obstetrics and GynecologyWayne State University School of Medicine Detroit Michigan
- Center for Molecular Medicine and GeneticsC.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine Detroit Michigan
| |
Collapse
|
18
|
Meinhardt A, Wang M, Schulz C, Bhushan S. Microenvironmental signals govern the cellular identity of testicular macrophages. J Leukoc Biol 2019; 104:757-766. [PMID: 30265772 DOI: 10.1002/jlb.3mr0318-086rr] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 12/24/2022] Open
Abstract
Testicular macrophages (TM) comprise the largest immune cell population in the mammalian testis. They are characterized by a subdued proinflammatory response upon adequate stimulation, and a polarization toward the immunoregulatory and immunotolerant M2 phenotype. This enables them to play a relevant role in supporting the archetypical functions of the testis, namely spermatogenesis and steroidogenesis. During infection, the characteristic blunted immune response of TM reflects the need for a delicate balance between a sufficiently strong reaction to counteract invading pathogens, and the prevention of excessive proinflammatory cytokine levels with the potential to disturb or destroy spermatogenesis. Local microenvironmental factors that determine the special phenotype of TM have just begun to be unraveled, and are discussed in this review.
Collapse
Affiliation(s)
- Andreas Meinhardt
- Unit of Reproductive Biology, Institute of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Ming Wang
- Unit of Reproductive Biology, Institute of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität, Ludwig-Maximilians-Universität, Munich, Germany
| | - Sudhanshu Bhushan
- Unit of Reproductive Biology, Institute of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
19
|
Feng Z, Liang C, Manthari RK, Wang C, Zhang J. Effects of Fluoride on Autophagy in Mouse Sertoli Cells. Biol Trace Elem Res 2019; 187:499-505. [PMID: 29915883 DOI: 10.1007/s12011-018-1405-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 05/31/2018] [Indexed: 12/15/2022]
Abstract
Fluoride had been reported to damage the structure and function of testicular tissues and reproductive cells; however, the mechanisms underlying its toxicity remained unclear. Autophagy plays a key role in reproductive function. In this study, we aimed to investigate the effect of fluoride on autophagy in Sertoli cells. Sertoli cells were exposed to 0, 0.125, 0.25, and 0.5 mM NaF for 24 h. The results showed that fluoride exposure up-regulated Beclin1 and p62 mRNA and protein expression levels with concomitant down-regulated mRNA and protein expression levels of LC3 and Atg5. In conclusion, exposure to fluoride impaired the autophagy process in Sertoli cells, which could be one of fluoride's mechanisms in male reproductive toxicity.
Collapse
Affiliation(s)
- Zhiyuan Feng
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| | - Chen Liang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| | - Ram Kumar Manthari
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
| | - Chong Wang
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, People's Republic of China
| | - Jianhai Zhang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China.
| |
Collapse
|
20
|
Lu Y, Rafiq A, Zhang Z, Aslani F, Fijak M, Lei T, Wang M, Kumar S, Klug J, Bergmann M, Chakraborty T, Meinhardt A, Bhushan S. Uropathogenic Escherichia coli virulence factor hemolysin A causes programmed cell necrosis by altering mitochondrial dynamics. FASEB J 2018; 32:4107-4120. [PMID: 29490169 DOI: 10.1096/fj.201700768r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Uropathogenic Escherichia coli (UPEC) is the most common cause of urinary tract infections. In this study, UPEC strains harboring hemolysin A (HlyA) did not induce programmed cell death pathways by the activation of caspases. Instead, the UPEC pore-forming toxin HlyA triggered an increase in mitochondrial Ca2+ levels and manipulated mitochondrial dynamics by causing fragmentation of the mitochondrial network. Alterations in mitochondrial dynamics resulted in severe impairment of mitochondrial functions by loss of membrane potential, increase in reactive oxygen species production, and ATP depletion. Moreover, HlyA caused disruption of plasma membrane integrity that was accompanied by extracellular release of the danger-associated molecules high-mobility group box 1 (HMGB1) and histone 3 (H3). Our results indicate that UPEC induced programmed cell necrosis by irreversibly impairing mitochondrial function. This finding suggests a strategy devised by UPEC at the onset of infection to escape early innate immune response and silently propagate inside host cells.-Lu, Y., Rafiq, A., Zhang, Z., Aslani, F., Fijak, M., Lei, T., Wang, M., Kumar, S., Klug, J., Bergmann, M., Chakraborty, T., Meinhardt, A., Bhushan, S. Uropathogenic Escherichia coli virulence factor hemolysin A causes programmed cell necrosis by altering mitochondrial dynamics.
Collapse
Affiliation(s)
- Yongning Lu
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Amir Rafiq
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Zhengguo Zhang
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Ferial Aslani
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Tao Lei
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Ming Wang
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Sanjeev Kumar
- Department of Medical Microbiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jörg Klug
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Martin Bergmann
- Institute of Veterinary Anatomy, Histology, and Embryology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Trinad Chakraborty
- Department of Medical Microbiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
21
|
Immunometabolism, pregnancy, and nutrition. Semin Immunopathol 2017; 40:157-174. [PMID: 29071391 DOI: 10.1007/s00281-017-0660-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/18/2017] [Indexed: 12/16/2022]
Abstract
The emerging field of immunometabolism has substantially progressed over the last years and provided pivotal insights into distinct metabolic regulators and reprogramming pathways of immune cell populations in various immunological settings. However, insights into immunometabolic reprogramming in the context of reproduction are still enigmatic. During pregnancy, the maternal immune system needs to actively adapt to the presence of the fetal antigens, i.e., by functional modifications of distinct innate immune cell subsets, the generation of regulatory T cells, and the suppression of an anti-fetal effector T cell response. Considering that metabolic pathways have been shown to affect the functional role of such immune cells in a number of settings, we here review the potential role of immunometabolism with regard to the molecular and cellular mechanisms necessary for successful reproduction. Since immunometabolism holds the potential for a therapeutic approach to alter the course of immune diseases, we further highlight how a targeted metabolic reprogramming of immune cells may be triggered by maternal anthropometric or nutritional aspects.
Collapse
|
22
|
Fidyk W, Mitrus I, Ciomber A, Smagur A, Chwieduk A, Głowala-Kosińska M, Giebel S. Evaluation of proinflammatory and immunosuppressive cytokines in blood and bone marrow of healthy hematopoietic stem cell donors. Cytokine 2017; 102:181-186. [PMID: 28927758 DOI: 10.1016/j.cyto.2017.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/17/2017] [Accepted: 09/02/2017] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Cytokine composition of bone marrow microenvironment in comparison to blood is poorly explored. The goal of this study was to investigate the levels of cytokines present in peripheral blood and bone marrow of healthy hematopoietic stem cells donors. The data obtained on this subject with addition to cytometric analysis can provide new insight into the hematopoietic stem cells microenvironment. METHODOLOGY Study consisted of cytokine concentration analysis performed by ELISA tests of peripheral blood of healthy peripheral blood stem cells donors and bone marrow of healthy bone marrow donors. Additionally we have tested the expression of CD47 and CD274 proteins on the surface of hematopoietic stem cells by the flow cytometry analysis. RESULTS The results has shown different composition of analyzed cytokines (IL-1 β, IL-2, IL-4, IL-6, IL-10, IL-17A, TGF-β1, IFN-γ and TNF-α) present in bone marrow and blood of stem cells donors. The hematopoietic stem cells in peripheral blood are subjected to higher levels of proinflammatory cytokines whilst the lower level of those cytokines in bone marrow with a very high level of TGF-β1 which possibly creates a more immunosuppressive environment. The IL-10 level was significantly higher in peripheral blood of PBSC donors after the administration of mobilizing factor (G-CSF). The percentage of CD47+HSCs was significantly higher in bone marrow compared to peripheral blood of mobilized donors.
Collapse
Affiliation(s)
- Wojciech Fidyk
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute Oncology Center, Gliwice Branch, 44-101 Gliwice, Wybrzeże Armii Krajowej 15 Street, Poland.
| | - Iwona Mitrus
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute Oncology Center, Gliwice Branch, 44-101 Gliwice, Wybrzeże Armii Krajowej 15 Street, Poland
| | - Agnieszka Ciomber
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute Oncology Center, Gliwice Branch, 44-101 Gliwice, Wybrzeże Armii Krajowej 15 Street, Poland
| | - Andrzej Smagur
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute Oncology Center, Gliwice Branch, 44-101 Gliwice, Wybrzeże Armii Krajowej 15 Street, Poland
| | - Agata Chwieduk
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute Oncology Center, Gliwice Branch, 44-101 Gliwice, Wybrzeże Armii Krajowej 15 Street, Poland
| | - Magdalena Głowala-Kosińska
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute Oncology Center, Gliwice Branch, 44-101 Gliwice, Wybrzeże Armii Krajowej 15 Street, Poland
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute Oncology Center, Gliwice Branch, 44-101 Gliwice, Wybrzeże Armii Krajowej 15 Street, Poland
| |
Collapse
|
23
|
Adhikary T, Wortmann A, Finkernagel F, Lieber S, Nist A, Stiewe T, Wagner U, Müller-Brüsselbach S, Reinartz S, Müller R. Interferon signaling in ascites-associated macrophages is linked to a favorable clinical outcome in a subgroup of ovarian carcinoma patients. BMC Genomics 2017; 18:243. [PMID: 28327095 PMCID: PMC5359932 DOI: 10.1186/s12864-017-3630-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 03/15/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Although tumor-associated macrophages (TAMs) are essential for cancer progression, connections between different clinical outcomes and transcriptional networks have not been reported. We have addressed this issue by analyzing global expression patterns of TAMs isolated from the ascites of ovarian cancer patients. RESULTS TAMs isolated from different ovarian cancer patients can be stratified by coexpression or principal component analysis into subgroups with specific biological features and associated with distinct clinical outcomes. A hallmark of subgroup A is a high expression of clinically unfavorable markers, including (i) high CD163 expression, a surface receptor characteristic of an anti-inflammatory activation state, (ii) increased PCOLCE2 expression, indicative of enhanced extracellular matrix organization, and (iii) elevated ascites levels of IL-6 and IL-10, linked to the aggressiveness of ovarian cancer and immune suppression. In contrast, subgroup B TAMs are characterized by the upregulation of genes linked to immune defense mechanisms and interferon (IFN) signaling. Intriguingly, analysis of published data for 1763 ovarian cancer patients revealed a strong association of this transcriptional signature with a longer overall survival. Consistent with these results, IFNγ was able to abrogate the suppressive effect of ovarian cancer ascites on the inducibility of IL12B expression and IL-12 secretion, a key determinant of a cytotoxic immune response. CONCLUSIONS The survival of ovarian cancer patients is linked to the presence of TAMs with a transcriptional signature that is characterized by a low expression of protumorigenic and immunosuppressive markers and an upregulation of genes linked to interferon signaling. The observed IFNγ-mediated restoration of the inducibility of IL-12 in the presence of ascites provides a possible explanation for the association of an interferon signaling-associated signature with a favorable clinical outcome.
Collapse
Affiliation(s)
- Till Adhikary
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Annika Wortmann
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany.
| |
Collapse
|
24
|
Jasienska G, Ellison PT, Galbarczyk A, Jasienski M, Kalemba-Drozdz M, Kapiszewska M, Nenko I, Thune I, Ziomkiewicz A. Apolipoprotein E (ApoE) polymorphism is related to differences in potential fertility in women: a case of antagonistic pleiotropy? Proc Biol Sci 2015; 282:20142395. [PMID: 25673673 DOI: 10.1098/rspb.2014.2395] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The alleles that are detrimental to health, especially in older age, are thought to persist in populations because they also confer some benefits for individuals (through antagonistic pleiotropy). The ApoE4 allele at the ApoE locus, encoding apolipoprotein E (ApoE), significantly increases risk of poor health, and yet it is present in many populations at relatively high frequencies. Why has it not been replaced by natural selection with the health-beneficial ApoE3 allele? ApoE is a major supplier of cholesterol precursor for the production of ovarian oestrogen and progesterone, thus ApoE has been suggested as the potential candidate gene that may cause variation in reproductive performance. Our results support this hypothesis showing that in 117 regularly menstruating women those with genotypes with at least one ApoE4 allele had significantly higher levels of mean luteal progesterone (144.21 pmol l(-1)) than women with genotypes without ApoE4 (120.49 pmol l(-1)), which indicates higher potential fertility. The hormonal profiles were based on daily data for entire menstrual cycles. We suggest that the finding of higher progesterone in women with ApoE4 allele could provide first strong evidence for an evolutionary mechanism of maintaining the ancestral and health-worsening ApoE4 allele in human populations.
Collapse
Affiliation(s)
- Grazyna Jasienska
- Department of Environmental Health, Jagiellonian University Medical College, Krakow, Poland
| | - Peter T Ellison
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Andrzej Galbarczyk
- Department of Environmental Health, Jagiellonian University Medical College, Krakow, Poland
| | - Michal Jasienski
- Center for Innovatics, Nowy Sacz Business School-National-Louis University, Zielona 27, 33-300 Nowy Sacz, Poland
| | | | | | - Ilona Nenko
- Department of Environmental Health, Jagiellonian University Medical College, Krakow, Poland
| | - Inger Thune
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway The Cancer Center, Oslo University Hospital, Oslo, Norway
| | - Anna Ziomkiewicz
- Polish Academy of Sciences, Unit of Anthropology in Wroclaw, Poland
| |
Collapse
|
25
|
Ludke A, Wu J, Nazari M, Hatta K, Shao Z, Li SH, Song H, Ni NC, Weisel RD, Li RK. Uterine-derived progenitor cells are immunoprivileged and effectively improve cardiac regeneration when used for cell therapy. J Mol Cell Cardiol 2015; 84:116-28. [PMID: 25939780 DOI: 10.1016/j.yjmcc.2015.04.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/07/2015] [Accepted: 04/26/2015] [Indexed: 01/03/2023]
Abstract
Cell therapy to prevent cardiac dysfunction after myocardial infarction (MI) is less effective in aged patients because aged cells have decreased regenerative capacity. Allogeneic transplanted stem cells (SCs) from young donors are usually rejected. Maintaining transplanted SC immunoprivilege may dramatically improve regenerative outcomes. The uterus has distinct immune characteristics, and we showed that reparative uterine SCs home to the myocardium post-MI. Here, we identify immunoprivileged uterine SCs and assess their effects on cardiac regeneration after allogeneic transplantation. We found more than 20% of cells in the mouse uterus have undetectable MHC I expression by flow cytometry. Uterine MHC I((neg)) and MHC I((pos)) cells were separated by magnetic cell sorting. The MHC I((neg)) population expressed the SC markers CD34, Sca-1 and CD90, but did not express MHC II or c-kit. In vitro, MHC I((neg)) and ((pos)) SCs show colony formation and endothelial differentiation capacity. In mixed leukocyte co-culture, MHC I((neg)) cells showed reduced cell death and leukocyte proliferation compared to MHC I((pos)) cells. MHC I((neg)) and ((pos)) cells had significantly greater angiogenic capacity than mesenchymal stem cells. The benefits of intramyocardial injection of allogeneic MHC I((neg)) cells after MI were comparable to syngeneic bone marrow cell transplantation, with engraftment in cardiac tissue and limited recruitment of CD4 and CD8 cells up to 21 days post-MI. MHC I((neg)) cells preserved cardiac function, decreased infarct size and improved regeneration post-MI. This new source of immunoprivileged cells can induce neovascularization and could be used as allogeneic cell therapy for regenerative medicine.
Collapse
Affiliation(s)
- Ana Ludke
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Jun Wu
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Mansoreh Nazari
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Kota Hatta
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Zhengbo Shao
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shu-Hong Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Huifang Song
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Anatomy, Shanxi Medical University, Taiyuan, China
| | - Nathan C Ni
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Richard D Weisel
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada.
| |
Collapse
|
26
|
Fijak M, Damm LJ, Wenzel JP, Aslani F, Walecki M, Wahle E, Eisel F, Bhushan S, Hackstein H, Baal N, Schuler G, Konrad L, Rafiq A, O'Hara L, Smith LB, Meinhardt A. Influence of Testosterone on Inflammatory Response in Testicular Cells and Expression of Transcription Factor Foxp3 in T Cells. Am J Reprod Immunol 2015; 74:12-25. [DOI: 10.1111/aji.12363] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/02/2015] [Indexed: 01/31/2023] Open
Affiliation(s)
- Monika Fijak
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Lara-Jil Damm
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Jan-Per Wenzel
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Ferial Aslani
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Magdalena Walecki
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Eva Wahle
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Florian Eisel
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine; Justus-Liebig-University; Giessen Germany
| | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine; Justus-Liebig-University; Giessen Germany
| | - Gerhard Schuler
- Clinic for Obstetrics; Gynecology and Andrology of Large and Small Animals; Faculty of Veterinary Medicine; Justus-Liebig-University; Giessen Germany
| | - Lutz Konrad
- Department of Obstetrics and Gynaecology; Faculty of Medicine; Justus-Liebig-University; Giessen Germany
| | - Amir Rafiq
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Laura O'Hara
- MRC Centre for Reproductive Health; University of Edinburgh; Edinburgh UK
| | - Lee B. Smith
- MRC Centre for Reproductive Health; University of Edinburgh; Edinburgh UK
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| |
Collapse
|
27
|
Arck PC, Hecher K, Solano ME. B Cells in Pregnancy: Functional Promiscuity or Tailored Function? Biol Reprod 2015; 92:12. [DOI: 10.1095/biolreprod.114.126110] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
28
|
Aslani F, Schuppe HC, Guazzone VA, Bhushan S, Wahle E, Lochnit G, Lustig L, Meinhardt A, Fijak M. Targeting high mobility group box protein 1 ameliorates testicular inflammation in experimental autoimmune orchitis. Hum Reprod 2014; 30:417-31. [PMID: 25452436 DOI: 10.1093/humrep/deu320] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Does high mobility group box protein 1 (HMGB1) regulate inflammatory reactions in a rat model of experimental autoimmune orchitis (EAO)? SUMMARY ANSWER HMGB1 appears to be involved in regulating inflammatory reactions in testes, as HMGB1 is translocated from testicular cells during the course of EAO and blocking its action by ethyl pyruvate (EP) reduces disease progression and spermatogenic damage. WHAT IS KNOWN ALREADY Despite its immune privileged status, the human testis is prone to inflammatory lesions associated with male factor infertility. Accumulating evidence shows that HMGB1 plays an important role in onset and progression of autoimmune diseases. STUDY DESIGN, SIZE, DURATION This is a cross sectional and longitudinal study involving Wistar male rats immunized with testicular homogenates to induce EAO 50 (EAO50; n = 10) and 80 (EAO80; n = 10) days after first immunization. Control adjuvant animals received saline instead of testicular homogenate (n = 16). Untreated animals (n = 10) were also studied. An interventional study was performed to block the action of HMGB1 starting 20 days after first immunization in EAO animals and respective controls (n = 17). Rats were treated i.p. with EP and the effect of EP treatment on testicular pathogenesis was evaluated 30 days later. Moreover, human testicular biopsies from infertile men with focal lymphocytic infiltrates (n = 7) and sections with intact spermatogenesis (n = 6) were probed with antibodies against HMGB1. PARTICIPANTS/MATERIALS, SETTING, METHODS Testicular RNA and protein extracts from EAO animals, EAO animals treated with EP and relevant controls were used for analysis of cytokine expression by real-time RT-PCR and enzyme-linked immunosorbent assay. HMGB1 was co-localized on rat testicular cross sections with antibodies against testicular macrophages (TM), peritubular cells (PTC) and Sertoli cells (SC). Interaction of HMGB1 and its receptors (RAGE, TLR4) as well signaling pathways after HMGB1 stimulation were studied in isolated TM, PTC and SC by proximity ligation assay and western blot, respectively. Furthermore, HMGB1 immunofluorescence on human testicular biopsies was performed. MAIN RESULTS AND THE ROLE OF CHANCE HMGB1 was translocated from the nuclei in EAO testes and testes of infertile men with impaired spermatogenesis and lymphocytic infiltrates. Elevated HMGB1 levels were observed during late phase of EAO. In testicular somatic cells HMGB1 receptors Toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE) were differentially expressed: HMGB1-TLR4 binding was predominant in TM, while HMGB1-RAGE interaction was prevalent in SC and PTC. In support, HMGB1 triggered extracellular signal regulated kinase (ERK)1/2 and cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) activation in SC and PTC, while TM responded to HMGB1 stimulation with p38 mitogen-activated protein kinase (MAPK) and p65 nuclear factor Kappa B (NF-ĸB) phosphorylation followed by increased tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) mRNA levels. In vivo treatment of EAO animals with EP 20 days after induction of disease revealed beneficial effects, as documented by reduced disease progression and spermatogenic damage, lower macrophage numbers, as well as decreased concentrations of HMGB1 and IL-6 in the testis compared with EAO controls. LIMITATIONS, REASONS FOR CAUTION The ability of HMGB1 to bind to a wide range of receptors makes it difficult to prevent its action by blockade of a specific receptor; therefore we applied EP, a drug preventing HMGB1 release from cells. Due to its mode of action EP decreases also the secretion of some other pro-inflammatory cytokines. Using isolated primary cells imposes limitations for cell transfection studies. As a compromise between purity and yield primary cells need to be isolated from animals of different age, which has to be considered when comparing their responses. WIDER IMPLICATIONS OF THE FINDINGS HMGB1 could be a promising target in attenuating testicular damage caused by inflammatory reactions.
Collapse
Affiliation(s)
- Ferial Aslani
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany
| | - Hans-Christian Schuppe
- Department of Urology, Pediatric Urology and Andrology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany
| | - Vanesa A Guazzone
- Instituto de Investigaciones Biomédicas UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1121 ABG, Buenos Aires, Argentina
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany
| | - Eva Wahle
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany
| | - Günter Lochnit
- Institute of Biochemistry, Faculty of Medicine, Justus-Liebig University of Giessen, 35392 Giessen, Germany
| | - Livia Lustig
- Instituto de Investigaciones Biomédicas UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1121 ABG, Buenos Aires, Argentina
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany
| |
Collapse
|
29
|
Meyron-Holtz EG, Cohen LA, Fahoum L, Haimovich Y, Lifshitz L, Magid-Gold I, Stuemler T, Truman-Rosentsvit M. Ferritin polarization and iron transport across monolayer epithelial barriers in mammals. Front Pharmacol 2014; 5:194. [PMID: 25202274 PMCID: PMC4142484 DOI: 10.3389/fphar.2014.00194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 08/02/2014] [Indexed: 12/20/2022] Open
Abstract
Epithelial barriers are found in many tissues such as the intestine, kidney and brain where they separate the external environment from the body or a specific compartment from its periphery. Due to the tight junctions that connect epithelial barrier-cells (EBCs), the transport of compounds takes place nearly exclusively across the apical or basolateral membrane, the cell-body and the opposite membrane of the polarized EBC, and is regulated on numerous levels including barrier-specific adapted trafficking-machineries. Iron is an essential element but toxic at excess. Therefore, all iron-requiring organisms tightly regulate iron concentrations on systemic and cellular levels. In contrast to most cell types that control just their own iron homeostasis, EBCs also regulate homeostasis of the compartment they enclose or the body as a whole. Iron is transported across EBCs by specialized transporters such as the transferrin receptor and ferroportin. Recently, the iron storage protein ferritin was also attributed a role in the regulation of systemic iron homeostasis and we gathered evidence from the literature and original data that ferritin is polarized in EBC, suggesting also a role for ferritin in iron trafficking across EBCs.
Collapse
Affiliation(s)
- Esther G Meyron-Holtz
- Laboratory for Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Technion City Haifa, Israel
| | - Lyora A Cohen
- Laboratory for Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Technion City Haifa, Israel
| | - Lulu Fahoum
- Laboratory for Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Technion City Haifa, Israel
| | - Yael Haimovich
- Laboratory for Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Technion City Haifa, Israel
| | - Lena Lifshitz
- Laboratory for Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Technion City Haifa, Israel
| | - Inbar Magid-Gold
- Laboratory for Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Technion City Haifa, Israel
| | - Tanja Stuemler
- Laboratory for Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Technion City Haifa, Israel
| | - Marianna Truman-Rosentsvit
- Laboratory for Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Technion City Haifa, Israel
| |
Collapse
|
30
|
Tiegs G, Karimi K, Brune K, Arck P. New problems arising from old drugs: second-generation effects of acetaminophen. Expert Rev Clin Pharmacol 2014; 7:655-62. [PMID: 25075430 DOI: 10.1586/17512433.2014.944502] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Acetaminophen (APAP)/paracetamol is one of the most commonly used over-the-counter drugs taken worldwide for treatment of pain and fever. Although considered as safe when taken in recommended doses not higher than 4 g/day, APAP overdose is currently the most important cause of acute liver failure (ALF). ALF may require liver transplantation and can be fatal. The reasons for APAP-related ALF are mostly intentional (suicidal) or unintentional overdose. However, results from large scale epidemiological studies provide increasing evidence for second generation effects of APAP, even when taken in pharmacological doses. Most strikingly, APAP medication during pregnancy has been associated with health problems including neurodevelopmental and behavioral disorders such as attention deficit hyperactivity disorder and increase in the risk of wheezing and incidence of asthma among offspring. This article reviews the epidemiological findings and aims to shed light into the molecular and cellular mechanisms responsible for APAP-mediated prenatal risk for asthma.
Collapse
Affiliation(s)
- Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | |
Collapse
|
31
|
Bissegger S, Martyniuk CJ, Langlois VS. Transcriptomic profiling in Silurana tropicalis testes exposed to finasteride. Gen Comp Endocrinol 2014; 203:137-45. [PMID: 24530632 DOI: 10.1016/j.ygcen.2014.01.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 01/24/2014] [Accepted: 01/28/2014] [Indexed: 01/14/2023]
Abstract
Investigations of endocrine disrupting chemicals found in aquatic ecosystems with estrogenic and androgenic modes of action have increased over the past two decades due to a surge of evidence of adverse effects in wildlife. Chemicals that disrupt androgen signalling and steroidogenesis can result in an imbalanced conversion of testosterone (T) into 17β-estradiol (E2) and other androgens such as 5α-dihydrotestosterone (5α-DHT). Therefore, a better understanding of how chemicals perturb these pathways is warranted. In this study, the brain, liver, and testes of Silurana tropicalis were exposed ex vivo to the human drug finasteride, a potent steroid 5α-reductase inhibitor and a model compound to study the inhibition of the conversion of T into 5α-DHT. These experiments were conducted (1) to determine organ specific changes in sex steroid production after treatment, and (2) to elucidate the transcriptomic response to finasteride in testicular tissue. Enzyme-linked immunosorbent assays were used to measure hormone levels in media following finasteride incubation for 6 h. Finasteride significantly increased T levels in the media of liver and testis tissue, but did not induce any changes in E2 and 5α-DHT production. Gene expression analysis was performed in frog testes and data revealed that finasteride treatment significantly altered 1,434 gene probes. Gene networks associated with male reproduction such as meiosis, hormone biosynthesis, sperm entry, gonadotropin releasing hormone were affected by finasteride exposure as well as other pathways such as oxysterol synthesis, apoptosis, and epigenetic regulation. For example, this study suggests that the mode of action by which finasteride induces cellular damage in testicular tissue as reported by others, is via oxidative stress in testes. This data also suggests that 5-reductase inhibition disrupts the expression of genes related to reproduction. It is proposed that androgen-disrupting chemicals may mediate their action via 5-reductases and that the effects of environmental pollutants are not limited to the androgen receptor signalling.
Collapse
Affiliation(s)
- Sonja Bissegger
- Chemistry and Chemical Engineering Department, Royal Military College of Canada, Kingston, ON, Canada.
| | - Christopher J Martyniuk
- Department of Biology and the Canadian River Institute, University of New Brunswick, NB, Canada.
| | - Valérie S Langlois
- Chemistry and Chemical Engineering Department, Royal Military College of Canada, Kingston, ON, Canada.
| |
Collapse
|
32
|
Hartwig IRV, Bruenahl CA, Ramisch K, Keil T, Inman M, Arck PC, Pincus M. Reduced levels of maternal progesterone during pregnancy increase the risk for allergic airway diseases in females only. J Mol Med (Berl) 2014; 92:1093-104. [PMID: 24890522 DOI: 10.1007/s00109-014-1167-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/25/2014] [Accepted: 05/09/2014] [Indexed: 12/17/2022]
Abstract
Observational as well as experimental studies support that prenatal challenges seemed to be associated with an increased risk for allergic airway diseases in the offspring. However, insights into biomarkers involved in mediating this risk are largely elusive. We here aimed to test the association between endogenous and exogenous factors documented in pregnant women, including psychosocial, endocrine, and life style parameters, and the risk for allergic airway diseases in the children later in life. We further pursued to functionally test identified factors in a mouse model of an allergic airway response. In a prospectively designed pregnancy cohort (n = 409 families), women were recruited between the 4th and 12th week of pregnancy. To investigate an association between exposures during pregnancy and the incidence of allergic airway disease in children between 3 and 5 years of age, multiple logistic regression analyses were applied. Further, in prenatally stressed adult offspring of BALB/c-mated BALB/c female mice, asthma was experimentally induced by ovalbumin (OVA) sensitization. In addition to the prenatal stress challenge, some pregnant females were treated with the progesterone derivative dihydrodydrogesterone (DHD). In humans, we observed that high levels of maternal progesterone in early human pregnancies were associated with a decreased risk for an allergic airway disease (asthma or allergic rhinitis) in daughters (adjusted OR 0.92; 95% confidence interval [CI] 0.84 to 1.00) but not sons (aOR 1.02, 95% CI 0.94-1.10). In mice, prenatal DHD supplementation of stress-challenged dams attenuated prenatal stress-induced airway hyperresponsiveness exclusively in female offspring. Reduced levels of maternal progesterone during pregnancy-which can result from high stress perception-increase the risk for allergic airway diseases in females but not in males. Key messages: Lower maternal progesterone during pregnancy increases the risk for allergic airway disease only in female offspring. Prenatal progesterone supplementation ameliorates airway hyperreactivity in prenatally stressed murine offspring.
Collapse
Affiliation(s)
- Isabel R V Hartwig
- Laboratory for Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,
| | | | | | | | | | | | | |
Collapse
|
33
|
Jones HE. Time to focus on improving the contraceptive method mix in high HIV prevalence settings and let go of unanswerable questions. Contraception 2014; 90:357-9. [PMID: 24993486 DOI: 10.1016/j.contraception.2014.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/12/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Affiliation(s)
- Heidi E Jones
- City University of New York (CUNY) School of Public Health, Hunter College.
| |
Collapse
|