1
|
Nakamura N, Yoshida N, Suwa T. Three major reasons why transgenerational effects of radiation are difficult to detect in humans. Int J Radiat Biol 2023; 100:1297-1311. [PMID: 36880868 DOI: 10.1080/09553002.2023.2187478] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/18/2023] [Indexed: 03/08/2023]
Abstract
PURPOSE Ionizing radiation can induce mutations in germ cells in various organisms, including fruit flies and mice. However, currently, there is no clear evidence for the transgenerational effects of radiation in humans. This review is an effort to identify possible reasons for the lack of such observations. METHODS Literature search and narrative review. RESULTS 1) In both mice and humans, resting oocytes locate primarily in the cortical region of the ovary where the number of blood vessels is very low especially when young and extra-cellular material is rich, and this region is consequently hypoxic, which probably leads to immature oocytes being resistant to the cell killing and mutagenic effects of radiation. 2) In studies of spermatogonia, the mouse genes used for specific locus test (SLT) studies, which include coat color genes, were hypermutable when compared to many other genes. Recent studies which examined over 1000 segments of genomic DNA indicate that the induction rate of deletion mutation per segment was on the order of 10-6 per Gy, which is one order of magnitude lower than that obtained from the SLT data. Therefore, it appears possible that detecting any transgenerational effects of radiation following human male exposures will be difficult due to a lack of mutable marker genes. 3) Fetal malformations were examined in studies in humans, but the genetic component in such malformations is low, and abnormal fetuses are prone to undergo miscarriage which does not occur in mice, and which leads to difficulties in detecting transgenerational effects. CONCLUSION The lack of clear evidence for radiation effects in humans probably does not result from any problem in the methodologies used but may be due largely to biological properties. Currently, whole genome sequencing studies of exposed parents and offspring are planned, but ethical guidelines need to be followed to avoid discrimination, which had once happened to the atomic bomb survivors.
Collapse
Affiliation(s)
- Nori Nakamura
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Noriaki Yoshida
- Department of Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Tatsuya Suwa
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Dong Z, Qian J, Law TSM, Chau MHK, Cao Y, Xue S, Tong S, Zhao Y, Kwok YK, Ng K, Chan DYL, Chiu PKF, Ng CF, Chung CHS, Mak JSM, Leung TY, Chung JPW, Morton CC, Choy KW. Mate-pair genome sequencing reveals structural variants for idiopathic male infertility. Hum Genet 2023; 142:363-377. [PMID: 36526900 DOI: 10.1007/s00439-022-02510-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Currently, routine genetic investigation for male infertility includes karyotyping analysis and PCR for Y chromosomal microdeletions to provide prognostic information such as sperm retrieval success rate. However, over 85% of male infertility remain idiopathic. We assessed 101 male patients with primary infertility in a retrospective cohort analysis who have previously received negative results from standard-of-care tests. Mate-pair genome sequencing (large-insert size library), an alternative long-DNA sequencing method, was performed to detect clinically significant structural variants (SVs) and copy-number neutral absence of heterozygosity (AOH). Candidate SVs were filtered against our in-house cohort of 1077 fertile men. Genes disrupted by potentially clinically significant variants were correlated with single-cell gene expression profiles of human fetal and postnatal testicular developmental lineages and adult germ cells. Follow-up studies were conducted for each patient with clinically relevant finding(s). Molecular diagnoses were made in 11.1% (7/63) of patients with non-obstructive azoospermia and 13.2% (5/38) of patients with severe oligozoospermia. Among them, 12 clinically significant SVs were identified in 12 cases, including five known syndromes, one inversion, and six SVs with direct disruption of genes by intragenic rearrangements or complex insertions. Importantly, a genetic defect related to intracytoplasmic sperm injection (ICSI) failure was identified in a patient with non-obstructive azoospermia, illustrating the additional value of an etiologic diagnosis in addition to determining sperm retrieval rate. Our study reveals a landscape of various genomic variants in 101 males with idiopathic infertility, not only advancing understanding of the underlying mechanisms of male infertility, but also impacting clinical management.
Collapse
Affiliation(s)
- Zirui Dong
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China. .,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China. .,The Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.
| | - Jicheng Qian
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China
| | - Tracy Sze Man Law
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China
| | - Matthew Hoi Kin Chau
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China.,The Chinese University of Hong Kong-Baylor College of Medicine Joint Center for Medical Genetics, Hong Kong, China
| | - Ye Cao
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China.,The Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuwen Xue
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China
| | - Steve Tong
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Yilin Zhao
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Yvonne K Kwok
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Karen Ng
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - David Yiu Leung Chan
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Peter K-F Chiu
- SH Ho Urology Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi-Fai Ng
- SH Ho Urology Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Cathy Hoi Sze Chung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Jennifer Sze Man Mak
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Tak Yeung Leung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China.,The Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,The Chinese University of Hong Kong-Baylor College of Medicine Joint Center for Medical Genetics, Hong Kong, China
| | - Jacqueline Pui Wah Chung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China.,The Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China.,The Chinese University of Hong Kong-Baylor College of Medicine Joint Center for Medical Genetics, Hong Kong, China
| | - Cynthia C Morton
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, 02115, USA.,Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,Manchester Centre for Audiology and Deafness, School of Health Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Kwong Wai Choy
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China. .,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China. .,The Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China. .,The Chinese University of Hong Kong-Baylor College of Medicine Joint Center for Medical Genetics, Hong Kong, China.
| |
Collapse
|
3
|
Liebich A, Schmid N, Koupourtidou C, Herrmann C, Dietrich KG, Welter H, Ninkovic J, Mayerhofer A. The Molecular Signature of Human Testicular Peritubular Cells Revealed by Single-Cell Analysis. Cells 2022; 11:cells11223685. [PMID: 36429113 PMCID: PMC9688777 DOI: 10.3390/cells11223685] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Peritubular cells of the human testis form a small compartment surrounding the seminiferous tubules. They are crucial for sperm transport, and they emerge as contributors to the spermatogonial stem cell niche. They are among the least known cell types of the human body. We employed single-cell RNA sequencing of cultured human testicular peritubular cells (HTPCs), which had been isolated from testicular samples of donors with normal spermatogenesis. The significant overlap between our results and recently published ex vivo data indicates that HTPCs are a highly adequate cellular model to define and study these cells. Thus, based on the expression of several markers, HTPCs can be classified as testicular smooth muscle cells. Small differences between the in vivo/in vitro expressed genes may be due to cellular plasticity. Plasticity was also shown upon addition of FCS to the culture medium. Based on transcriptome similarities, four cellular states were identified. Further analyses confirmed the presence of known stem cell niche-relevant factors (e.g., GDNF) and identified unknown functions, e.g., the ability to produce retinoic acid. Therefore, HTPCs allow us to define the signature(s) and delineate the functions of human testicular peritubular cells. The data may also serve as a resource for future studies to better understand male (in)fertility.
Collapse
Affiliation(s)
- Annika Liebich
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Nina Schmid
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Christina Koupourtidou
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
- Helmholtz Center Munich, Institute of Stem Cell Research, 85764 Neuherberg, Germany
| | - Carola Herrmann
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Kim-Gwendolyn Dietrich
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Harald Welter
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Jovica Ninkovic
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
- Helmholtz Center Munich, Institute of Stem Cell Research, 85764 Neuherberg, Germany
| | - Artur Mayerhofer
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
- Correspondence: ; Tel.: +49-89-2180-75859
| |
Collapse
|
4
|
Stepanov YK, Speidel JD, Herrmann C, Schmid N, Behr R, Köhn FM, Stöckl JB, Pickl U, Trottmann M, Fröhlich T, Mayerhofer A, Welter H. Profound Effects of Dexamethasone on the Immunological State, Synthesis and Secretion Capacity of Human Testicular Peritubular Cells. Cells 2022; 11:cells11193164. [PMID: 36231125 PMCID: PMC9562650 DOI: 10.3390/cells11193164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/21/2022] [Accepted: 10/04/2022] [Indexed: 11/21/2022] Open
Abstract
The functions of human testicular peritubular cells (HTPCs), forming a small compartment located between the seminiferous epithelium and the interstitial areas of the testis, are not fully known but go beyond intratesticular sperm transport and include immunological roles. The expression of the glucocorticoid receptor (GR) indicates that they may be regulated by glucocorticoids (GCs). Herein, we studied the consequences of the GC dexamethasone (Dex) in cultured HTPCs, which serves as a unique window into the human testis. We examined changes in cytokines, mainly by qPCR and ELISA. A holistic mass-spectrometry-based proteome analysis of cellular and secreted proteins was also performed. Dex, used in a therapeutic concentration, decreased the transcript level of proinflammatory cytokines, e.g., IL6, IL8 and MCP1. An siRNA-mediated knockdown of GR reduced the actions on IL6. Changes in IL6 were confirmed by ELISA measurements. Of note, Dex also lowered GR levels. The proteomic results revealed strong responses after 24 h (31 significantly altered cellular proteins) and more pronounced ones after 72 h of Dex exposure (30 less abundant and 42 more abundant cellular proteins). Dex also altered the composition of the secretome (33 proteins decreased, 13 increased) after 72 h. Among the regulated proteins were extracellular matrix (ECM) and basement membrane components (e.g., FBLN2, COL1A2 and COL3A1), as well as PTX3 and StAR. These results pinpoint novel, profound effects of Dex in HTPCs. If transferrable to the human testis, changes specifically in ECM and the immunological state of the testis may occur in men upon treatment with Dex for medical reasons.
Collapse
Affiliation(s)
| | - Jan Dominik Speidel
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Carola Herrmann
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Nina Schmid
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | | | - Jan Bernd Stöckl
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 München, Germany
| | | | | | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 München, Germany
| | - Artur Mayerhofer
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
- Correspondence: (A.M.); (H.W.); Tel.: +49-89218075859 (A.M.); +49-89218071882 (H.W.)
| | - Harald Welter
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
- Correspondence: (A.M.); (H.W.); Tel.: +49-89218075859 (A.M.); +49-89218071882 (H.W.)
| |
Collapse
|
5
|
Riviere E, Rossi SP, Tavalieri YE, Muñoz de Toro MM, Calandra RS, Mayerhofer A, Matzkin ME, Frungieri MB. Pleiotropic actions of melatonin in testicular peritubular myoid cells of immature Syrian hamsters. Biochim Biophys Acta Gen Subj 2022; 1866:130187. [PMID: 35691458 DOI: 10.1016/j.bbagen.2022.130187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Peritubular myoid cells are emerging as key regulators of testicular function in adulthood. However, little is known about the role of testicular peritubular myoid cells (TPMCs) in the development of the male gonad. We found that, compared to testes of young adult hamsters, gonads of 21 day-old animals show increased melatonin concentration, seminiferous tubular wall thickening and a heterogeneous packaging of its collagen fibers thus raising the question whether melatonin may be involved in the regulation of TPMCs. METHODS We established primary cultures of TPMCs from immature hamsters (ihaTPMCs), which we found express melatonergic receptors. RESULTS Exogeneous melatonin decreased the levels of inflammatory markers (NLRP3 inflammasome, IL1β) but increased the expression of cyclooxygenase 2 (COX2, key enzyme mediating prostaglandin synthesis) and of the glial cell line-derived neurotrophic factor (GDNF) in ihaTPMCs. Melatonin also stimulated ihaTPMCs proliferation and the expression of extracellular matrix proteins such as collagen type I and IV. Furthermore, collagen gel contraction assays revealed an enhanced ability of ihaTPMCs to contract in the presence of melatonin. CONCLUSION Melatonin regulates immune and inflammatory functions as well as contractile phenotype of the peritubular wall in the hamster testis. GENERAL SIGNIFICANCE If transferable to the in vivo situation, melatonin-dependent induction of ihaTPMCs to produce factors known to exert paracrine effects in other somatic cell populations of the gonad suggests that the influence of melatonin may go beyond the peritubular wall and indicates its contribution to testicular development and the establishment of a normal and sustainable spermatogenesis.
Collapse
Affiliation(s)
- Eugenia Riviere
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Ciudad de Buenos Aires C1428ADN, Argentina
| | - Soledad P Rossi
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Ciudad de Buenos Aires C1428ADN, Argentina; Cátedra de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Yamil E Tavalieri
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
| | - Mónica M Muñoz de Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
| | - Ricardo S Calandra
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Ciudad de Buenos Aires C1428ADN, Argentina
| | - Artur Mayerhofer
- Cell Biology, Anatomy III, Faculty of Medicine, Biomedical Center Munich (BMC), Ludwig-Maximilian-University (LMU), 82152 Martinsried, Germany
| | - María E Matzkin
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Ciudad de Buenos Aires C1428ADN, Argentina; Cátedra de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Mónica B Frungieri
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Ciudad de Buenos Aires C1428ADN, Argentina; Cátedra de Química, Ciclo Básico Común, Ciudad de Buenos Aires C1405CAE, Argentina.
| |
Collapse
|
6
|
Willems M, Seβenhausen P, Gies I, Vloeberghs V, Tournaye H, Goossens E, Van Saen D. To graft or not to graft? Intratesticular xenografting of (pre)pubertal testicular tissue from Klinefelter patients as potential ex vivo model to study testicular fibrosis. Reprod Biomed Online 2022; 44:896-906. [DOI: 10.1016/j.rbmo.2022.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/07/2022] [Accepted: 01/18/2022] [Indexed: 12/01/2022]
|
7
|
Hachem NE, Humpfle L, Simon P, Kaese M, Weinhold B, Günther J, Galuska SP, Middendorff R. The Loss of Polysialic Acid Impairs the Contractile Phenotype of Peritubular Smooth Muscle Cells in the Postnatal Testis. Cells 2021; 10:1347. [PMID: 34072405 PMCID: PMC8230264 DOI: 10.3390/cells10061347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
In the testis, the germinal epithelium of seminiferous tubules is surrounded by contractile peritubular cells, which are involved in sperm transport. Interestingly, in postnatal testis, polysialic acid (polySia), which is also an essential player for the development of the brain, was observed around the tubules. Western blotting revealed a massive decrease of polySia from postnatal day 1 towards puberty, together with a fundamental reduction of the net-like intertubular polySia. Using polysialyltransferase knockout mice, we investigated the consequences of the loss of polySia in the postnatal testis. Compared to postnatal wild-type animals, polySia knockouts showed slightly reduced smooth muscle actin (SMA) immunostaining of peritubular smooth muscle cells (SMCs), while calponin, marking more differentiated SMCs, dramatically decreased. In contrast, testicular SMA and calponin immunostaining remained unchanged in vascular SMCs in all genotypes. In addition, the cGMP-dependent protein kinase PKG I, a key enzyme of SMC relaxation, was nearly undetectable in the peritubular SMCs. Cell proliferation in the peritubular layer increased significantly in the knockouts, as shown by proliferating cell nuclear anti (PCNA) staining. Taken together, in postnatal testis, the absence of polySia resulted in an impaired differentiation of peritubular, but not vascular, SMCs to a more synthetic phenotype. Thus, polySia might influence the maintenance of a differentiated phenotype of non-vascular SMCs.
Collapse
Affiliation(s)
- Nadim E. Hachem
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany; (N.E.H.); (L.H.)
| | - Luisa Humpfle
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany; (N.E.H.); (L.H.)
| | - Peter Simon
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany; (P.S.); (M.K.)
| | - Miriam Kaese
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany; (P.S.); (M.K.)
| | - Birgit Weinhold
- Institute of Clinical Biochemistry, OE 4340, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany;
| | - Juliane Günther
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany;
| | - Sebastian P. Galuska
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany; (P.S.); (M.K.)
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany;
| | - Ralf Middendorff
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany; (N.E.H.); (L.H.)
| |
Collapse
|
8
|
Stöckl JB, Schmid N, Flenkenthaler F, Drummer C, Behr R, Mayerhofer A, Arnold GJ, Fröhlich T. Age-Related Alterations in the Testicular Proteome of a Non-Human Primate. Cells 2021; 10:cells10061306. [PMID: 34074003 PMCID: PMC8225046 DOI: 10.3390/cells10061306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/17/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Aging of human testis and associated cellular changes is difficult to assess. Therefore, we used a translational, non-human primate model to get insights into underlying cellular and biochemical processes. Using proteomics and immunohistochemistry, we analyzed testicular tissue of young (age 2 to 3) and old (age 10 to 12) common marmosets (Callithrix jacchus). Using a mass spectrometry-based proteomics approach, we identified 63,124 peptides, which could be assigned to 5924 proteins. Among them, we found proteins specific for germ cells and somatic cells, such as Leydig and Sertoli cells. Quantitative analysis showed 31 differentially abundant proteins, of which 29 proteins were more abundant in older animals. An increased abundance of anti-proliferative proteins, among them CDKN2A, indicate reduced cell proliferation in old testes. Additionally, an increased abundance of several small leucine rich repeat proteoglycans and other extracellular matrix proteins was observed, which may be related to impaired cell migration and fibrotic events. Furthermore, an increased abundance of proteins with inhibitory roles in smooth muscle cell contraction like CNN1 indicates functional alterations in testicular peritubular cells and may mirror a reduced capacity of these cells to contract in old testes.
Collapse
Affiliation(s)
- Jan B. Stöckl
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 München, Germany; (J.B.S.); (F.F.)
| | - Nina Schmid
- Biomedical Center (BMC), Anatomy III–Cell Biology, Medical Faculty, LMU München, 82152 Martinsried, Germany; (N.S.); (A.M.)
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 München, Germany; (J.B.S.); (F.F.)
| | - Charis Drummer
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (C.D.); (R.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37077 Göttingen, Germany
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (C.D.); (R.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37077 Göttingen, Germany
| | - Artur Mayerhofer
- Biomedical Center (BMC), Anatomy III–Cell Biology, Medical Faculty, LMU München, 82152 Martinsried, Germany; (N.S.); (A.M.)
| | - Georg J. Arnold
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 München, Germany; (J.B.S.); (F.F.)
- Correspondence: (G.J.A.); (T.F.)
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 München, Germany; (J.B.S.); (F.F.)
- Correspondence: (G.J.A.); (T.F.)
| |
Collapse
|
9
|
Fleck D, Kenzler L, Mundt N, Strauch M, Uesaka N, Moosmann R, Bruentgens F, Missel A, Mayerhofer A, Merhof D, Spehr J, Spehr M. ATP activation of peritubular cells drives testicular sperm transport. eLife 2021; 10:e62885. [PMID: 33502316 PMCID: PMC7840184 DOI: 10.7554/elife.62885] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Spermatogenesis, the complex process of male germ cell proliferation, differentiation, and maturation, is the basis of male fertility. In the seminiferous tubules of the testes, spermatozoa are constantly generated from spermatogonial stem cells through a stereotyped sequence of mitotic and meiotic divisions. The basic physiological principles, however, that control both maturation and luminal transport of the still immotile spermatozoa within the seminiferous tubules remain poorly, if at all, defined. Here, we show that coordinated contractions of smooth muscle-like testicular peritubular cells provide the propulsive force for luminal sperm transport toward the rete testis. Using a mouse model for in vivo imaging, we describe and quantify spontaneous tubular contractions and show a causal relationship between peritubular Ca2+ waves and peristaltic transport. Moreover, we identify P2 receptor-dependent purinergic signaling pathways as physiological triggers of tubular contractions both in vitro and in vivo. When challenged with extracellular ATP, transport of luminal content inside the seminiferous tubules displays stage-dependent directionality. We thus suggest that paracrine purinergic signaling coordinates peristaltic recurrent contractions of the mouse seminiferous tubules to propel immotile spermatozoa to the rete testis.
Collapse
Affiliation(s)
- David Fleck
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Lina Kenzler
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Nadine Mundt
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
- Research Training Group 2416 MultiSenses – MultiScales, RWTH Aachen UniversityAachenGermany
| | - Martin Strauch
- Institute of Imaging and Computer Vision, RWTH Aachen UniversityAachenGermany
| | - Naofumi Uesaka
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
- Department of Cognitive Neurobiology, Tokyo Medical and Dental UniversityTokyoJapan
| | - Robert Moosmann
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Felicitas Bruentgens
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Annika Missel
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilians-Universität MünchenPlanegg-MartinsriedGermany
| | - Artur Mayerhofer
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilians-Universität MünchenPlanegg-MartinsriedGermany
| | - Dorit Merhof
- Institute of Imaging and Computer Vision, RWTH Aachen UniversityAachenGermany
| | - Jennifer Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
- Research Training Group 2416 MultiSenses – MultiScales, RWTH Aachen UniversityAachenGermany
| |
Collapse
|
10
|
Neto FTL, Flannigan R, Goldstein M. Regulation of Human Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:255-286. [PMID: 34453741 DOI: 10.1007/978-3-030-77779-1_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human spermatogenesis (HS) is an intricate network of sequential processes responsible for the production of the male gamete, the spermatozoon. These processes take place in the seminiferous tubules (ST) of the testis, which are small tubular structures considered the functional units of the testes. Each human testicle contains approximately 600-1200 STs [1], and are capable of producing up to 275 million spermatozoa per day [2].
Collapse
Affiliation(s)
| | - Ryan Flannigan
- Department of Urology, Weill Cornell Medicine, New York, NY, USA.,University of British Columbia, Vancouver, BC, Canada
| | - Marc Goldstein
- Department of Urology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Welter H, Herrmann C, Fröhlich T, Flenkenthaler F, Eubler K, Schorle H, Nettersheim D, Mayerhofer A, Müller-Taubenberger A. Filamin A Orchestrates Cytoskeletal Structure, Cell Migration and Stem Cell Characteristics in Human Seminoma TCam-2 Cells. Cells 2020; 9:E2563. [PMID: 33266100 PMCID: PMC7761120 DOI: 10.3390/cells9122563] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Filamins are large dimeric F-actin cross-linking proteins, crucial for the mechanosensitive properties of a number of cell types. Due to their interaction with a variety of different proteins, they exert important regulatory functions. However, in the human testis the role of filamins has been insufficiently explored. Immunohistochemical staining of human testis samples identified filamin A (FLNA) in spermatogonia and peritubular myoid cells. Investigation of different testicular tumor samples indicated that seminoma also express FLNA. Moreover, mass spectrometric analyses identified FLNA as one of the most abundant proteins in human seminoma TCam-2 cells. We therefore focused on FLNA in TCam-2 cells, and identified by co-immunoprecipitation LAD1, RUVBL1 and DAZAP1, in addition to several cytoskeletal proteins, as interactors of FLNA. To study the role of FLNA in TCam-2 cells, we generated FLNA-deficient cells using the CRISPR/Cas9 system. Loss of FLNA causes an irregular arrangement of the actin cytoskeleton and mechanical instability, impaired adhesive properties and disturbed migratory behavior. Furthermore, transcriptional activity of typical stem cell factors is increased in the absence of FLNA. In summary, our data suggest that FLNA is crucially involved in balancing stem cell characteristics and invasive properties in human seminoma cells and possibly human testicular germ cells.
Collapse
Affiliation(s)
- Harald Welter
- Anatomy III, Cell Biology, Biomedical Center, Ludwig Maximillian University of Munich, 82152 Planegg, Martinsried, Germany; (H.W.); (C.H.); (K.E.); (A.M.-T.)
| | - Carola Herrmann
- Anatomy III, Cell Biology, Biomedical Center, Ludwig Maximillian University of Munich, 82152 Planegg, Martinsried, Germany; (H.W.); (C.H.); (K.E.); (A.M.-T.)
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig Maximilian University of Munich, 81377 Munich, Germany; (T.F.); (F.F.)
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig Maximilian University of Munich, 81377 Munich, Germany; (T.F.); (F.F.)
| | - Katja Eubler
- Anatomy III, Cell Biology, Biomedical Center, Ludwig Maximillian University of Munich, 82152 Planegg, Martinsried, Germany; (H.W.); (C.H.); (K.E.); (A.M.-T.)
| | - Hubert Schorle
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Daniel Nettersheim
- Department of Urology, Urological Research Lab, Translational UroOncology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Artur Mayerhofer
- Anatomy III, Cell Biology, Biomedical Center, Ludwig Maximillian University of Munich, 82152 Planegg, Martinsried, Germany; (H.W.); (C.H.); (K.E.); (A.M.-T.)
| | - Annette Müller-Taubenberger
- Anatomy III, Cell Biology, Biomedical Center, Ludwig Maximillian University of Munich, 82152 Planegg, Martinsried, Germany; (H.W.); (C.H.); (K.E.); (A.M.-T.)
| |
Collapse
|
12
|
Stöckl JB, Schmid N, Flenkenthaler F, Drummer C, Behr R, Mayerhofer A, Arnold GJ, Fröhlich T. Proteomic Insights into Senescence of Testicular Peritubular Cells from a Nonhuman Primate Model. Cells 2020; 9:cells9112498. [PMID: 33213088 PMCID: PMC7698562 DOI: 10.3390/cells9112498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
Age-related changes in the human testis may include morphological alterations, disturbed steroidogenesis, and impaired spermatogenesis. However, the specific impact of cell age remains poorly understood and difficult to assess. Testicular peritubular cells fulfill essential functions, including sperm transport, contributions to the spermatogonial stem cell niche, and paracrine interactions within the testis. To study their role in age-associated decline of testicular functions, we performed comprehensive proteome and secretome analyses of repeatedly passaged peritubular cells from Callithrix jacchus. This nonhuman primate model better reflects the human testicular biology than rodents and further gives access to young donors unavailable from humans. Among 5095 identified proteins, 583 were differentially abundant between samples with low and high passage numbers. The alterations indicate a reduced ability of senescent peritubular cells to contract and secrete proteins, as well as disturbances in nuclear factor (NF)-κB signaling and a reduced capacity to handle reactive oxygen species. Since this in vitro model may not exactly mirror all molecular aspects of in vivo aging, we investigated the proteomes and secretomes of testicular peritubular cells from young and old donors. Even though the age-related alterations at the protein level were less pronounced, we found evidence for impaired protein secretion, altered NF-κB signaling, and reduced contractility of these in vivo aged peritubular cells.
Collapse
Affiliation(s)
- Jan B. Stöckl
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 Munich, Germany; (J.B.S.); (F.F.)
| | - Nina Schmid
- LMU München, Biomedical Center (BMC), Anatomy III—Cell Biology, 82152 Martinsried, Germany; (N.S.); (A.M.)
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 Munich, Germany; (J.B.S.); (F.F.)
| | - Charis Drummer
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (C.D.); (R.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37077 Göttingen, Germany
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (C.D.); (R.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37077 Göttingen, Germany
| | - Artur Mayerhofer
- LMU München, Biomedical Center (BMC), Anatomy III—Cell Biology, 82152 Martinsried, Germany; (N.S.); (A.M.)
| | - Georg J. Arnold
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 Munich, Germany; (J.B.S.); (F.F.)
- Correspondence: (G.J.A.); (T.F.)
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 Munich, Germany; (J.B.S.); (F.F.)
- Correspondence: (G.J.A.); (T.F.)
| |
Collapse
|
13
|
Kauerhof AC, Nicolas N, Bhushan S, Wahle E, Loveland KA, Fietz D, Bergmann M, Groome NP, Kliesch S, Schuppe HC, Pilatz A, Meinhardt A, Hedger MP, Fijak M. Investigation of activin A in inflammatory responses of the testis and its role in the development of testicular fibrosis. Hum Reprod 2020; 34:1536-1550. [PMID: 31340036 DOI: 10.1093/humrep/dez109] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 05/27/2019] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION Does activin A contribute to testicular fibrosis under inflammatory conditions? SUMMARY ANSWER Our results show that activin A and key fibrotic proteins are increased in human testicular biopsies with leukocytic infiltrates and impaired spermatogenesis and in murine experimental autoimmune orchitis (EAO) and that activin A stimulates fibrotic responses in peritubular cells (PTCs) and NIH 3T3 fibroblasts. WHAT IS KNOWN ALREADY Fibrosis is a feature of EAO. Activin A, a regulator of fibrosis, was increased in testes of mice with EAO and its expression correlated with severity of the disease. STUDY DESIGN, SIZE, DURATION This is a cross-sectional and longitudinal study of adult mice immunized with testicular homogenate (TH) in adjuvant to induce EAO, collected at 30 (n = 6), 50 (n = 6) and 80 (n = 5) days after first immunization. Age-matched mice injected with adjuvant alone (n = 14) and untreated mice (n = 15) were included as controls. TH-immunized mice with elevated endogenous follistatin, injected with a non-replicative recombinant adeno-associated viral vector carrying a gene cassette of follistatin (rAAV-FST315; n = 3) or vector with an empty cassette (empty vector controls; n = 2) 30 days prior to the first immunization, as well as appropriate adjuvant (n = 2) and untreated (n = 2) controls, were also examined.Human testicular biopsies showing focal inflammatory lesions associated with impaired spermatogenesis (n = 7) were included. Biopsies showing intact spermatogenesis without inflammation, from obstructive azoospermia patients, served as controls (n = 7).Mouse primary PTC and NIH 3T3 fibroblasts were stimulated with activin A and follistatin 288 (FST288) to investigate the effect of activin A on the expression of fibrotic markers. Production of activin A by mouse primary Sertoli cells (SCs) was also investigated. PARTICIPANTS/MATERIALS, SETTING, METHODS Testicular RNA and protein extracts collected from mice at days 30, 50 and 80 after first immunization were used for analysis of fibrotic marker genes and proteins, respectively. Total collagen was assessed by hydroxyproline assay and fibronectin; collagen I, III and IV, α-smooth muscle actin (α-SMA) expression and phosphorylation of suppressor of mothers against decapentaplegic (SMAD) family member 2 were measured by western blot. Immunofluorescence was used to detect fibronectin. Fibronectin (Fn), αSMA (Acta2), collagen I (Col1a2), III (Col3a1) and IV (Col4a1) mRNA in PTC and NIH 3T3 cells treated with activin A and/or FST288 were measured by quantitative RT-PCR (qRT-PCR). Activin A in SC following tumour necrosis factor (TNF) or FST288 stimulation was measured by ELISA. Human testicular biopsies were analysed by qRT-PCR for PTPRC (CD45) and activin A (INHBA), hydroxyproline assay and immunofluorescence. MAIN RESULTS AND THE ROLE OF CHANCE Production of activin A by SC was stimulated by 25 and 50 ng/ml TNF (P < 0.01, P < 0.001, respectively) as compared to untreated cells. INHBA mRNA was increased in human testicular biopsies with leukocytic infiltrates and impaired spermatogenesis, compared with control biopsies (P < 0.05), accompanied by increased total collagen (P < 0.01) and fibronectin deposition. Total testicular collagen (P < 0.0001) and fibronectin protein expression (P < 0.05) were also increased in EAO, and fibronectin expression was correlated with the severity of the disease (r = 0.9028). In animals pre-treated with rAAV-FST315 prior to immunization with TH, protein expression of fibronectin was comparable to control. Stimulation of PTC and NIH 3T3 cells with activin A increased fibronectin mRNA (P < 0.05) and the production of collagen I (P < 0.001; P < 0.01) and fibronectin (P < 0.05). Moreover, activin A also increased collagen IV mRNA (P < 0.05) in PTC, while αSMA mRNA (P < 0.01) and protein (P < 0.0001) were significantly increased by activin A in NIH 3T3 cells. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION A limited number of human testicular specimens was available for the study. Part of the study was performed in vitro, including NIH 3T3 cells as a surrogate for testicular fibroblasts. WIDER IMPLICATIONS OF THE FINDINGS Resident fibroblasts and PTC may contribute to the progression of testicular fibrosis following inflammation, and activin A is implicated as a key mediator of this process. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Health and Medical Research Council of Australia, the Victorian Government's Operational Infrastructure Support Program and the International Research Training Group between Justus Liebig University (Giessen) and Monash University (Melbourne) (GRK 1871/1-2) on `Molecular pathogenesis on male reproductive disorders' funded by the Deutsche Forschungsgemeinschaft and Monash University. The authors declare no competing financial interests.
Collapse
Affiliation(s)
- A Christine Kauerhof
- Department of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus Liebig University, Giessen, Germany.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Nour Nicolas
- Department of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany.,Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Australia
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus Liebig University, Giessen, Germany
| | - Eva Wahle
- Department of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus Liebig University, Giessen, Germany
| | - Kate A Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Daniela Fietz
- Hessian Centre of Reproductive Medicine, Justus Liebig University, Giessen, Germany.,Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Martin Bergmann
- Hessian Centre of Reproductive Medicine, Justus Liebig University, Giessen, Germany.,Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Nigel P Groome
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University of Münster, Münster, Germany
| | - Hans-Christian Schuppe
- Hessian Centre of Reproductive Medicine, Justus Liebig University, Giessen, Germany.,Department of Urology, Paediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Adrian Pilatz
- Hessian Centre of Reproductive Medicine, Justus Liebig University, Giessen, Germany.,Department of Urology, Paediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany.,Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Australia
| | - Mark P Hedger
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia.,Shared last authorship
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany.,Hessian Centre of Reproductive Medicine, Justus Liebig University, Giessen, Germany.,Shared last authorship
| |
Collapse
|
14
|
Heinrich A, DeFalco T. Essential roles of interstitial cells in testicular development and function. Andrology 2020; 8:903-914. [PMID: 31444950 PMCID: PMC7036326 DOI: 10.1111/andr.12703] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Testicular architecture and sperm production are supported by a complex network of communication between various cell types. These signals ensure fertility by: regulating spermatogonial stem/progenitor cells; promoting steroidogenesis; and driving male-specific differentiation of the gonad. Sertoli cells have long been assumed to be the major cellular player in testis organogenesis and spermatogenesis. However, cells in the interstitial compartment, such as Leydig, vascular, immune, and peritubular cells, also play prominent roles in the testis but are less well understood. OBJECTIVES Here, we aim to outline our current knowledge of the cellular and molecular mechanisms by which interstitial cell types contribute to spermatogenesis and testicular development, and how these diverse constituents of the testis play essential roles in ensuring male sexual differentiation and fertility. METHODS We surveyed scientific literature and summarized findings in the field that address how interstitial cells interact with other interstitial cell populations and seminiferous tubules (i.e., Sertoli and germ cells) to support spermatogenesis, male-specific differentiation, and testicular function. These studies focused on 4 major cell types: Leydig cells, vascular cells, immune cells, and peritubular cells. RESULTS AND DISCUSSION A growing number of studies have demonstrated that interstitial cells play a wide range of functions in the fetal and adult testis. Leydig cells, through secretion of hormones and growth factors, are responsible for steroidogenesis and progression of spermatogenesis. Vascular, immune, and peritubular cells, apart from their traditionally acknowledged physiological roles, have a broader importance than previously appreciated and are emerging as essential players in stem/progenitor cell biology. CONCLUSION Interstitial cells take part in complex signaling interactions with both interstitial and tubular cell populations, which are required for several biological processes, such as steroidogenesis, Sertoli cell function, spermatogenesis, and immune regulation. These various processes are essential for testicular function and demonstrate how interstitial cells are indispensable for male fertility.
Collapse
Affiliation(s)
- Anna Heinrich
- Division of Reproductive Sciences, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, MLC 7045, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, MLC 7045, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Suite E-870, Cincinnati, OH, 45267, USA
| |
Collapse
|
15
|
Van Saen D, Vloeberghs V, Gies I, De Schepper J, Tournaye H, Goossens E. Characterization of the stem cell niche components within the seminiferous tubules in testicular biopsies of Klinefelter patients. Fertil Steril 2020; 113:1183-1195.e3. [PMID: 32418646 DOI: 10.1016/j.fertnstert.2020.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To characterize the tubular environment in testicular biopsy tissues from patients with Klinefelter syndrome (KS). DESIGN Observational immunohistochemical study. SETTING Academic research unit. PATIENT(S) Males with KS and controls at different developmental time points: fetal, prepubertal, peripubertal, and adult. INTERVENTION(S) Immunohistochemical analysis of testicular biopsies samples to characterize maturation of Sertoli cells and tubular wall components-peritubular myoid cells (PTMC) and extracellular matrix (ECM) proteins. MAIN OUTCOME MEASURE(S) Intensity of antimüllerian hormone staining; proportion of Sertoli cells expressing androgen receptor (AR); and expression of tubular wall markers as characterized by identifying abnormal staining patterns. RESULT(S) Decreased expression for alpha smooth muscle actin 2 (ACTA2) was observed in peripubertal and adult KS as well as in Sertoli cell only (SCO) patients. Altered expression patterns for all ECM proteins were observed in SCO and KS biopsy tissues compared with controls. Only for collagen I and IV were altered expression patterns observed between KS and SCO patients. In peripubertal samples, no statistically significant differences were observed in the maturation markers, but altered ECM patterns were already present in some samples. CONCLUSION(S) The role of loss of ACTA2 expression in PTMC in the disintegration of tubules in KS patients should be further investigated. Future research is necessary to identify the causes of testicular fibrosis in KS patients. If the mechanism behind this fibrotic process could be identified, this process might be altered toward increasing the chances of fertility in KS patients.
Collapse
Affiliation(s)
- Dorien Van Saen
- Biology of the Testis, Laboratory for Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| | - Veerle Vloeberghs
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Inge Gies
- Department of Pediatrics, Division of Pediatric Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jean De Schepper
- Biology of the Testis, Laboratory for Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Department of Pediatrics, Division of Pediatric Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium; Pediatric Endocrinology, Universitair Ziekenhuis Gent, Gent, Belgium
| | - Herman Tournaye
- Biology of the Testis, Laboratory for Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium; Department of Surgical and Clinical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Ellen Goossens
- Biology of the Testis, Laboratory for Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
16
|
The Glucocorticoid Receptor NR3C1 in Testicular Peritubular Cells is Developmentally Regulated and Linked to the Smooth Muscle-Like Cellular Phenotype. J Clin Med 2020; 9:jcm9040961. [PMID: 32244354 PMCID: PMC7230580 DOI: 10.3390/jcm9040961] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/16/2022] Open
Abstract
Whether glucocorticoids (GC) can directly affect human testicular functions is not well understood. A predominant site of GC receptor (GR; NR3C1) expression in the adult testis are peritubular smooth muscle-like cells, which express smooth muscle actin (ACTA2), contract and thereby are involved in sperm transport. In contrast to the adult, neither GR nor ACTA2, or elastin (ELN) were detected in the peritubular compartment before puberty in non-human primate testes. In isolated human testicular peritubular cells (HTPCs), activation of GR by dexamethasone (Dex) caused the translocation of GR to the nucleus and stimulated expression of ACTA2 and ELN, without affecting the expression of collagens. Cytoskeletal ACTA2-rearrangements were observed and were associated with an increased ability to contract. Our results indicate post-pubertal testicular roles of GC in the maintenance of the contractile, smooth muscle-like phenotype of peritubular cells.
Collapse
|
17
|
Schmid N, Flenkenthaler F, Stöckl JB, Dietrich KG, Köhn FM, Schwarzer JU, Kunz L, Luckner M, Wanner G, Arnold GJ, Fröhlich T, Mayerhofer A. Insights into replicative senescence of human testicular peritubular cells. Sci Rep 2019; 9:15052. [PMID: 31636313 PMCID: PMC6803627 DOI: 10.1038/s41598-019-51380-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/27/2019] [Indexed: 01/10/2023] Open
Abstract
There is evidence for an age-related decline in male reproductive functions, yet how the human testis may age is not understood. Human testicular peritubular cells (HTPCs) transport sperm, contribute to the spermatogonial stem cell (SSC) niche and immune surveillance, and can be isolated and studied in vitro. Consequences of replicative senescence of HTPCs were evaluated to gain partial insights into human testicular aging. To this end, early and advanced HTPC passages, in which replicative senescence was indicated by increased cell size, altered nuclear morphology, enhanced β-galactosidase activity, telomere attrition and reduced mitochondrial DNA (mtDNA), were compared. These alterations are typical for senescent cells, in general. To examine HTPC-specific changes, focused ion beam scanning electron microscopy (FIB/SEM) tomography was employed, which revealed a reduced mitochondrial network and an increased lysosome population. The results coincide with the data of a parallel proteomic analysis and indicate deranged proteostasis. The mRNA levels of typical contractility markers and growth factors, important for the SSC niche, were not significantly altered. A secretome analysis identified, however, elevated levels of macrophage migration inhibitory factor (MIF) and dipeptidyl peptidase 4 (DPP4), which may play a role in spermatogenesis. Testicular DPP4 may further represent a possible drug target.
Collapse
Affiliation(s)
- Nina Schmid
- LMU München, Biomedical Center (BMC), Anatomy III - Cell Biology, 82152, Planegg-Martinsried, Germany
| | - Florian Flenkenthaler
- LMU München, Gene Center, Laboratory for Functional Genome Analysis (LAFUGA), 81377 München, Germany
| | - Jan B Stöckl
- LMU München, Gene Center, Laboratory for Functional Genome Analysis (LAFUGA), 81377 München, Germany
| | - Kim-Gwendolyn Dietrich
- LMU München, Biomedical Center (BMC), Anatomy III - Cell Biology, 82152, Planegg-Martinsried, Germany
| | | | | | - Lars Kunz
- LMU München, Department Biology II, Division of Neurobiology, 82152, Planegg-Martinsried, Germany
| | - Manja Luckner
- LMU München, Department Biology I, Ultrastructural Research, 82152, Planegg-Martinsried, Germany
| | - Gerhard Wanner
- LMU München, Department Biology I, Ultrastructural Research, 82152, Planegg-Martinsried, Germany
| | - Georg J Arnold
- LMU München, Gene Center, Laboratory for Functional Genome Analysis (LAFUGA), 81377 München, Germany
| | - Thomas Fröhlich
- LMU München, Gene Center, Laboratory for Functional Genome Analysis (LAFUGA), 81377 München, Germany
| | - Artur Mayerhofer
- LMU München, Biomedical Center (BMC), Anatomy III - Cell Biology, 82152, Planegg-Martinsried, Germany.
| |
Collapse
|
18
|
Mayerhofer A. Peritubular cells of the human testis: prostaglandin E 2 and more. Andrology 2019; 8:898-902. [PMID: 31237067 DOI: 10.1111/andr.12669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Several layers of slender, smooth muscle-like, peritubular cells and extracellular matrix (ECM) form the peritubular compartment of the human testis. Peritubular cells are the least explored testicular cells. MATERIALS AND METHODS Human testicular peritubular cells (HTPCs) can be isolated from small testicular fragments of patients and studied in vitro. We have used this cellular model, in combination with human testicular samples, to examine how peritubular cells may contribute to male (in)fertility. RESULTS Human testicular peritubular cells (HTPCs) retain contractile abilities in vitro and secrete many proteins. Among them are factors, which serve intra-testicular roles, for example, glial cell line-derived neurotrophic factor (GDNF), thought to be important for the renewal of spermatogonial stem cells (SSCs). Studies in mutant mice indicated that peritubular cell-derived GDNF is crucial for lifelong spermatogenesis. Thus, peritubular cells are a functional part of the SSC niche. Peritubular cells of mice and men express androgen receptors (AR). In mouse peritubular cells, androgens enhanced GDNF production, but not in HTPCs. Rather, AR activation increased the levels of AR and smooth muscle proteins and thereby enhanced the smooth muscle-like phenotype. Following the lead of a proteomic analysis, which identified the key prostaglandin (PG)-synthesizing enzyme (PTGS1 = COX1), we found that HTPCs secrete PGE2 . COX1, and PGE2 receptors (EP1, 2, and 4) were identified in peritubular cells in situ, supporting in vivo relevance. In HTPCs, activation of EP1/4 increased GDNF and a smooth muscle protein. Ibuprofen is a nonsteroidal anti-inflammatory drug (NSAID), which blocks PG synthesis. Added to HTPCs it reduced PGE2 and GDNF production and lowered smooth muscle protein levels. If applicable to the in vivo situation, the results suggest that ibuprofen and possibly other NSAIDs may impair important peritubular cell functions and consequently testicular functions. CONCLUSION The few examples highlighted, together with others not mentioned here, indicate that HTPCs provide an experimental window into the human testis.
Collapse
Affiliation(s)
- Artur Mayerhofer
- Biomedical Center Munich (BMC), Cell Biology - Anatomy III, Ludwig-Maximilians-Universität (LMU), Planegg-Martinsried, Germany
| |
Collapse
|
19
|
Rebourcet D, O'Shaughnessy PJ, Smith LB. The expanded roles of Sertoli cells: lessons from Sertoli cell ablation models. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.coemr.2019.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
20
|
Mayerhofer A, Walenta L, Mayer C, Eubler K, Welter H. Human testicular peritubular cells, mast cells and testicular inflammation. Andrologia 2019; 50:e13055. [PMID: 30569646 DOI: 10.1111/and.13055] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/20/2018] [Accepted: 04/29/2018] [Indexed: 12/13/2022] Open
Abstract
In man, the wall of seminiferous tubules forms a testicular compartment, which contains several layers of smooth muscle-like, "myoid", peritubular cells and extracellular matrix. Its architecture and its cellular composition change in male infertility associated with impaired spermatogenesis. Increased deposits of extracellular matrix, changes in the smooth muscle-like phenotype of peritubular cells and accumulation of immune cells, especially mast cells, are among the striking alterations. Taken together, the changes indicate that inflammatory events take place in particular within this compartment. This short review summarises recent studies, which pinpoint possible mechanisms of the interplay between peritubular cells and mast cells, which may contribute to sterile inflammation and impairments of testicular function. These insights are based mainly on cellular studies, for which we used isolated human testicular peritubular cells (HTPCs), and on the examination of human testicular sections. Recent data on immunological properties of peritubular cells, unexpected roles of the extracellular matrix factor, biglycan, which is secreted by peritubular cells and functions of mast cell products (chymase, tryptase and ATP) are presented. We believe that the results may foster a better understanding of peritubular cells, their roles in the human testis and specifically their involvement in infertility.
Collapse
Affiliation(s)
- Artur Mayerhofer
- Anatomy III - Cell Biology, Biomedical Center Munich, LMU München, Planegg-Martinsried, Germany
| | - Lena Walenta
- Anatomy III - Cell Biology, Biomedical Center Munich, LMU München, Planegg-Martinsried, Germany
| | - Christine Mayer
- Anatomy III - Cell Biology, Biomedical Center Munich, LMU München, Planegg-Martinsried, Germany
| | - Katja Eubler
- Anatomy III - Cell Biology, Biomedical Center Munich, LMU München, Planegg-Martinsried, Germany
| | - Harald Welter
- Anatomy III - Cell Biology, Biomedical Center Munich, LMU München, Planegg-Martinsried, Germany
| |
Collapse
|
21
|
Rossi SP, Walenta L, Rey-Ares V, Köhn FM, Schwarzer JU, Welter H, Calandra RS, Frungieri MB, Mayerhofer A. Alpha 1 adrenergic receptor-mediated inflammatory responses in human testicular peritubular cells. Mol Cell Endocrinol 2018; 474:1-9. [PMID: 29407194 DOI: 10.1016/j.mce.2018.01.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/14/2017] [Accepted: 01/29/2018] [Indexed: 01/15/2023]
Abstract
Stress activates the sympathetic nervous system and is linked to impaired fertility in man. We hypothesized that catecholamines by acting on testicular cells have a role in these events, possibly by fostering an inflammatory environment. The cells of the wall of seminiferous tubules, human testicular peritubular cells (HTPCs), express adrenergic receptors (ADRs) α1B, α1D, β1 and β2. A selective α1-ADR agonist, phenylephrine, increased intracellular Ca2+-levels in cultured HTPCs and induced COX-2, IL-6 and MCP-1 mRNA expression without affecting IL-1β mRNA. These changes were paralleled by a significant increase in the secretion of IL-6 and MCP-1. Epinephrine was also effective, but salbutamol, a selective β2-ADR agonist was not. Our results suggest that stress-associated elevation of catecholamines may be able to promote inflammatory events by targeting peritubular cells in the human testis. Blockage of α1-ADRs may therefore be a novel way to interfere with stress-related impairment of male reproductive functions.
Collapse
Affiliation(s)
- Soledad Paola Rossi
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany; Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Lena Walenta
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany
| | - Verónica Rey-Ares
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany
| | | | | | - Harald Welter
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany
| | - Ricardo Saúl Calandra
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Mónica Beatriz Frungieri
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Artur Mayerhofer
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| |
Collapse
|
22
|
Rey-Ares V, Rossi SP, Dietrich KG, Köhn FM, Schwarzer JU, Welter H, Frungieri MB, Mayerhofer A. Prostaglandin E 2 (PGE 2) is a testicular peritubular cell-derived factor involved in human testicular homeostasis. Mol Cell Endocrinol 2018; 473:217-224. [PMID: 29408603 DOI: 10.1016/j.mce.2018.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 12/06/2017] [Accepted: 01/29/2018] [Indexed: 10/18/2022]
Abstract
In man, blockage of prostaglandin (PG)-production e.g. by non-steroidal anti-inflammatory drug (NSAIDs) may have negative testicular side effects, implying beneficial actions of PGs in the testis. We examined human testicular samples and isolated human testicular peritubular cells (HTPCs) to explore sites of PG-synthesis and targets. HTPCs express cyclooxygenase 1 (COX1) and secrete PGE2. Receptors (EP1, 2, 4) were specifically identified in peritubular cells. In HTPCs PGE2 significantly increased mRNA levels of the contractility protein calponin, but did not induce contractions. PGE2, as well as EP1 and EP4 receptor agonists, significantly increased glia cell line derived neurotrophic factor (GDNF) mRNA and/or protein levels. Importantly, the NSAID ibuprofen reduced PGE2 and this action also lowered SMA and calponin mRNA levels and levels of secreted GDNF protein. The results reveal an unknown PGE2 system in the human testis, in involving peritubular cells, which may be prone to interference by NSAIDs.
Collapse
Affiliation(s)
- Verónica Rey-Ares
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| | - Soledad Paola Rossi
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany; Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| | - Kim-Gwendolyn Dietrich
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| | | | | | - Harald Welter
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| | - Mónica Beatriz Frungieri
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| | - Artur Mayerhofer
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| |
Collapse
|
23
|
Mayer C, Adam M, Walenta L, Schmid N, Heikelä H, Schubert K, Flenkenthaler F, Dietrich KG, Gruschka S, Arnold GJ, Fröhlich T, Schwarzer JU, Köhn FM, Strauss L, Welter H, Poutanen M, Mayerhofer A. Insights into the role of androgen receptor in human testicular peritubular cells. Andrology 2018; 6:756-765. [PMID: 29869453 DOI: 10.1111/andr.12509] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 01/07/2023]
Abstract
Contractile smooth muscle-like peritubular cells build the wall of seminiferous tubules in men. They are crucial for sperm transport and complement the functions of Sertoli cells by secreting factors, including glial cell line-derived neurotrophic factor. Previous studies revealed that they also secrete the chemokine C-X-C motif chemokine ligand 12 (CXCL12), which has known roles in spermatogenesis. Peritubular cells express the androgen receptor (AR), which is retained in isolated human testicular peritubular cells. We aimed to explore AR-regulated functions in human testicular peritubular cells. Bearing in mind that infertile men often have high aromatase activity, which may lower intratesticular androgen concentrations, an animal model for male infertility was studied. These mice display an age-dependent loss in spermatogenesis due to high aromatase activity. Human testicular peritubular cells were exposed to dihydrotestosterone or the antiandrogen flutamide. We studied AR, smooth muscle cell markers, glial cell line-derived neurotrophic factor and 15 secreted factors previously identified, including CXCL12. We used qPCR, Western blotting, ELISA or selected reaction monitoring (SRM). In the animal model for male infertility, we employed qPCR and immunohistochemistry. Dihydrotestosterone increased AR and flutamide prevented these actions. The smooth muscle cell markers calponin and smooth muscle actin were likewise increased, while cell size or cellular proliferation was not changed. Dihydrotestosterone did not increase glial cell line-derived neurotrophic factor or CXCL12 secretion but increased levels of serine proteinase inhibitor (SERPIN) E1. The animal model for male infertility with high aromatase activity showed reduced numbers of AR-immunoreactive testicular peritubular cells, suggesting that altered androgen and/or oestrogen levels could influence AR-mediated responses in peritubular cells. Androgens act on human testicular peritubular cells to enhance AR levels, their contractile phenotype and to modulate the secretion of some secreted factors. This study suggests that some aspects of human peritubular cell functions are regulated by androgens.
Collapse
Affiliation(s)
- C Mayer
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - M Adam
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany.,Turku Center for Disease Modeling and Institute of Biomedicine, University of Turku, Turku, Finland
| | - L Walenta
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - N Schmid
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - H Heikelä
- Turku Center for Disease Modeling and Institute of Biomedicine, University of Turku, Turku, Finland
| | - K Schubert
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - F Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Germany
| | - K-G Dietrich
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - S Gruschka
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - G J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Germany
| | - T Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Germany
| | | | | | - L Strauss
- Turku Center for Disease Modeling and Institute of Biomedicine, University of Turku, Turku, Finland
| | - H Welter
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - M Poutanen
- Turku Center for Disease Modeling and Institute of Biomedicine, University of Turku, Turku, Finland
| | - A Mayerhofer
- Cell Biology, Anatomy III, BMC Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| |
Collapse
|
24
|
ATP-mediated Events in Peritubular Cells Contribute to Sterile Testicular Inflammation. Sci Rep 2018; 8:1431. [PMID: 29362497 PMCID: PMC5780482 DOI: 10.1038/s41598-018-19624-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/03/2018] [Indexed: 12/19/2022] Open
Abstract
Peritubular myoid cells, which form the walls of seminiferous tubules in the testis, are functionally unexplored. While they transport sperm and contribute to the spermatogonial stem cell niche, specifically their emerging role in the immune surveillance of the testis and in male infertility remains to be studied. Recently, cytokine production and activation of Toll-like receptors (TLRs) were uncovered in cultured peritubular cells. We now show that human peritubular cells express purinergic receptors P2RX4 and P2RX7, which are functionally linked to TLRs, with P2RX4 being the prevalent ATP-gated ion channel. Subsequent ATP treatment of cultured peritubular cells resulted in up-regulated (pro-)inflammatory cytokine expression and secretion, while characteristic peritubular proteins, that is smooth muscle cell markers and extracellular matrix molecules, decreased. These findings indicate that extracellular ATP may act as danger molecule on peritubular cells, able to promote inflammatory responses in the testicular environment.
Collapse
|
25
|
Mincheva M, Sandhowe-Klaverkamp R, Wistuba J, Redmann K, Stukenborg JB, Kliesch S, Schlatt S. Reassembly of adult human testicular cells: can testis cord-like structures be created in vitro? Mol Hum Reprod 2017; 24:55-63. [DOI: 10.1093/molehr/gax063] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/11/2017] [Indexed: 02/06/2023] Open
Affiliation(s)
- M Mincheva
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - R Sandhowe-Klaverkamp
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - J Wistuba
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - K Redmann
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - J -B Stukenborg
- Department of Women’s and Children’s Health, NORDFERTIL research lab Stockholm, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, SE-17176 Stockholm, Sweden
| | - S Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - S Schlatt
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| |
Collapse
|
26
|
Xia H, Chen Y, Wu KJ, Zhao H, Xiong CL, Huang DH. Role of C-type natriuretic peptide in the function of normal human sperm. Asian J Androl 2016; 18:80-4. [PMID: 25926602 PMCID: PMC4736361 DOI: 10.4103/1008-682x.150254] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
C-type natriuretic peptide (CNP) is a newly discovered type of local regulatory factor that mediates its biological effects through the specific, membrane-bound natriuretic peptide receptor-B (NPR-B). Recent studies have established that CNP is closely related to male reproductive function. The aims of this study were to determine the distribution of CNP/NPR-B in human ejaculated spermatozoa through different methods (such as immunolocalization, real time polymerase chain reaction and Western Blot), and then to evaluate the influence of CNP on sperm function i n vitro, such as motility and acrosome reaction. Human semen samples were collected from consenting donors who met the criteria of the World Health Organization for normozoospermia. Our results show that the specific receptor NPR-B of CNP is localized in the acrosomal region of the head and the membrane of the front-end tail of the sperm, and there is no signal of CNP in human sperm. Compared with the control, CNP can induce a significant dose-dependent increase in spermatozoa motility and acrosome reaction. In summary, CNP/NPR-B can affect sperm motility and acrosome reaction, thus regulating the reproductive function of males. CNP may be a new key factor in regulating sperm function.
Collapse
Affiliation(s)
| | | | | | | | | | - Dong-Hui Huang
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
27
|
Maqdasy S, El Hajjaji FZ, Baptissart M, Viennois E, Oumeddour A, Brugnon F, Trousson A, Tauveron I, Volle D, Lobaccaro JMA, Baron S. Identification of the Functions of Liver X Receptor-β in Sertoli Cells Using a Targeted Expression-Rescue Model. Endocrinology 2015; 156:4545-57. [PMID: 26402841 DOI: 10.1210/en.2015-1382] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Liver X receptors (LXRs) are key regulators of lipid homeostasis and are involved in multiple testicular functions. The Lxrα(-/-);Lxrβ(-/-) mice have illuminated the roles of both isoforms in maintenance of the epithelium in the seminiferous tubules, spermatogenesis, and T production. The requirement for LXRβ in Sertoli cells have been emphasized by early abnormal cholesteryl ester accumulation in the Lxrβ(-/-) and Lxrα(-/-);Lxrβ(-/-) mice. Other phenotypes, such as germ cell loss and hypogonadism, occur later in life in the Lxrα(-/-);Lxrβ(-/-) mice. Thus, LXRβ expression in Sertoli cells seems to be essential for normal testicular physiology. To decipher the roles of LXRβ within the Sertoli cells, we generated Lxrα(-/-);Lxrβ(-/-):AMH-Lxrβ transgenic mice, which reexpress Lxrβ in Sertoli cells in the context of Lxrα(-/-);Lxrβ(-/-) mice. In addition to lipid homeostasis, LXRβ is necessary for maintaining the blood-testis barrier and the integrity of the germ cell epithelium. LXRβ is also implicated in the paracrine action of Sertoli cells on Leydig cells to modulate T synthesis. The Lxrα(-/-);Lxrβ(-/-) and Lxrα(-/-);Lxrβ(-/-):AMH-Lxrβ mice exhibit lipid accumulation in germ cells after the Abcg8 down-regulation, suggesting an intricate LXRβ-dependent cooperation between the Sertoli cells and germ cells to ensure spermiogenesis. Further analysis revealed also peritubular smooth muscle defects (abnormal lipid accumulation and disorganized smooth muscle actin) and spermatozoa stagnation in the seminiferous tubules. Together the present work elucidates specific roles of LXRβ in Sertoli cell physiology in vivo beyond lipid homeostasis.
Collapse
Affiliation(s)
- Salwan Maqdasy
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Fatim-Zohra El Hajjaji
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Marine Baptissart
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Emilie Viennois
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Abdelkader Oumeddour
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Florence Brugnon
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Amalia Trousson
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Igor Tauveron
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - David Volle
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Jean-Marc A Lobaccaro
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Silvère Baron
- Department of Génétique Reproduction et Développement (GReD) (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Université Blaise Pascal, Centre de Recherche en Nutrition Humaine d'Auvergne (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., D.V., J.-M.A.L., S.B.), and Department of Assistance Médicale à la Procréation (F.B.), CECOS, Centre Hospitalier Universitaire Clermont Ferrand, Centre Hospitalier Universitaire Estaing, F-63000 Clermont-Ferrand, France; Centre National de la Recherche Scientifique (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.) and INSERM (S.M., F.-Z.E.H., M.B., A.O., F.B., A.T., I.T., D.V., J.-M.A.L., S.B.), Unité Mixte de Recherche 6293, GReD, F-63177 Aubiere, France; Center for Diagnostics and Therapeutics (E.V.), Georgia State University, Atlanta, Georgia 30302-4010; Veterans Affairs Medical Center (E.V.), Decatur, Georgia 30033; Service d'Endocrinologie, Diabétologie, et Maladies Métaboliques (S.M., I.T.), Hôpital Gabriel Montpied, F-63003 Clermont-Ferrand, France; and Service de Médecine Nucléaire (S.M.), Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| |
Collapse
|
28
|
Nygaard MB, Almstrup K, Lindbæk L, Christensen ST, Svingen T. Cell context-specific expression of primary cilia in the human testis and ciliary coordination of Hedgehog signalling in mouse Leydig cells. Sci Rep 2015; 5:10364. [PMID: 25992706 PMCID: PMC4438617 DOI: 10.1038/srep10364] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/09/2015] [Indexed: 12/04/2022] Open
Abstract
Primary cilia are sensory organelles that coordinate numerous cellular signalling pathways during development and adulthood. Defects in ciliary assembly or function lead to a series of developmental disorders and diseases commonly referred to as ciliopathies. Still, little is known about the formation and function of primary cilia in the mammalian testis. Here, we characterized primary cilia in adult human testis and report a constitutive expression of cilia in peritubular myoid cells and a dynamic expression of cilia in differentiating Leydig cells. Primary cilia are generally absent from cells of mature seminiferous epithelium, but present in Sertoli cell-only tubules in Klinefelter syndrome testis. Peritubular cells in atrophic testis produce overly long cilia. Furthermore cultures of growth-arrested immature mouse Leydig cells express primary cilia that are enriched in components of Hedgehog signalling, including Smoothened, Patched-1, and GLI2, which are involved in regulating Leydig cell differentiation. Stimulation of Hedgehog signalling increases the localization of Smoothened to the cilium, which is followed by transactivation of the Hedgehog target genes, Gli1 and Ptch1. Our findings provide new information on the spatiotemporal formation of primary cilia in the testis and show that primary cilia in immature Leydig cells mediate Hedgehog signalling.
Collapse
Affiliation(s)
- Marie Berg Nygaard
- 1] University Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen DK-2100, Denmark [2] Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Kristian Almstrup
- University Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen DK-2100, Denmark
| | - Louise Lindbæk
- Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | | | - Terje Svingen
- 1] University Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen DK-2100, Denmark [2] Department of Toxicology and Risk Assessment, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| |
Collapse
|
29
|
Frungieri MB, Calandra RS, Mayerhofer A, Matzkin ME. Cyclooxygenase and prostaglandins in somatic cell populations of the testis. Reproduction 2015; 149:R169-80. [DOI: 10.1530/rep-14-0392] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prostaglandins (PGs) are synthesized through the action of the rate-limiting enzyme cyclooxygenase (COX) and further specific enzymes. The development ofCox-deficient mice in the 1990s gave insights into the reproductive roles of PGs. FemaleCox-knockout mice were subfertile or infertile. Interestingly, fertility was not affected in male mice deficient inCox, suggesting that PGs may not be critical for the functioning of the testis. However, this conclusion has recently been challenged by observations of important roles for PGs in both physiological and pathological processes in the testis. The two key somatic cell types in the testis, Leydig and Sertoli cells, express the inducible isoenzyme COX2 and produce PGs. Testicular COX2 expression in these somatic cells is regulated by hormonal input (FSH, prolactin (PRL), and testosterone) as well as by IL1β. PGs modulate steroidogenesis in Leydig cells and glucose uptake in Sertoli cells. Hence, the COX2/PG system in Leydig and Sertoli cells acts as a local modulator of testicular activity, and consequently may regulate spermatogenic efficiency. In addition to its expression in Leydig and Sertoli cells, COX2 has been detected in the seminiferous tubule wall, and in testicular macrophages and mast cells of infertile patients. These observations highlight the possible relevance of PGs in testicular inflammation associated with idiopathic infertility. Collectively, these data indicate that the COX2/PG system plays crucial roles not only in testicular physiology (i.e., development, steroidogenesis, and spermatogenesis), but more importantly in the pathogenesis or maintenance of infertility status in the male gonad. Further studies of these actions could lead to new therapeutic approaches to idiopathic male infertility.Free German abstractA German translation of this abstract is freely available athttp://www.reproduction-online.org/content/149/4/R169/suppl/DC1.Free Spanish abstractA Spanish translation of this abstract is freely available athttp://www.reproduction-online.org/content/149/4/R169/suppl/DC2.
Collapse
|
30
|
Rossitto M, Ujjan S, Poulat F, Boizet-Bonhoure B. Multiple roles of the prostaglandin D2 signaling pathway in reproduction. Reproduction 2015; 149:R49-58. [DOI: 10.1530/rep-14-0381] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Prostaglandins signaling molecules are involved in numerous physiological processes. They are produced by several enzyme-limited reactions upon fatty acids, which are catalyzed by two cyclooxygenases and prostaglandin synthases. In particular, the prostaglandins E2(PGE2), D2(PGD2), and F2(PGF2α) have been shown to be involved in female reproductive mechanisms. Furthermore, widespread expression of lipocalin- and hematopoietic-PGD2synthases in the male reproductive tract supports the purported roles of PGD2in the development of both embryonic and adult testes, sperm maturation, and spermatogenesis. In this review, we summarize the putative roles of PGD2signaling and the roles of both PGD2synthases in testicular formation and function. We review the data reporting the involvement of PGD2signaling in the differentiation of Sertoli and germ cells of the embryonic testis. Furthermore, we discuss the roles of lipocalin-PGD2synthase in steroidogenesis and spermatogenesis, in terms of lipid molecule transport and PGD2production. Finally, we discuss the hypothesis that PGD2signaling may be affected in certain reproductive diseases, such as infertility, cryptorchidism, and testicular cancer.
Collapse
|
31
|
Welter H, Huber A, Lauf S, Einwang D, Mayer C, Schwarzer JU, Köhn FM, Mayerhofer A. Angiotensin II regulates testicular peritubular cell function via AT1 receptor: a specific situation in male infertility. Mol Cell Endocrinol 2014; 393:171-8. [PMID: 24970685 DOI: 10.1016/j.mce.2014.06.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/28/2014] [Accepted: 06/16/2014] [Indexed: 10/25/2022]
Abstract
We observed that peritubular myoid cells in the human testis are immunoreactive for angiotensin II (AngII) receptors (AT1R) and explored AngII actions in cultured human testicular peritubular cells (HTPCs). In response to AngII they contracted within minutes. The AT1R-blocker losartan blocked contraction, implying involvement of AngII and AT1R in intratesticular sperm transport. AngII also significantly increased IL-6 mRNA levels and IL-6 secretion within hours and losartan again prevented this action. This suggests involvement in inflammatory processes, which may play a role in male infertility. AngII can be generated locally by mast cell (MC)-derived chymase (CHY), which cleaves AngI. In testicular biopsies from infertile men we found abundant MCs, which express CHY, within the wall of seminiferous tubules. In contrast, CHY-positive MCs are hardly found in normal human testis. Testicular inflammatory events may fuel processes resulting in impaired spermatogenesis. Therefore therapeutic interference with MCs, CHY or AT1R might be novel options in male infertility.
Collapse
Affiliation(s)
- H Welter
- Anatomy III - Cell Biology, Ludwig Maximilian University, Schillerstrasse 42, 80336 Munich, Germany.
| | - A Huber
- Anatomy III - Cell Biology, Ludwig Maximilian University, Schillerstrasse 42, 80336 Munich, Germany
| | - S Lauf
- Anatomy III - Cell Biology, Ludwig Maximilian University, Schillerstrasse 42, 80336 Munich, Germany
| | - D Einwang
- Anatomy III - Cell Biology, Ludwig Maximilian University, Schillerstrasse 42, 80336 Munich, Germany
| | - C Mayer
- Anatomy III - Cell Biology, Ludwig Maximilian University, Schillerstrasse 42, 80336 Munich, Germany
| | | | - F M Köhn
- Andrologicum, 80331 Munich, Germany
| | - A Mayerhofer
- Anatomy III - Cell Biology, Ludwig Maximilian University, Schillerstrasse 42, 80336 Munich, Germany.
| |
Collapse
|
32
|
Sandner F, Welter H, Schwarzer JU, Köhn FM, Urbanski HF, Mayerhofer A. Expression of the oestrogen receptor GPER by testicular peritubular cells is linked to sexual maturation and male fertility. Andrology 2014; 2:695-701. [PMID: 25052196 DOI: 10.1111/j.2047-2927.2014.00243.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 05/08/2014] [Accepted: 06/02/2014] [Indexed: 12/01/2022]
Abstract
Besides the two nuclear oestrogen receptors (ESR1/ESR2), the G protein-coupled oestrogen receptor (GPER) was described in the human testis but little is known about testicular GPER during development or male infertility. We performed an immunohistochemical analysis using human and rhesus monkey testicular samples. The results obtained in adult primate testes showed GPER in interstitial and vascular cells as well as in smooth muscle-like peritubular cells, which build the wall of seminiferous tubules. Expression of GPER was also found in cultured human testicular peritubular cells (HPTCs) by Western blotting and RT-PCR/sequencing. Furthermore, as seen in time-lapse videos of cultured cells, addition of a specific GPER agonist (G1) significantly reduced the numbers of HTPCs within 24 h. A GPER antagonist (G15) prevented this action, implying a role for GPER related to the control of cell proliferation or cell death of peritubular cells. Peritubular cell functions and their phenotype change, for example, during post-natal development and in the cases of male infertility. The study of non-human primate samples revealed that GPER in peritubular cells was detectable only from the time of puberty onwards, while in samples from infantile and prepubertal monkeys only interstitial cells showed immunopositive staining. In testicular biopsies of men with mixed atrophy, a reduction or loss of immunoreactive GPER was found in peritubular cells surrounding those tubules, in which spermatogenesis was impaired. In other cases of impaired spermatogenesis, namely when the tubular wall was fibrotically remodelled, a complete loss of GPER was seen. Thus, the observed inverse relation between the state of fertility and GPER expression by peritubular cells implies that the regulation of primate testicular peritubular cells by oestrogens is mediated by GPER in both, health and disease.
Collapse
Affiliation(s)
- F Sandner
- Anatomy III, Cell Biology, LMU München, München, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Reuter K, Ehmcke J, Stukenborg JB, Simoni M, Damm OS, Redmann K, Schlatt S, Wistuba J. Reassembly of somatic cells and testicular organogenesis in vitro. Tissue Cell 2013; 46:86-96. [PMID: 24411714 DOI: 10.1016/j.tice.2013.12.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/30/2013] [Accepted: 12/03/2013] [Indexed: 02/07/2023]
Abstract
Testicular organogenesis in vitro requires an environment allowing a reassembly of testicular cell types. Previous in vitro studies using male murine germ cells cultured in a defined three-dimensional environment demonstrated tubulogenesis and differentiation into spermatozoa. Combining scaffolds as artificial culture substrates with testicular cell culture, we analysed the colonization of collagen sponges by rat testicular cells focusing on cell survival and reassembly of tubule-like-structures in vitro. Isolated testicular cells obtained from juvenile Sprague Dawley and eGFP transgenic rats were cultured on collagen sponges (DMEM high glucose+Glutamax, 35°C, 5% CO2 with or without gonadotropins). Live cell imaging revealed the colonization of cells across the entire scaffold for up to 35 days. After two days, histology showed cell clusters attached to the collagen fibres and displaying signs of tubulogenesis. Clusters consisted mainly of Sertoli and peritubular cells which surrounded some undifferentiated spermatogonia. Flow cytometry confirmed lack of differentiation as no haploid cells were detected. Leydig cell activity was detected by a rise of testosterone after gonadotropin stimulation. Our approach provides a novel method which is in particular suitable to follow the somatic testicular cells in vitro an issue of growing importance for the analysis of germ line independent failure of spermatogenesis.
Collapse
Affiliation(s)
- Karin Reuter
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Clinics, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Jens Ehmcke
- Central Animal Facility of the Faculty of Medicine, University of Münster, Albert-Schweitzer-Campus 1 Building A8, 48149 Münster, Germany
| | - Jan-Bernd Stukenborg
- Department of Women's and Children's Health, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, 17176 Stockholm, Sweden
| | - Manuela Simoni
- University of Modena and Reggio Emilia, Department of Medicine, Metabolism and Neural Sciences, NOCSAE, Via Giardini 1355, I-41126 Modena, Italy
| | - Oliver S Damm
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Clinics, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Klaus Redmann
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Clinics, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Stefan Schlatt
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Clinics, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Joachim Wistuba
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Clinics, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany.
| |
Collapse
|
34
|
Abstract
In healthy men, several layers of inconspicuously flat cells and extracellular matrix (ECM) proteins build the wall of the seminiferous tubules. The cells of this wall, peritubular cells, are not well characterized. They are smooth-muscle-like and contractile and transport immotile sperm, a function important for male fertility. However, their full functional importance, especially their potential contribution to the paracrine regulation of the male gonad, is unknown. In men with impaired spermatogenesis, the architecture of the tubular wall is frequently altered. Deposits of ECM and morphological changes of peritubular cells imply that functions of peritubular cells may be fundamentally altered. To be able to study human peritubular cells and their functions, a culture method was established. It is based on small biopsies of patients with obstructive azoospermia but normal spermatogenesis (human testicular peritubular cells, HTPCs) and non-obstructive azoospermia, impaired spermatogenesis, and testicular fibrosis (HTPCFs). Results obtained from cellular studies and parallel examinations of biopsies provide insights into the repertoire of the secretion products, contractile properties, and plasticity of human peritubular cells. They produce ECM components, including the proteoglycan decorin, which may influence paracrine signaling between testicular cells. They may contribute to the spermatogonial stem cell niche via secreted factors. They are regulated by mast cell and macrophage products, and in response produce factors that can fuel inflammatory changes. They possess a high degree of plasticity, which results in hypertrophy and loss of contractile abilities. The data collectively indicate important roles of inconspicuous testicular peritubular cells in human male fertility and infertility.
Collapse
Affiliation(s)
- Artur Mayerhofer
- Anatomy III-Cell Biology, Ludwig Maximilian University Munich, Biedersteiner Strasse 29, D-80802 Munich, Germany.
| |
Collapse
|