1
|
Kim YY, Kwak J, Kang BC, Ku SY. Non-human primate: the new frontier model of female reproductive engineering. Front Bioeng Biotechnol 2025; 13:1536750. [PMID: 40242357 PMCID: PMC12001037 DOI: 10.3389/fbioe.2025.1536750] [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: 11/29/2024] [Accepted: 03/13/2025] [Indexed: 04/18/2025] Open
Abstract
Reproductive engineering encompasses a range of advanced tissue engineering techniques aimed at addressing infertility that is non-curable with current assisted reproductive technology (ART). The use of animal models has been crucial for these advancements, with a notable preference for non-human primates (NHPs) given their genetic, anatomical, and physiological similarities to humans. Therefore, NHPs are invaluable for studying reproductive engineering. Thus, in reproductive studies, NHPs bridge the anatomical and physiological gaps between rodent models and humans. Their shared features with humans, such as menstrual cycles, placentation, and hormonal regulation, allow for more accurate modeling of reproductive physiology and pathology. These traits make NHPs indispensable in the exploration of reproductive engineering, including infertility treatments, genetic engineering, and uterine transplantation. Reproductive engineering is a transformative field that addresses infertility and enhances reproductive health. By leveraging the unique traits of NHPs, researchers can deepen their understanding of reproductive processes and refine ART techniques for human use. Advances in genetic engineering have enabled the creation of transgenic NHP models, which have been used to modify genes to investigate roles for various purposes, and the process, as mentioned earlier, is closely related to the ART technique, including fertility, embryogenesis, and pregnancy. Therefore, the relation to reproductive studies and the necessity of the NHP model are prerequisites for reproductive engineering. The engineering of NHPs is critically related to integrating ethical practices and exploring complementary methodologies. This review overviews the types of NHP frequently used and studies using NHP for reproductive engineering. These studies may suggest a broader way to use NHP for reproductive engineering.
Collapse
Affiliation(s)
- Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Jina Kwak
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Byeong-Cheol Kang
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Translational Medicine, Seoul, Republic of Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Meng J, Feng J, Xiao L, Hu N, Lan X, Wang S. Oral vaccination with inhibin DNA vaccine for promoting spermatogenesis in rats. Anim Reprod 2024; 21:e20230079. [PMID: 39371539 PMCID: PMC11452157 DOI: 10.1590/1984-3143-ar2023-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/28/2024] [Indexed: 10/08/2024] Open
Abstract
The objective of the present study was to evaluate the effects of a novel Inhibin (INH) DNA vaccine (C500/pVAX-asd-IS) on the immune response, reproductive hormone levels, and spermatogenesis of rats. Forty healthy male rats were divided into four groups, and respectively immunized (thrice, 14 d apart) with 1×108, 1×109, and 1×1010 CFU of the recombinant inhibin vaccine (group C500/pVAX-asd-IS-L, C500/pVAX-asd-IS-M, and C500/pVAX-asd-IS-H) or 1×1010 CFU C500. P/N values increased after vaccination and differed (p <0.05) at 7 d, and sharply increased at 14 d following the booster vaccination (p <0.01); The weight and volume of testes in C500/pVAX-asd-IS groups were increased (p < 0.05) at decapitation, respectively; Histological evaluation showed that the number of spermatogenic cells in the lumen was increased, and the cytoplasmic remnants of sperms were allergy increased significantly compared with the control group. Oral vaccination with INH DNA reduced (P < 0.05) serum concentrations of INH B, enhanced serum concentrations of testosterone (T) and FSH. Furthermore, mRNA expressions of VIM and SMAD4 in the testes were increased in C500/pVAX-asd-IS-M and C500/pVAX-asd-IS-H groups (p < 0.05 or p < 0.01). The mRNA amount of INHβ-B in C500/pVAX-asd-IS-M group was greater than control group (p < 0.05).These results suggested that neutralization of endogenous INH through oral vaccination with INH DNA delivered by C500 strain successfully elicited a humoral immune response. INH gene immunization may have a positive effect on spermatogenesis and reproductive efficiency in male rats.
Collapse
Affiliation(s)
- Jinzhu Meng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Guizhou Provincial Key Laboratory for Biodiversity Conservation and Utilization in the Fanjing Mountain Region, Tongren University, Tongren, China
| | - Jianhao Feng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Lilin Xiao
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Nan Hu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Xianyong Lan
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, China
| | - Shuilian Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| |
Collapse
|
3
|
Stallmeyer B, Bühlmann C, Stakaitis R, Dicke AK, Ghieh F, Meier L, Zoch A, MacKenzie MacLeod D, Steingröver J, Okutman Ö, Fietz D, Pilatz A, Riera-Escamilla A, Xavier MJ, Ruckert C, Di Persio S, Neuhaus N, Gurbuz AS, Şalvarci A, Le May N, McEleny K, Friedrich C, van der Heijden G, Wyrwoll MJ, Kliesch S, Veltman JA, Krausz C, Viville S, Conrad DF, O'Carroll D, Tüttelmann F. Inherited defects of piRNA biogenesis cause transposon de-repression, impaired spermatogenesis, and human male infertility. Nat Commun 2024; 15:6637. [PMID: 39122675 PMCID: PMC11316121 DOI: 10.1038/s41467-024-50930-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
piRNAs are crucial for transposon silencing, germ cell maturation, and fertility in male mice. Here, we report on the genetic landscape of piRNA dysfunction in humans and present 39 infertile men carrying biallelic variants in 14 different piRNA pathway genes, including PIWIL1, GTSF1, GPAT2, MAEL, TDRD1, and DDX4. In some affected men, the testicular phenotypes differ from those of the respective knockout mice and range from complete germ cell loss to the production of a few morphologically abnormal sperm. A reduced number of pachytene piRNAs was detected in the testicular tissue of variant carriers, demonstrating impaired piRNA biogenesis. Furthermore, LINE1 expression in spermatogonia links impaired piRNA biogenesis to transposon de-silencing and serves to classify variants as functionally relevant. These results establish the disrupted piRNA pathway as a major cause of human spermatogenic failure and provide insights into transposon silencing in human male germ cells.
Collapse
Affiliation(s)
- Birgit Stallmeyer
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Clara Bühlmann
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Rytis Stakaitis
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ann-Kristin Dicke
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Farah Ghieh
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Luisa Meier
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Ansgar Zoch
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
- Wellcome Centre for Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - David MacKenzie MacLeod
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
- Wellcome Centre for Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Johanna Steingröver
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Özlem Okutman
- Laboratoire de Génétique Médicale LGM, institut de génétique médicale d'Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, Strasbourg, France
- Hôpital Universitaire de Bruxelles, Hôpital Erasme, Service de Gynécologie-Obstétrique, Clinique de Fertilité, Université libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Daniela Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Adrian Pilatz
- Clinic for Urology, Paediatric Urology and Andrology, Justus Liebig University Gießen, Gießen, Germany
| | - Antoni Riera-Escamilla
- Andrology Department, Fundació Puigvert, Universitat Autònoma de Barcelona, Instituto de Investigaciones Biomédicas Sant Pau, Barcelona, Catalonia, Spain
| | - Miguel J Xavier
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Christian Ruckert
- Centre of Medical Genetics, Department of Medical Genetics, University of Münster, Münster, Germany
| | - Sara Di Persio
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University Hospital Münster, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University Hospital Münster, Münster, Germany
| | - Ali Sami Gurbuz
- Department of Gynecology and Obstetrics Novafertil IVF Center, Konya, Turkey
| | - Ahmet Şalvarci
- Department of Andrology Novafertil IVF Center, Konya, Turkey
| | - Nicolas Le May
- Laboratoire de Génétique Médicale LGM, institut de génétique médicale d'Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, Strasbourg, France
| | - Kevin McEleny
- Newcastle Fertility Centre, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Corinna Friedrich
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Godfried van der Heijden
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Margot J Wyrwoll
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University Hospital Münster, Münster, Germany
| | - Joris A Veltman
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Csilla Krausz
- Andrology Department, Fundació Puigvert, Universitat Autònoma de Barcelona, Instituto de Investigaciones Biomédicas Sant Pau, Barcelona, Catalonia, Spain
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, University Hospital Careggi, Florence, Italy
| | - Stéphane Viville
- Laboratoire de Génétique Médicale LGM, institut de génétique médicale d'Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, Strasbourg, France
- Laboratoire de Diagnostic Génétique, UF3472-génétique de l'infertilité, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Donald F Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Dónal O'Carroll
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
- Wellcome Centre for Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Frank Tüttelmann
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany.
| |
Collapse
|
4
|
Zoch A, Konieczny G, Auchynnikava T, Stallmeyer B, Rotte N, Heep M, Berrens RV, Schito M, Kabayama Y, Schöpp T, Kliesch S, Houston B, Nagirnaja L, O'Bryan MK, Aston KI, Conrad DF, Rappsilber J, Allshire RC, Cook AG, Tüttelmann F, O'Carroll D. C19ORF84 connects piRNA and DNA methylation machineries to defend the mammalian germ line. Mol Cell 2024; 84:1021-1035.e11. [PMID: 38359823 PMCID: PMC10960678 DOI: 10.1016/j.molcel.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/01/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024]
Abstract
In the male mouse germ line, PIWI-interacting RNAs (piRNAs), bound by the PIWI protein MIWI2 (PIWIL4), guide DNA methylation of young active transposons through SPOCD1. However, the underlying mechanisms of SPOCD1-mediated piRNA-directed transposon methylation and whether this pathway functions to protect the human germ line remain unknown. We identified loss-of-function variants in human SPOCD1 that cause defective transposon silencing and male infertility. Through the analysis of these pathogenic alleles, we discovered that the uncharacterized protein C19ORF84 interacts with SPOCD1. DNMT3C, the DNA methyltransferase responsible for transposon methylation, associates with SPOCD1 and C19ORF84 in fetal gonocytes. Furthermore, C19ORF84 is essential for piRNA-directed DNA methylation and male mouse fertility. Finally, C19ORF84 mediates the in vivo association of SPOCD1 with the de novo methylation machinery. In summary, we have discovered a conserved role for the human piRNA pathway in transposon silencing and C19ORF84, an uncharacterized protein essential for orchestrating piRNA-directed DNA methylation.
Collapse
Affiliation(s)
- Ansgar Zoch
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Institute for Stem Cell Research, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK.
| | - Gabriela Konieczny
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Institute for Stem Cell Research, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Tania Auchynnikava
- Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Birgit Stallmeyer
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Nadja Rotte
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Madeleine Heep
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Institute for Stem Cell Research, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Rebecca V Berrens
- Institute for Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Drive, Oxford OX37TY, UK
| | - Martina Schito
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Institute for Stem Cell Research, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Yuka Kabayama
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Institute for Stem Cell Research, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Theresa Schöpp
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Institute for Stem Cell Research, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University Hospital Münster, Münster, Germany
| | - Brendan Houston
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Liina Nagirnaja
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Kenneth I Aston
- Andrology and In Vitro Fertilization Laboratory, Department of Surgery (Urology), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Donald F Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA; Center for Embryonic Cell and Gene Therapy, Oregon Health and Science University, Portland, OR, USA
| | - Juri Rappsilber
- Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK; Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Robin C Allshire
- Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Atlanta G Cook
- Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Dónal O'Carroll
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Institute for Stem Cell Research, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK.
| |
Collapse
|
5
|
Bayurova E, Zhitkevich A, Avdoshina D, Kupriyanova N, Kolyako Y, Kostyushev D, Gordeychuk I. Common Marmoset Cell Lines and Their Applications in Biomedical Research. Cells 2023; 12:2020. [PMID: 37626830 PMCID: PMC10453182 DOI: 10.3390/cells12162020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/19/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Common marmosets (Callithrix jacchus; CMs) are small New World primates widely used in biomedical research. Early stages of such research often include in vitro experiments which require standardized and well-characterized CM cell cultures derived from different tissues. Despite the long history of laboratory work with CMs and high translational potential of such studies, the number of available standardized, well-defined, stable, and validated CM cell lines is still small. While primary cells and immortalized cell lines are mostly used for the studies of infectious diseases, biochemical research, and targeted gene therapy, the main current applications of CM embryonic stem cells and induced pluripotent stem cells are regenerative medicine, stem cell research, generation of transgenic CMs, transplantology, cell therapy, reproductive physiology, oncology, and neurodegenerative diseases. In this review we summarize the data on the main advantages, drawbacks and research applications of CM cell lines published to date including primary cells, immortalized cell lines, lymphoblastoid cell lines, embryonic stem cells, and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Ekaterina Bayurova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
| | - Alla Zhitkevich
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
| | - Daria Avdoshina
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
| | - Natalya Kupriyanova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
- Institute for Translational Medicine and Biotechnology, Sechenov University, 117418 Moscow, Russia
| | - Yuliya Kolyako
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
- Institute for Translational Medicine and Biotechnology, Sechenov University, 117418 Moscow, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119435 Moscow, Russia;
- Scientific Center for Genetics and Life Sciences, Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Ilya Gordeychuk
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
- Institute for Translational Medicine and Biotechnology, Sechenov University, 117418 Moscow, Russia
| |
Collapse
|
6
|
da Silveira Firmiano EM, Machado‐Santos C, Ribeiro Ricardo Brito A, Sousa BM, Lima Pinheiro N, das Neves Cardoso N, Alves do Nascimento A. Histological study and immunohistochemical location of cytoskeletal proteins in the testis and epididymis of the three species of lizards of the family Leiosauridae (Reptilia: Squamata). ACTA ZOOL-STOCKHOLM 2022. [DOI: 10.1111/azo.12422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Enely Maris da Silveira Firmiano
- Postgraduate Program in Animal Biology, Institute of Biological and Health Sciences Federal Rural University of Rio de Janeiro‐UFRRJ Seropédica Brazil
| | - Clarice Machado‐Santos
- Laboratory of Teaching and Research in Histology and Compared Embryology (LEPHEC) Federal Fluminense University Niterói Brazil
| | - Amanda Ribeiro Ricardo Brito
- Laboratory of Teaching and Research in Histology and Compared Embryology (LEPHEC) Federal Fluminense University Niterói Brazil
| | - Bernadete Maria Sousa
- Herpetology Laboratory, Department of Zoology, Institute of Biological Sciences, Federal University of Juiz de Fora‐UFJF University Campus noc number Juiz de Fora Brazil
| | - Nadja Lima Pinheiro
- Postgraduate Program in Animal Biology, Institute of Biological and Health Sciences Federal Rural University of Rio de Janeiro‐UFRRJ Seropédica Brazil
| | - Nathália das Neves Cardoso
- Postgraduate Program in Animal Biology, Institute of Biological and Health Sciences Federal Rural University of Rio de Janeiro‐UFRRJ Seropédica Brazil
| | - Aparecida Alves do Nascimento
- Postgraduate Program in Animal Biology, Institute of Biological and Health Sciences Federal Rural University of Rio de Janeiro‐UFRRJ Seropédica Brazil
| |
Collapse
|
7
|
Sertoli, Leydig, and Spermatogonial Cells’ Specific Gene and Protein Expressions as Dog Testes Evolve from Immature into Mature States. Animals (Basel) 2022; 12:ani12030271. [PMID: 35158595 PMCID: PMC8833615 DOI: 10.3390/ani12030271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
Sertoli, Leydig, and spermatogonial cells proliferate and differentiate from birth to puberty and then stay stable in adulthood. We hypothesized that expressions of spermatogenesis-associated genes are not enhanced with a mere increase of these cells’ numbers. To accept this postulation, we investigated the abundances of Sertoli cell-specific FSHR and AMH, Leydig cell-specific LHR and INSL3, and spermatogonia-specific THY1 and CDH1 markers in immature and mature canine testis. Four biological replicates of immature and mature testes were processed, and RT-PCR was performed to elucidate the cells’ specific markers. The data were analyzed by ANOVA, using the 2−∆∆Ct method to ascertain differences in mRNA expressions. In addition, Western blot and IHC were performed. Gene expressions of all the studied cells’ specific markers were down-regulated (p < 0.05) in adult testis compared with immature testis. Western blot and immunohistochemistry showed the presence of these proteins in the testis. Protein expressions were greater in immature testis compared with mature testis (p < 0.05). Despite the obvious expansion of these cells’ numbers from immature to adult testis, the cells’ specific markers were not enriched in mature testis compared with immature dog testis. The results support the postulation that the gene expressions do not directly correlate with the increase of the cell numbers during post-natal development but changes in gene expressions show functional significance.
Collapse
|
8
|
Park JK, Song Y, Kim DW, Cho K, Yeo JM, Lee R, Lim YS, Lee WY, Park HJ. Helix-loop-helix protein ID4 expressed in bovine Sertoli cells. Acta Histochem 2021; 123:151800. [PMID: 34673438 DOI: 10.1016/j.acthis.2021.151800] [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: 07/14/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 10/20/2022]
Abstract
Stage- and cell type-specific biomarkers are important for understanding spermatogenesis in mammalian testis. The present study identified several testicular cell marker proteins in 6- and 24-month old bovine testes. In 6-month old bovine testes, spermatogonia and spermatocytes were detected but complete spermatogenesis occurred in 24-month old testes. The diameters of the seminiferous tubules increased significantly in the 24-month old testes compared with those in the 6-month old testes. Protein Gene Product 9.5 (PGP9.5), also known as the undifferentiated spermatogonium marker, and GATA4 (GATA binding protein 4), vimentin, and SOX9 (SRY-Box Transcription Factor 9) were detected in the basement membrane region. Interestingly, ID4 (inhibitor of DNA binding protein 4; previously known as the undifferentiated cell marker) proteins were located in the basement membrane region but their expression patterns were different from those of PGP9.5. Co-immunohistochemistry results showed that ID4 was detected in the Sertoli cells expressing vimentin and SOX9 in 6- and 24-month old bovine testes. This result indicated that ID4 is a putative biomarker of Sertoli cell in the bovine system, which is different from the rodent models. Thus, these results will contribute in understanding the process of spermatogenesis that is different in bovines compared to other species.
Collapse
|
9
|
Di Persio S, Tekath T, Siebert-Kuss LM, Cremers JF, Wistuba J, Li X, Meyer Zu Hörste G, Drexler HCA, Wyrwoll MJ, Tüttelmann F, Dugas M, Kliesch S, Schlatt S, Laurentino S, Neuhaus N. Single-cell RNA-seq unravels alterations of the human spermatogonial stem cell compartment in patients with impaired spermatogenesis. CELL REPORTS MEDICINE 2021; 2:100395. [PMID: 34622232 PMCID: PMC8484693 DOI: 10.1016/j.xcrm.2021.100395] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/01/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023]
Abstract
Despite the high incidence of male infertility, only 30% of infertile men receive a causative diagnosis. To explore the regulatory mechanisms governing human germ cell function in normal and impaired spermatogenesis (crypto), we performed single-cell RNA sequencing (>30,000 cells). We find major alterations in the crypto spermatogonial compartment with increased numbers of the most undifferentiated spermatogonia (PIWIL4+). We also observe a transcriptional switch within the spermatogonial compartment driven by increased and prolonged expression of the transcription factor EGR4. Intriguingly, the EGR4-regulated chromatin-associated transcriptional repressor UTF1 is downregulated at transcriptional and protein levels. This is associated with changes in spermatogonial chromatin structure and fewer Adark spermatogonia, characterized by tightly compacted chromatin and serving as reserve stem cells. These findings suggest that crypto patients are disadvantaged, as fewer cells safeguard their germline’s genetic integrity. These identified spermatogonial regulators will be highly interesting targets to uncover genetic causes of male infertility.
Crypto(zoospermic) men show increased number of PIWIL4+/EGR4+ spermatogonia Crypto undifferentiated spermatogonia over-activate the EGR4 regulatory network The predicted EGR4 target UTF1 is downregulated in crypto spermatogonia Crypto testes show reduced numbers of UTF1+ Adark reserve spermatogonia
Collapse
Affiliation(s)
- Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, 48149 Münster, Germany
| | - Tobias Tekath
- Institute of Medical Informatics, University Hospital of Münster, 48149 Münster, Germany
| | - Lara Marie Siebert-Kuss
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, 48149 Münster, Germany
| | - Jann-Frederik Cremers
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University Hospital of Münster, 48149 Münster, Germany
| | - Joachim Wistuba
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, 48149 Münster, Germany
| | - Xiaolin Li
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, 48149 Münster, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital of Münster, 48149 Münster, Germany
| | - Hannes C A Drexler
- Bioanalytical Mass Spectrometry Unit, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Margot Julia Wyrwoll
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University Hospital of Münster, 48149 Münster, Germany.,Institute of Reproductive Genetics, University of Münster, 48149 Münster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, 48149 Münster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University Hospital of Münster, 48149 Münster, Germany
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University Hospital of Münster, 48149 Münster, Germany
| | - Stefan Schlatt
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, 48149 Münster, Germany
| | - Sandra Laurentino
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, 48149 Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, 48149 Münster, Germany
| |
Collapse
|
10
|
Heckmann L, Langenstroth-Röwer D, Wistuba J, Portela JMD, van Pelt AMM, Redmann K, Stukenborg JB, Schlatt S, Neuhaus N. The initial maturation status of marmoset testicular tissues has an impact on germ cell maintenance and somatic cell response in tissue fragment culture. Mol Hum Reprod 2021; 26:374-388. [PMID: 32236422 DOI: 10.1093/molehr/gaaa024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 03/13/2020] [Indexed: 11/13/2022] Open
Abstract
Successful in vitro spermatogenesis was reported using immature mouse testicular tissues in a fragment culture approach, raising hopes that this method could also be applied for fertility preservation in humans. Although maintaining immature human testicular tissue fragments in culture is feasible for an extended period, it remains unknown whether germ cell survival and the somatic cell response depend on the differentiation status of tissue. Employing the marmoset monkey (Callithrix jacchus), we aimed to assess whether the maturation status of prepubertal and peri-/pubertal testicular tissues influence the outcome of testis fragment culture. Testicular tissue fragments from 4- and 8-month-old (n = 3, each) marmosets were cultured and evaluated after 0, 7, 14, 28 and 42 days. Immunohistochemistry was performed for identification and quantification of germ cells (melanoma-associated antigen 4) and Sertoli cell maturation status (anti-Müllerian hormone: AMH). During testis fragment culture, spermatogonial numbers were significantly reduced (P < 0.05) in the 4- but not 8-month-old monkeys, at Day 0 versus Day 42 of culture. Moreover, while Sertoli cells from 4-month-old monkeys maintained an immature phenotype (i.e. AMH expression) during culture, AMH expression was regained in two of the 8-month-old monkeys. Interestingly, progression of differentiation to later meiotic stage was solely observed in one 8-month-old marmoset, which was at an intermediate state regarding germ cell content, with gonocytes as well as spermatocytes present, as well as Sertoli cell maturation status. Although species-specific differences might influence the outcome of testis fragment experiments in vitro, our study demonstrated that the developmental status of the testicular tissues needs to be considered as it seems to be decisive for germ cell maintenance, somatic cell response and possibly the differentiation potential.
Collapse
Affiliation(s)
- L Heckmann
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - D Langenstroth-Röwer
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - J Wistuba
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - J M D Portela
- Center for Reproductive Medicine, Research Institute Reproduction and Development, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - A M M van Pelt
- Center for Reproductive Medicine, Research Institute Reproduction and Development, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - K Redmann
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - J B Stukenborg
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, 17164 Solna, Sweden
| | - S Schlatt
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - N Neuhaus
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| |
Collapse
|
11
|
The guardians of germ cells; Sertoli-derived exosomes against electromagnetic field-induced oxidative stress in mouse spermatogonial stem cells. Theriogenology 2021; 173:112-122. [PMID: 34371438 DOI: 10.1016/j.theriogenology.2021.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/02/2021] [Accepted: 08/01/2021] [Indexed: 01/10/2023]
Abstract
Nowadays, prolonged exposure to electromagnetic fields (EMF) has raised public concern about the detrimental potential of EMF on spermatogonial stem cells (SSCs) and spermatogenesis. Recent studies introduced the fundamental role of Sertoli cell paracrine signaling in the regulation of SSCs maintenance and differentiation in fertility preservation. Thus we investigated the therapeutic effect of Sertoli-derived exosomes (Sertoli-EXOs) as powerful paracrine mediators in SSCs subjected to EMF and its underlying mechanisms. SSCs and Sertoli cells were isolated from neonate mice testis, and identified by their specific markers. Then SSCs were exposed to 50 Hz EMF with intensity of 2.5 mT (1 h for 5 days) and supplemented with exosomes that were isolated from pre-pubertal Sertoli cells. Sertoli-EXOs were characterized and the uptake was observed by PKH26 labeling. The cell viability, colonization efficiency, reactive oxygen species (ROS) balance, cell cycle arrest and apoptosis induction were then analysed. SSCs were confirmed by immunocytochemistry (Oct4, Plzf) and Sertoli cells were identified through Sox9 and vimentin expression by immunocytochemistry and Real-time PCR (qRT-PCR), respectively. Our results demonstrated the detrimental effect of EMF via ROS accumulation that reduced the expression of catalase antioxidant, cell viability and colonization of SSCs. Also, AO/PI and flow cytometry analysis demonstrated the elevation of apoptosis in SSCs exposed to EMF in comparison with control. qRT-PCR data confirmed the up-regulation of apoptotic gene (Caspase-3) and down-regulation of SSCs specific gene (GFRα1). Consequently, the administration of Sertoli-EXOs exerted ameliorative effect on SSCs and significantly improved these changes through the regulation of oxidative stress. These findings suggest that Sertoli-EXOs have positive impact on SSCs exposed to EMF and can be useful in further investigation of Sertoli-EXOs as a novel therapeutic agent which may recover the deregulated SSCs microenvironment and spermatogenesis after exposure to EMF.
Collapse
|
12
|
Schneider F, Dabel J, Sandhowe-Klaverkamp R, Neuhaus N, Schlatt S, Kliesch S, Wistuba J. Serum and intratesticular inhibin B, AMH, and spermatogonial numbers in trans women at gender-confirming surgery: An observational study. Andrology 2021; 9:1781-1789. [PMID: 34085780 DOI: 10.1111/andr.13059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/30/2021] [Accepted: 05/23/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Anti-Müllerian hormone and inhibin B are produced by Sertoli cells. Anti-Müllerian hormone secretion indicates an immature Sertoli cell state. Inhibin B serves as a marker of male fertility. Identification of markers reflecting the presence of germ cells is of particular relevance in trans persons undergoing gender-affirming hormone therapy in order to offer individualized fertility preservation methods. OBJECTIVES Serum and intratesticular inhibin B and anti-Müllerian hormone values were assessed and related to clinical features, laboratory values, and germ cell numbers. MATERIALS AND METHODS Twenty-two trans women from three clinics were included. As gender-affirming hormone therapy, 10-12.5 mg of cyproterone acetate plus estrogens were administered. Height, weight, age, medication, and treatment duration were inquired by questionnaires. Serum luteinizing hormone, follicle-stimulating hormone, testosterone, and estradiol were measured by immuno-assays. Serum and intratesticular inhibin B and anti-Müllerian hormone were measured by commercially available ELISAs. Spermatogonia were quantified as spermatogonia per cubic millimeter testicular tissue applying a morphometric analysis of two independent testicular cross-sections per individual after MAGEA4 immunostaining. RESULTS Patients with high inhibin B levels presented with a higher number of spermatogonia (*p < 0.05). Furthermore, mean serum inhibin B was associated with low age (*p < 0.05), low follicle-stimulating hormone (*p < 0.05), and low testosterone (*p < 0.05). Serum anti-Müllerian hormone, however, was not related to spermatogonial numbers. It correlated with high testosterone (*p < 0.05) and high follicle-stimulating hormone (*p < 0.05) only. High intratesticular inhibin B was accompanied by high luteinizing hormone (*p < 0.05), high follicle-stimulating hormone (**p < 0.01), and high testosterone levels (**p < 0.01). Higher the intratesticular anti-Müllerian hormone levels, the longer gender-affirming hormone therapy was administered (*p < 0.05). DISCUSSION AND CONCLUSION Serum inhibin B levels indicate the presence of spermatogonia, whereas anti-Müllerian hormone seems not to be a reliable marker concerning germ cell abundance.
Collapse
Affiliation(s)
- Florian Schneider
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany.,Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Jennifer Dabel
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Reinhild Sandhowe-Klaverkamp
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Nina Neuhaus
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Stefan Schlatt
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Sabine Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Joachim Wistuba
- Institute of Reproductive and Regenerative Medicine, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
13
|
Zomer HD, Reddi PP. Characterization of rodent Sertoli cell primary cultures. Mol Reprod Dev 2020; 87:857-870. [PMID: 32743879 PMCID: PMC7685524 DOI: 10.1002/mrd.23402] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 07/16/2020] [Indexed: 12/25/2022]
Abstract
Sertoli cells play a vital role in spermatogenesis by offering physical and nutritional support to the differentiating male germ cells. They form the blood-testis barrier and secrete growth factors essential for germ cell differentiation. Sertoli cell primary cultures are critical for understanding the regulation of spermatogenesis; however, obtaining pure cultures has been a challenge. Rodent Sertoli cell isolation protocols do not rule out contamination by the interstitial or connective tissue cells. Sertoli cell-specific markers could be helpful, but there is no consensus. Vimentin, the most commonly used marker, is not specific for Sertoli cells since its expression has been reported in peritubular myoid cells, mesenchymal stem cells, fibroblasts, macrophages, and endothelial cells, which contaminate Sertoli cell preparations. Markers based on transcription and growth factors also have limitations. Thus, the impediment to obtaining pure Sertoli cell cultures pertains to both the method of isolation and marker usage. The aim of this review is to discuss improvements to current methods of rodent Sertoli cell primary cultures, assess the properties of prepubertal versus mature Sertoli cell cultures, and propose steps to improve cellular characterization. Potential benefits of using contemporary approaches, including lineage tracing, specific cell ablation, and RNA-seq for obtaining Sertoli-specific transcript markers are discussed. Evaluating the specificity and applicability of these markers at the protein level to characterize Sertoli cells in culture would be critical. This review is expected to positively impact future work using primary cultures of rodent Sertoli cells.
Collapse
Affiliation(s)
- Helena D Zomer
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana Champaign, Urbana, Illinois
| | - Prabhakara P Reddi
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana Champaign, Urbana, Illinois
| |
Collapse
|
14
|
Zomer HD, Reddi PP. Mouse Sertoli cells isolation by lineage tracing and sorting. Mol Reprod Dev 2020; 87:871-879. [PMID: 32735067 DOI: 10.1002/mrd.23406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 11/05/2022]
Abstract
Sertoli cells play a key role in spermatogenesis by supporting the germ cells throughout differentiation. The isolation of Sertoli cells is essential to study their functions. However, the close contact of Sertoli cells with other testicular cell types and the high proliferation of contaminating cells are obstacles to obtain pure primary cultures. Current rodent Sertoli cell isolation protocols result in enriched, rather than pure Sertoli cells. Therefore, novel approaches are necessary to improve the purity of Sertoli cell primary cultures. The goal of this study is to obtain pure mouse Sertoli cells using lineage tracing and fluorescence-activated cell sorting (FACS). We bred the Amh-Cre mouse line with tdTomato line to generate mice constitutively expressing red fluorescence specifically in Sertoli cells. Primary cultures of Sertoli cells isolated from prepubertal mice showed that 79% of cells expressed tdTomato, as evaluated by fluorescence microscopy and flow cytometry; however, nearly all adherent cells were positive for vimentin. Most of the tomato-negative cells expressed α-smooth muscle actin (α-SMA), a peritubular myoid cell marker, but double-negative populations were also present. These findings suggest that vimentin lacks Sertoli cell-specificity and that α-SMA is not adequate to identify all of the contaminating cells. Upon FACS sorting; however, virtually 100% of the cells were tdTomato positive, expressed vimentin, but not α-SMA. Prepubertal mice yielded a higher number of Sertoli cells compared to adults, but both could be adequately sorted. In conclusion, our study shows that lineage tracing and sorting is an efficient strategy for acquiring pure populations of murine Sertoli cells.
Collapse
Affiliation(s)
- Helena D Zomer
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Prabhakara P Reddi
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
15
|
Portela JMD, Heckmann L, Wistuba J, Sansone A, van Pelt AMM, Kliesch S, Schlatt S, Neuhaus N. Development and Disease-Dependent Dynamics of Spermatogonial Subpopulations in Human Testicular Tissues. J Clin Med 2020; 9:jcm9010224. [PMID: 31947706 PMCID: PMC7019285 DOI: 10.3390/jcm9010224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/03/2020] [Accepted: 01/10/2020] [Indexed: 12/22/2022] Open
Abstract
Cancer therapy and conditioning treatments of non-malignant diseases affect spermatogonial function and may lead to male infertility. Data on the molecular properties of spermatogonia and the influence of disease and/or treatment on spermatogonial subpopulations remain limited. Here, we assessed if the density and percentage of spermatogonial subpopulation changes during development (n = 13) and due to disease and/or treatment (n = 18) in tissues stored in fertility preservation programs, using markers for spermatogonia (MAGEA4), undifferentiated spermatogonia (UTF1), proliferation (PCNA), and global DNA methylation (5mC). Throughout normal prepubertal testicular development, only the density of 5mC-positive spermatogonia significantly increased with age. In comparison, patients affected by disease and/or treatment showed a reduced density of UTF1-, PCNA- and 5mC-positive spermatogonia, whereas the percentage of spermatogonial subpopulations remained unchanged. As an exception, sickle cell disease patients treated with hydroxyurea displayed a reduction in both density and percentage of 5mC- positive spermatogonia. Our results demonstrate that, in general, a reduction in spermatogonial density does not alter the percentages of undifferentiated and proliferating spermatogonia, nor the establishment of global methylation. However, in sickle cell disease patients’, establishment of spermatogonial DNA methylation is impaired, which may be of importance for the potential use of this tissues in fertility preservation programs.
Collapse
Affiliation(s)
- Joana M. D. Portela
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany; (J.M.D.P.); (L.H.); (J.W.); (A.S.); (S.S.)
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Laura Heckmann
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany; (J.M.D.P.); (L.H.); (J.W.); (A.S.); (S.S.)
| | - Joachim Wistuba
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany; (J.M.D.P.); (L.H.); (J.W.); (A.S.); (S.S.)
| | - Andrea Sansone
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany; (J.M.D.P.); (L.H.); (J.W.); (A.S.); (S.S.)
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Ans M. M. van Pelt
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Sabine Kliesch
- Center of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany;
| | - Stefan Schlatt
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany; (J.M.D.P.); (L.H.); (J.W.); (A.S.); (S.S.)
| | - Nina Neuhaus
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany; (J.M.D.P.); (L.H.); (J.W.); (A.S.); (S.S.)
- Correspondence:
| |
Collapse
|
16
|
Park HJ, Song H, Woo JS, Chung HJ, Park JK, Cho KH, Mo Yeo J, Lee WY. Expression patterns of male germ cell markers in cryptorchid pig testes. Acta Histochem 2019; 121:784-790. [PMID: 31324385 DOI: 10.1016/j.acthis.2019.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/27/2019] [Accepted: 06/27/2019] [Indexed: 11/29/2022]
Abstract
Male germ cell apoptosis has been described in heat-damaged testes by cryptorchidism. In the present study, wild type pig testes were compared with cryptorchid testes via histological and immunohistological analyses. Spermatozoa were not detected in two cryptorchid testes and the diameters of seminiferous tubules were significantly reduced in cryptorchid pig testes compared with wild type pig testes. Cells expressing marker genes for undifferentiated spermatogonia, such as protein gene product 9.5 was significantly decreased in cryptochid pig testes. In addition, the numbers of cells expressing DEAD-box polypeptide 4 (VASA), synaptonemal complex protein 3, protamine, and acrosin (a biomarker of spermatocyte, spermatid, and spermatozoa) were significantly reduced in cryptochid pig testes. However, the number of vimentin-expressing Sertoli cells was not changed or was significantly increased in cryptorchid pig testes. This result indicates that male germ cells are specifically damaged by heat in cryptorchid pig testes and not Sertoli cells. These findings will facilitate the further study of spermatogenesis and the specific mechanisms by which cryptorchidism causes male infertility.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Stem Cell and Regenerative Biology, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jae-Seok Woo
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeonbuk 55365, Republic of Korea
| | - Hak-Jae Chung
- Swine Science Division, National Institute of Animal Science, RDA, Cheoan-si 31000, Republic of Korea
| | - Jin-Ki Park
- Department of Swine & Poultry Science, Korea National College of Agriculture and Fisheries, Jeonbuk 54874, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Beef & Dairy Science, Korea National College of Agricultures and Fisheries, Jeonbuk 54874, Republic of Korea
| | - Joon Mo Yeo
- Department of Beef & Dairy Science, Korea National College of Agricultures and Fisheries, Jeonbuk 54874, Republic of Korea
| | - Won-Young Lee
- Department of Beef & Dairy Science, Korea National College of Agricultures and Fisheries, Jeonbuk 54874, Republic of Korea.
| |
Collapse
|
17
|
Laurentino S, Heckmann L, Di Persio S, Li X, Meyer Zu Hörste G, Wistuba J, Cremers JF, Gromoll J, Kliesch S, Schlatt S, Neuhaus N. High-resolution analysis of germ cells from men with sex chromosomal aneuploidies reveals normal transcriptome but impaired imprinting. Clin Epigenetics 2019; 11:127. [PMID: 31462300 PMCID: PMC6714305 DOI: 10.1186/s13148-019-0720-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/02/2019] [Indexed: 12/18/2022] Open
Abstract
Background The most common sex chromosomal aneuploidy in males is Klinefelter syndrome, which is characterized by at least one supernumerary X chromosome. While these men have long been considered infertile, focal spermatogenesis can be observed in some patients, and sperm can be surgically retrieved and used for artificial reproductive techniques. Although these gametes can be used for fertility treatments, little is known about the molecular biology of the germline in Klinefelter men. Specifically, it is unclear if germ cells in Klinefelter syndrome correctly establish the androgenetic DNA methylation profile and transcriptome. This is due to the low number of germ cells in the Klinefelter testes available for analysis. Results Here, we overcame these difficulties and successfully investigated the epigenetic and transcriptional profiles of germ cells in Klinefelter patients employing deep bisulfite sequencing and single-cell RNA sequencing. On the transcriptional level, the germ cells from Klinefelter men clustered together with the differentiation stages of normal spermatogenesis. Klinefelter germ cells showed a normal DNA methylation profile of selected germ cell-specific markers compared with spermatogonia and sperm from men with normal spermatogenesis. However, germ cells from Klinefelter patients showed variations in the DNA methylation of imprinted regions. Conclusions These data indicate that Klinefelter germ cells have a normal transcriptome but might present aberrant imprinting, showing impairment in germ cell development that goes beyond mere germ cell loss. Electronic supplementary material The online version of this article (10.1186/s13148-019-0720-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sandra Laurentino
- Centre of Reproductive Medicine and Andrology, University of Münster, Domagkstrasse 11, 48149, Münster, Germany
| | - Laura Heckmann
- Centre of Reproductive Medicine and Andrology, University of Münster, Domagkstrasse 11, 48149, Münster, Germany
| | - Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University of Münster, Domagkstrasse 11, 48149, Münster, Germany
| | - Xiaolin Li
- Department of Neurology, Institute of Translational Neurology, University Hospital of Münster, Münster, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology, Institute of Translational Neurology, University Hospital of Münster, Münster, Germany
| | - Joachim Wistuba
- Centre of Reproductive Medicine and Andrology, University of Münster, Domagkstrasse 11, 48149, Münster, Germany
| | - Jann-Frederik Cremers
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University of Münster, Münster, Germany
| | - Jörg Gromoll
- Centre of Reproductive Medicine and Andrology, University of Münster, Domagkstrasse 11, 48149, Münster, Germany
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University of Münster, Münster, Germany
| | - Stefan Schlatt
- Centre of Reproductive Medicine and Andrology, University of Münster, Domagkstrasse 11, 48149, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, University of Münster, Domagkstrasse 11, 48149, Münster, Germany.
| |
Collapse
|
18
|
Pohl E, Höffken V, Schlatt S, Kliesch S, Gromoll J, Wistuba J. Ageing in men with normal spermatogenesis alters spermatogonial dynamics and nuclear morphology in Sertoli cells. Andrology 2019; 7:827-839. [PMID: 31250567 DOI: 10.1111/andr.12665] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/28/2019] [Accepted: 05/14/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Ageing in men is believed to be associated with fertility decline and elevated risk of congenital disorders for the offspring. The previous studies also reported reduced germ and Sertoli cell numbers in older men. However, it is not clear whether ageing in men with normal spermatogenesis affects the testis and germ cell population dynamics in a way sufficient for transmitting adverse age effects to the offspring. OBJECTIVES We examined men with normal spermatogenesis at different ages concerning effects on persisting testicular cell types, that is the germ line and Sertoli cells, as these cell populations are prone to be exposed to age effects. MATERIAL AND METHODS Ageing was assessed in testicular biopsies of 32 patients assigned to three age groups: (i) 28.8 ± 2.7 years; (ii) 48.1 ± 1 years; and (iii) 70.9 ± 6.2 years, n = 8 each, with normal spermatogenesis according to the Bergmann-Kliesch score, and in a group of meiotic arrest patients (29.9 ± 3.8 years, n = 8) to decipher potential links between different germ cell types. Besides morphometry of seminiferous tubules and Sertoli cell nuclei, we investigated spermatogenic output/efficiency, and dynamics of spermatogonial populations via immunohistochemistry for MAGE A4, PCNA, CREM and quantified A-pale/A-dark spermatogonia. RESULTS We found a constant spermatogenic output (CREM-positive round spermatids) in all age groups studied. In men beyond their mid-40s (group 2), we detected increased nuclear and nucleolar size in Sertoli cells, indirectly indicating an elevated protein turnover. From the 7th decade (group 3) of life onwards, testes showed increased proliferation of undifferentiated spermatogonia, decreased spermatogenic efficiency and elevated numbers of proliferating A-dark spermatogonia. DISCUSSION AND CONCLUSION Maintaining normal sperm output seems to be an intrinsic determinant of spermatogenesis. Ageing appears to affect this output and might provoke compensatory proliferation increase in A spermatogonia which, in turn, might hamper germ cell integrity.
Collapse
Affiliation(s)
- E Pohl
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - V Höffken
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - S Schlatt
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - S Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - J Gromoll
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - J Wistuba
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| |
Collapse
|
19
|
Efficiency of colony formation and differentiation of human spermatogenic cells in two different culture systems. Reprod Biol 2018; 18:397-403. [DOI: 10.1016/j.repbio.2018.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/08/2018] [Accepted: 09/22/2018] [Indexed: 01/15/2023]
|
20
|
Heckmann L, Langenstroth-Röwer D, Pock T, Wistuba J, Stukenborg JB, Zitzmann M, Kliesch S, Schlatt S, Neuhaus N. A diagnostic germ cell score for immature testicular tissue at risk of germ cell loss. Hum Reprod 2018; 33:636-645. [DOI: 10.1093/humrep/dey025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 01/24/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- L Heckmann
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - D Langenstroth-Röwer
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - T Pock
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - J Wistuba
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - J -B Stukenborg
- Department of Women's and Children's Health, NORDFERTIL Research Lab Stockholm, Paediatric Endocrinology Unit, Q2:08, Karolinska Institutet and Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - M Zitzmann
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - S Kliesch
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - S Schlatt
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - N Neuhaus
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| |
Collapse
|
21
|
Neuhaus N, Yoon J, Terwort N, Kliesch S, Seggewiss J, Huge A, Voss R, Schlatt S, Grindberg RV, Schöler HR. Single-cell gene expression analysis reveals diversity among human spermatogonia. Mol Hum Reprod 2018; 23:79-90. [PMID: 28093458 DOI: 10.1093/molehr/gaw079] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 01/12/2017] [Indexed: 12/16/2022] Open
Abstract
STUDY QUESTION Is the molecular profile of human spermatogonia homogeneous or heterogeneous when analysed at the single-cell level? SUMMARY ANSWER Heterogeneous expression profiles may be a key characteristic of human spermatogonia, supporting the existence of a heterogeneous stem cell population. WHAT IS KNOWN ALREADY Despite the fact that many studies have sought to identify specific markers for human spermatogonia, the molecular fingerprint of these cells remains hitherto unknown. STUDY DESIGN, SIZE, DURATION Testicular tissues from patients with spermatogonial arrest (arrest, n = 1) and with qualitatively normal spermatogenesis (normal, n = 7) were selected from a pool of 179 consecutively obtained biopsies. Gene expression analyses of cell populations and single-cells (n = 105) were performed. Two OCT4-positive individual cells were selected for global transcriptional capture using shallow RNA-seq. Finally, expression of four candidate markers was assessed by immunohistochemistry. PARTICIPANTS/MATERIALS, SETTING, METHODS Histological analysis and blood hormone measurements for LH, FSH and testosterone were performed prior to testicular sample selection. Following enzymatic digestion of testicular tissues, differential plating and subsequent micromanipulation of individual cells was employed to enrich and isolate human spermatogonia, respectively. Endpoint analyses were qPCR analysis of cell populations and individual cells, shallow RNA-seq and immunohistochemical analyses. MAIN RESULTS AND THE ROLE OF CHANCE Unexpectedly, single-cell expression data from the arrest patient (20 cells) showed heterogeneous expression profiles. Also, from patients with normal spermatogenesis, heterogeneous expression patterns of undifferentiated (OCT4, UTF1 and MAGE A4) and differentiated marker genes (BOLL and PRM2) were obtained within each spermatogonia cluster (13 clusters with 85 cells). Shallow RNA-seq analysis of individual human spermatogonia was validated, and a spermatogonia-specific heterogeneous protein expression of selected candidate markers (DDX5, TSPY1, EEF1A1 and NGN3) was demonstrated. LIMITATIONS, REASONS FOR CAUTION The heterogeneity of human spermatogonia at the RNA and protein levels is a snapshot. To further assess the functional meaning of this heterogeneity and the dynamics of stem cell populations, approaches need to be developed to facilitate the repeated analysis of individual cells. WIDER IMPLICATIONS OF THE FINDINGS Our data suggest that heterogeneous expression profiles may be a key characteristic of human spermatogonia, supporting the model of a heterogeneous stem cell population. Future studies will assess the dynamics of spermatogonial populations in fertile and infertile patients. LARGE SCALE DATA RNA-seq data is published in the GEO database: GSE91063. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Max Planck Society and the Deutsche Forschungsgemeinschaft DFG-Research Unit FOR 1041 Germ Cell Potential (grant numbers SCHO 340/7-1, SCHL394/11-2). The authors declare that there is no conflict of interest.
Collapse
Affiliation(s)
- N Neuhaus
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Domagkstrasse 11, 48149 Münster , Germany
| | - J Yoon
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149 Münster , Germany
| | - N Terwort
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Domagkstrasse 11, 48149 Münster , Germany
| | - S Kliesch
- Department of Clinical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Domagkstrasse 11, 48149 Münster , Germany
| | - J Seggewiss
- Institute of Human Genetics, University Hospital Münster, Vesaliusweg 12-14, 48149 Münster , Germany
| | - A Huge
- Core Facility Genomik, Medical Faculty of Münster, Domagkstrasse 3, 48149 Münster , Germany
| | - R Voss
- Interdisciplinary Centre for Clinical Research in the Faculty of Medicine, Domagkstrasse 3, 48149 Münster , Germany
| | - S Schlatt
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Domagkstrasse 11, 48149 Münster , Germany
| | - R V Grindberg
- University Hospital Zurich, Department of Infectious Diseases and Hospital Epidemiology, 8091 Zurich , Switzerland
| | - H R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149 Münster , Germany
| |
Collapse
|
22
|
Gat I, Maghen L, Filice M, Kenigsberg S, Wyse B, Zohni K, Saraz P, Fisher AG, Librach C. Initial germ cell to somatic cell ratio impacts the efficiency of SSC expansion in vitro. Syst Biol Reprod Med 2018; 64:39-50. [PMID: 29193985 DOI: 10.1080/19396368.2017.1406013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 10/12/2017] [Indexed: 12/23/2022]
Abstract
Spermatogonial Stem Cell (SSC) expansion in vitro remains a major challenge in efforts to preserve fertility among pubertal cancer survivor boys. The current study focused on innovative approaches to optimize SSC expansion. Six- to eight-week-old CD-1 murine testicular samples were harvested by mechanical and enzymatic digestion. Cell suspensions were incubated for differential plating (DP). After DP, we established two experiments comparing single vs. repetitive DP (S-DP and R-DP, respectively) until passage 2 (P2) completion. Each experiment included a set of cultures consisting of 5 floating-to-attached cell ratios (5, 10, 15, 20, and 25) and control cultures containing floating cells only. We found similar cell and colony count drops during P0 in both S- and R-DP. During P2, counts increased in S-DP in middle ratios (10, 15, and especially 20) relative to low and high ratios (5 and 25, respectively). Counts dropped extensively in R-DP after passage 2. The superiority of intermediate ratios was demonstrated by enrichment of GFRα1 by qPCR. The optimal ratio of 20 in S-DP contained significantly increased proportions of GFRα1-positive cells (25.8±5.8%) as measured by flow cytometry compared to after DP (1.9±0.7%, p<0.0001), as well as positive immunostaining for GFRα1 and UTF1, with rare Sox9-positive cells. This is the first report of the impact of initial floating-to-attached cell ratios on SSC proliferation in vitro. ABBREVIATIONS SSC: spermatogonial stem cells; DP: differential plating; NOA: non-obstructive azoospermia; MACS: magnetic-activated cells sorting; FACS: fluorescence-activated cells sorting.
Collapse
Affiliation(s)
- Itai Gat
- a CReATe Fertility Centre , Toronto , Ontario , Canada
- b Pinchas Borenstein Talpiot Medical Leadership Program , Sheba Medical Center, Tel HaShomer , Ramat Gan , Israel
- c Sackler Medical School, University of Tel Aviv , Israel
| | - Leila Maghen
- a CReATe Fertility Centre , Toronto , Ontario , Canada
| | | | | | - Brandon Wyse
- a CReATe Fertility Centre , Toronto , Ontario , Canada
| | - Khaled Zohni
- a CReATe Fertility Centre , Toronto , Ontario , Canada
| | - Peter Saraz
- a CReATe Fertility Centre , Toronto , Ontario , Canada
| | | | - Clifford Librach
- a CReATe Fertility Centre , Toronto , Ontario , Canada
- d Department of Obstetrics & Gynecology , University of Toronto , Toronto , Ontario , Canada
- e Department of Gynecology , Women's College Hospital , Toronto , Ontario , Canada
| |
Collapse
|
23
|
Sharma S, Portela JMD, Langenstroth-Röwer D, Wistuba J, Neuhaus N, Schlatt S. Male germline stem cells in non-human primates. Primate Biol 2017; 4:173-184. [PMID: 32110705 PMCID: PMC7041516 DOI: 10.5194/pb-4-173-2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/17/2017] [Indexed: 12/22/2022] Open
Abstract
Over the past few decades, several studies have attempted to decipher the
biology of mammalian germline stem cells (GSCs). These studies provide
evidence that regulatory mechanisms for germ cell specification and migration
are evolutionarily conserved across species. The characteristics and
functions of primate GSCs are highly distinct from rodent species; therefore
the findings from rodent models cannot be extrapolated to primates. Due to
limited availability of human embryonic and testicular samples for research
purposes, two non-human primate models (marmoset and macaque monkeys) are
extensively employed to understand human germline development and
differentiation. This review provides a broader introduction to the in vivo
and in vitro germline stem cell terminology from primordial to
differentiating germ cells. Primordial germ cells (PGCs) are the most
immature germ cells colonizing the gonad prior to sex differentiation into
testes or ovaries. PGC specification and migratory patterns among different
primate species are compared in the review. It also reports the distinctions
and similarities in expression patterns of pluripotency markers (OCT4A,
NANOG, SALL4 and LIN28) during embryonic developmental stages, among
marmosets, macaques and humans. This review presents a comparative summary
with immunohistochemical and molecular evidence of germ cell marker
expression patterns during postnatal developmental stages, among humans and
non-human primates. Furthermore, it reports findings from the recent
literature investigating the plasticity behavior of germ cells and stem cells
in other organs of humans and monkeys. The use of non-human primate models
would enable bridging the knowledge gap in primate GSC research and
understanding the mechanisms involved in germline development. Reported
similarities in regulatory mechanisms and germ cell expression profile in
primates demonstrate the preclinical significance of monkey models for
development of human fertility preservation strategies.
Collapse
Affiliation(s)
- Swati Sharma
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Medicine, Albert Schweitzer Campus 1, Building D11, Münster, Germany.,These authors contributed equally to this work
| | - Joana M D Portela
- Center for Reproductive Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands.,These authors contributed equally to this work
| | - Daniel Langenstroth-Röwer
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Medicine, Albert Schweitzer Campus 1, Building D11, Münster, Germany
| | - Joachim Wistuba
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Medicine, Albert Schweitzer Campus 1, Building D11, Münster, Germany
| | - Nina Neuhaus
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Medicine, Albert Schweitzer Campus 1, Building D11, Münster, Germany
| | - Stefan Schlatt
- Center of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Medicine, Albert Schweitzer Campus 1, Building D11, Münster, Germany
| |
Collapse
|
24
|
Gat I, Maghen L, Filice M, Wyse B, Zohni K, Jarvi K, Lo KC, Gauthier Fisher A, Librach C. Optimal culture conditions are critical for efficient expansion of human testicular somatic and germ cells in vitro. Fertil Steril 2017; 107:595-605.e7. [PMID: 28259258 DOI: 10.1016/j.fertnstert.2016.12.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To optimize culture conditions for human testicular somatic cells (TSCs) and spermatogonial stem cells. DESIGN Basic science study. SETTING Urology clinic and stem cell research laboratory. PATIENT(S) Eight human testicular samples. INTERVENTIONS(S) Testicular tissues were processed by mechanical and enzymatic digestion. Cell suspensions were subjected to differential plating (DP) after which floating cells (representing germ cells) were removed and attached cells (representing TSCs) were cultured for 2 passages (P0-P1) in StemPro-34- or DMEM-F12-based medium. Germ cell cultures were established in both media for 12 days. MAIN OUTCOME MEASURE(S) TSC cultures: proliferation doubling time (PDT), fluorescence-activated cell sorting for CD90, next-generation sequencing for 89 RNA transcripts, immunocytochemistry for TSC and germ cell markers, and conditioned media analysis; germ cell cultures: number of aggregates. RESULT(S) TSCs had significantly prolonged PDT in DMEM-F12 versus StemPro-34 (319.6 ± 275.8 h and 110.5 ± 68.3 h, respectively). The proportion of CD90-positive cells increased after P1 in StemPro-34 and DMEM-F12 (90.1 ± 10.8% and 76.5 ± 17.4%, respectively) versus after DP (66.3 ± 7%). Samples from both media after P1 clustered closely in the principle components analysis map whereas those after DP did not. After P1 in either medium, CD90-positive cells expressed TSC markers only, and fibroblast growth factor 2 and bone morphogenetic protein 4 were detected in conditioned medium. A higher number of germ cell aggregates formed in DMEM-F12 (59 ± 39 vs. 28 ± 17, respectively). CONCLUSION(S) Use of DMEM-F12 reduces TSC proliferation while preserving their unique characteristics, leading to improved germ cell aggregates formation compared with StemPro-34, the standard basal medium used in the majority of previous reports.
Collapse
Affiliation(s)
- Itai Gat
- Create Fertility Centre, Toronto, Ontario, Canada; Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center, Ramat Gan, Israel; Sackler school of medicine, Tel Aviv university, Tel Aviv, Israel
| | - Leila Maghen
- Create Fertility Centre, Toronto, Ontario, Canada
| | | | - Brandon Wyse
- Create Fertility Centre, Toronto, Ontario, Canada
| | - Khaled Zohni
- Create Fertility Centre, Toronto, Ontario, Canada; Department of Reproductive Health and Family Planning, National Research Center, Cairo, Egypt
| | - Keith Jarvi
- Division of Urology, Department of Surgery, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Kirk C Lo
- Division of Urology, Department of Surgery, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | - Clifford Librach
- Create Fertility Centre, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, Women's College Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
25
|
Ahmed N, Yufei H, Yang P, Muhammad Yasir W, Zhang Q, Liu T, Hong C, Lisi H, Xiaoya C, Chen Q. Cytological study on Sertoli cells and their interactions with germ cells during annual reproductive cycle in turtle. Ecol Evol 2016; 6:4050-64. [PMID: 27516863 PMCID: PMC4972231 DOI: 10.1002/ece3.2193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 04/07/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022] Open
Abstract
Sertoli cells (SCs) play a central role in the development of germ cells within functional testes and exhibit varying morphology during spermatogenesis. This present study investigated the seasonal morphological changes in SCs in the reproductive cycle of Pelodiscus sinensis by light microscopy, transmission electron microscopy (TEM), and immunohistochemistry. During hibernation period with the quiescent of spermatogenesis, several autophagosomes were observed inside the SCs, the processes of which retracted. In early spermatogenesis, when the germ cells started to proliferate, the SCs contained numerous lipid droplets instead of autophagosomes. In late spermatogenesis, the SCs processes became very thin and contacted several round/elongated spermatids in pockets. At this time, abundant endoplasmic reticulum and numerous mitochondria were present in the SCs. The organization of the tight junctions and the adherens junctions between the SCs and germ cells also changed during the reproductive cycle. Moreover, SCs were involved in the formation of cytoplasmic bridges, phagophores, and exosome secretions during spermatogenesis. Tubulobulbar complexes (TBC) were also developed by SCs around the nucleus of the spermatid at the time of spermiation. Strong, positive expression of vimentin was noted on the SCs during late spermatogenesis compared with the hibernation stage and the early stage of spermatogenesis. These data provide clear cytological evidence about the seasonal changes in SCs, corresponding with their different roles in germ cells within the Chinese soft‐shelled turtle Pelodiscus sinensis.
Collapse
Affiliation(s)
- Nisar Ahmed
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China; Faculty of Veterinary and Animal Sciences LUAWMS Uthal 90150 Pakistan
| | - Huang Yufei
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| | - Ping Yang
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| | - Waqas Muhammad Yasir
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| | - Qian Zhang
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| | - Tengfei Liu
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| | - Chen Hong
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| | - Hu Lisi
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| | - Chu Xiaoya
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| | - Qiusheng Chen
- Laboratory of Animal Cell Biology and Embryology College of Veterinary Medicine Nanjing Agricultural University Nanjing 210095 China
| |
Collapse
|
26
|
Pohl E, Gromoll J, Kliesch S, Wistuba J. An alternative interpretation of cellular 'selfish spermatogonial selection'-clusters in the human testis indicates the need for 3-D-analyses. Andrology 2016; 4:213-7. [PMID: 26891892 DOI: 10.1111/andr.12142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/30/2015] [Accepted: 11/09/2015] [Indexed: 02/01/2023]
Abstract
The 'selfish spermatogonial selection'- model was proposed to explain the paternal age effect (PAE) of some congenital disorders associated with point mutations in male germ cells. According to this, spermatogonia carrying pathogenic mutations gain a selection advantage over non-mutated spermatogonia which leads to an increased number of mutated spermatogonia and consequently spermatozoa over time. Recently, an immunohistochemical approach using the premeiotic marker melanoma antigen family A4 (MAGE A4) was undertaken by the Wilkie group to confirm the presence of microclones of putatively mutated spermatogonia in testes of elderly men. The objective of our study was the age-dependent assessment of testes from men with normal spermatogenesis using MAGE A4 immunohistochemistry to identify and corroborate cellular clusters indicative for 'selfish spermatogonial selection' in our cohort. We analyzed testicular tissues obtained from men with normal spermatogenesis assigned to three age groups [(1) 28.8 ± 2.7 years; (2) 48.1 ± 1 years; (3) 71.9 ± 6.8 years, n/group = 8]. We could detect very similar distribution patterns of MAGE A4-positive cells and the presence of several types of microclusters as reported previously. However, these cellular clusters, indicative for clonal expansion, were not only present in testes from elderly men but also in those from age group 1 and 2. Using graphical three-dimensional modelling, we identified that cross-section directions e.g. longitudinal sections might provoke misleading interpretation of spermatogonial clusters, in particular when the tissue processing is limited. Thus, appropriate fixation and embedding is needed for reliable analysis of testicular sections. We therefore propose a more careful interpretation of such spermatogonial clusters and recommend a 3-D analysis to unequivocally determine 'selfish spermatogonial selection'-manifestations.
Collapse
Affiliation(s)
- E Pohl
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Germany
| | - J Gromoll
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Germany
| | - S Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Germany
| | - J Wistuba
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Germany
| |
Collapse
|
27
|
Lin ZYC, Hikabe O, Suzuki S, Hirano T, Siomi H, Sasaki E, Imamura M, Okano H. Sphere-formation culture of testicular germ cells in the common marmoset, a small New World monkey. Primates 2015; 57:129-35. [PMID: 26530217 DOI: 10.1007/s10329-015-0500-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/21/2015] [Indexed: 01/15/2023]
Abstract
Spermatogonia are specialized cells responsible for continuous spermatogenesis and the production of offspring. Because of this biological property, in vitro culture of spermatogonia provides a powerful methodology to advance reproductive biology and engineering. However, methods for culturing primate spermatogonia are poorly established. We have designed a novel method for culturing spermatogonia in the common marmoset (Callithrix jacchus), a small primate. By using our method with a suite of growth factors, adult marmoset testis-derived germ cells could be cultured in the form of a floating sphere for several weeks. Notably, this method could be applied not only to freshly isolated cells but also to cryopreserved cell stocks. The spheres enriched spermatogonia and early spermatocytes, and could be assembled from a C-KIT(+) spermatogonial population. Techniques for culturing spermatogonia could facilitate increased understanding of primate reproduction as well as the preservation of valuable biomaterials from nonhuman primates.
Collapse
Affiliation(s)
- Zachary Yu-Ching Lin
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Orie Hikabe
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Sadafumi Suzuki
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takamasa Hirano
- Department of Molecular Biology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Haruhiko Siomi
- Department of Molecular Biology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Erika Sasaki
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Applied Developmental Biology, Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki, 210-0821, Japan.,PRESTO Japan Science and Technology Agency, Tokyo, Japan
| | - Masanori Imamura
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan. .,Molecular Biology Section, Department of Cellular and Molecular Biology, Primate Research Institute, Kyoto University, Inuyama, Aichi, 484-8506, Japan.
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
28
|
Schneider F, Redmann K, Wistuba J, Schlatt S, Kliesch S, Neuhaus N. Comparison of enzymatic digestion and mechanical dissociation of human testicular tissues. Fertil Steril 2015; 104:302-11.e3. [PMID: 26056924 DOI: 10.1016/j.fertnstert.2015.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 04/10/2015] [Accepted: 05/01/2015] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To compare mechanical dissociation, employing the Medimachine system, and enzymatic digestion of human testicular tissues with respect to the proportion of spermatogonia and somatic cells, with the long-term objective of establishing human spermatogonial cultures. DESIGN Experimental basic science study. SETTING Reproductive biology laboratory. PATIENT(S) Testicular tissues were obtained from patients with gender dysphoria on the day of sex reassignment surgery. On the basis of the histological evaluation, tissue samples with complete spermatogenesis (fresh, n = 6; cryopreserved, n = 7) and with meiotic arrest (cryopreserved, n = 4) were selected. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) The composition of testicular cell suspensions was assessed performing quantitative real-time polymerase chain reaction (qPCR) analyses for germ cell-specific (FGFR3, SALL4, UTF1, MAGE-A4) and somatic marker genes (ACTA2 and VIM). Additionally, flow-cytometric analyses were used to evaluate the percentage of SALL4-and vimentin-positive cells. RESULT(S) While Medimachine dissociation yielded higher cell numbers in all patient groups, viability of cells was highly variable and correlated with the histological status of the tissue. Interestingly, qPCR analysis revealed a significantly decreased expression of the somatic marker genes ACTA2 and VIM and an increased expression of the spermatogonial marker genes FGFR3 and SALL4 after Medimachine dissociation. These findings were corroborated by flow-cytometric analyses that demonstrated that the proportion of SALL4-positive cells was up to 4 times higher after mechanical dissociation. CONCLUSION(S) Medimachine dissociation of human testicular tissues is comparably fast and leads to an enrichment of SALL4-positive spermatogonia. The use of this method may therefore constitute an advantage for the establishment of human spermatogonial cell cultures.
Collapse
Affiliation(s)
- Florian Schneider
- Institute for Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus, Münster, Germany; Department of Clinical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus, Münster, Germany
| | - Klaus Redmann
- Institute for Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus, Münster, Germany
| | - Joachim Wistuba
- Institute for Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus, Münster, Germany
| | - Stefan Schlatt
- Institute for Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus, Münster, Germany
| | - Sabine Kliesch
- Department of Clinical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus, Münster, Germany
| | - Nina Neuhaus
- Institute for Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus, Münster, Germany.
| |
Collapse
|
29
|
Pimenta MT, Francisco RAR, Silva RP, Porto CS, Lazari MFM. Relaxin affects cell organization and early and late stages of spermatogenesis in a coculture of rat testicular cells. Andrology 2015; 3:772-86. [PMID: 26041439 DOI: 10.1111/andr.12056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/15/2015] [Accepted: 04/30/2015] [Indexed: 01/03/2023]
Abstract
Relaxin and its receptor RXFP1 are co-expressed in Sertoli cells, and relaxin can stimulate proliferation of Sertoli cells. In this study, we investigated a role of relaxin in spermatogenesis, using a short-term culture of testicular cells of the rat that allowed differentiation of spermatogonia to spermatids. Sertoli, germ, and peritubular myoid cells were the predominant cell types in the culture. Sertoli and germ cells expressed RXFP1. Cultures were incubated without (control) or with 0.5% fetal bovine serum (FBS) or 100 ng/mL H2 relaxin (RLN) for 2 days. Cell organization, number, and differentiation were analyzed after 2 (D2), 5 (D5) or 8 (D8) days of culturing. Although the proportion of germ cells decayed from D2 to D5, the relative contribution of HC, 1C, 2C, and 4C germ cell populations remained constant in the control group during the whole culture. RLN did not affect the proportion of germ cell populations compared with control, but increased gene and/or protein expression of the undifferentiated and differentiated spermatogonia markers PLZF and c-KIT, and of the post-meiotic marker Odf2 in D5. RLN favored organization of cells in tubule-like structures, the arrangement of myoid cells around the tubules, arrangement of c-KIT-positive spermatogonia at the basal region of the tubules, and expression of the cell junction protein β-catenin close to the plasma membrane region. Knockdown of relaxin with small interfering RNA (siRNA) reduced expression of β-catenin at the cell junctions, and shifted its expression to the nucleus. We propose that relaxin may affect spermatogenesis by modulating spermatogonial self renewal and favoring cell contact.
Collapse
Affiliation(s)
- M T Pimenta
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - R A R Francisco
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - R P Silva
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - C S Porto
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - M F M Lazari
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| |
Collapse
|
30
|
Westernströer B, Langenstroth D, Kliesch S, Troppmann B, Redmann K, Macdonald J, Mitchell R, Wistuba J, Schlatt S, Neuhaus N. Developmental expression patterns of chemokines CXCL11, CXCL12 and their receptor CXCR7 in testes of common marmoset and human. Cell Tissue Res 2015; 361:885-98. [PMID: 25810367 DOI: 10.1007/s00441-015-2164-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 02/24/2015] [Indexed: 01/07/2023]
Abstract
The chemokine receptor CXCR7 interacts with the chemokines CXCL11 and CXCL12. During development, this ligand receptor system (C-X-C) provokes cell-type-specific responses in terms of migration, adhesion or ligand sequestration. It is active in zebrafish and rodents but no data are available for its presence or function in primate testes. Real-time quantitative polymerase chain reaction was performed in monkeys to detect CXCL11, CXCL12 and CXCR7. At the protein level, CXCL12 and CXCR7 were localized in the testes of the marmoset (Callitrix jacchus) whereas CXCR7 patterns were determined for various stages in human testes. Morphometry and flow cytometry were applied to quantify CXCR7-positive cells in monkeys. Transcript levels and protein expression of CXCR7 were detectable throughout testicular development. In both species, CXCR7 protein expression was restricted to premeiotic germ cells. In immature marmoset testes, 69.9% ± 9% of the total germ cell population were labelled for CXCR7, whereas in the adult, 4.7% ± 2.7% were positive for CXCR7. CXCL12 mRNA was detectable in all developmental stages in marmosets. The CXCL12 protein was exclusively localized to Sertoli cells. This pattern of CXCL12/CXCR7 indicates their involvement in regulatory processes that possibly orchestrate the interaction between undifferentiated germ cells and Sertoli cells.
Collapse
Affiliation(s)
- Birgit Westernströer
- Centre of Reproductive Medicine and Andrology, CeRA, Institute of Reproductive and Regenerative Biology, Albert-Schweizer-Campus 1, Building D11, 48149, Münster, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Langenstroth D, Kossack N, Westernströer B, Wistuba J, Behr R, Gromoll J, Schlatt S. Separation of somatic and germ cells is required to establish primate spermatogonial cultures. Hum Reprod 2014; 29:2018-31. [PMID: 24963164 DOI: 10.1093/humrep/deu157] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Can primate spermatogonial cultures be optimized by application of separation steps and well defined culture conditions? SUMMARY ANSWER We identified the cell fraction which provides the best source for primate spermatogonia when prolonged culture is desired. WHAT IS KNOWN ALREADY Man and marmoset show similar characteristics in regard to germ cell development and function. Several protocols for isolation and culture of human testis-derived germline stem cells have been described. Subsequent analysis revealed doubts on the germline origin of these cells and characterized them as mesenchymal stem cells or fibroblasts. Studies using marmosets as preclinical model confirmed that the published isolation protocols did not lead to propagation of germline cells. STUDY DESIGN, SIZE, DURATION Testicular cells derived from nine adult marmoset monkeys (Callithrix jacchus) were cultured for 1, 3, 6 and 11 days and consecutively analyzed for the presence of spermatogonia, differentiating germ cells and testicular somatic cells. PARTICIPANTS/MATERIALS, SETTING, METHODS Testicular tissue of nine adult marmoset monkeys was enzymatically dissociated and subjected to two different cell culture approaches. In the first approach all cells were kept in the same dish (non-separate culture, n = 5). In the second approach the supernatant cells were transferred into a new dish 24 h after seeding and subsequently supernatant and attached cells were cultured separately (separate culture, n = 4). Real-time quantitative PCR and immunofluorescence were used to analyze the expression of reliable germ cell and somatic markers throughout the culture period. Germ cell transplantation assays and subsequent wholemount analyses were performed to functionally evaluate the colonization of spermatogonial cells. MAIN RESULTS AND THE ROLE OF CHANCE This is the first report revealing an efficient isolation and culture of putative marmoset spermatogonial stem cells with colonization ability. Our results indicate that a separation of spermatogonia from testicular somatic cells is a crucial step during cell preparation. We identified the overgrowth of more rapidly expanding somatic cells to be a major problem when establishing spermatogonial cultures. Initiating germ cell cultures from the supernatant and maintaining germ cells in suspension cultures minimized the somatic cell contamination and provided enriched germ cell fractions which displayed after 11 days of culture a significantly higher expression of germ cell markers genes (DDX-4, MAGE A-4; P < 0.05) compared with separately cultured attached cells. Additionally, germ cell transplantation experiments demonstrated a significantly higher absolute number of cells with colonization ability (P < 0.001) in supernatant cells after 11 days of separate culture. LIMITATIONS, REASONS FOR CAUTION This study presents a relevant aspect for the successful setup of spermatogonial cultures but provides limited data regarding the question of whether the long-term maintenance of spermatogonia can be achieved. Transfer of these preclinical data to man may require modifications of the protocol. WIDER IMPLICATIONS OF THE FINDINGS Spermatogonial cultures from rodents have become important and innovative tools for basic and applied research in reproductive biology and veterinary medicine. It is expected that spermatogonia-based strategies will be transformed into clinical applications for the treatment of male infertility. Our data in the marmoset monkey may be highly relevant to establish spermatogonial cultures of human testes. STUDY FUNDING/COMPETING INTERESTS Funding was provided by the DFG-Research Unit FOR 1041 Germ Cell Potential (SCHL394/11-2) and by the Graduate Program Cell Dynamics and Disease (CEDAD) together with the International Max Planck Research School - Molecular Biomedicine (IMPRS-MBM). The authors declare that there is no conflict of interest. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Daniel Langenstroth
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Nina Kossack
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Birgit Westernströer
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Joachim Wistuba
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Rüdiger Behr
- Stem Cell Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Jörg Gromoll
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Stefan Schlatt
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| |
Collapse
|
32
|
Kossack N, Terwort N, Wistuba J, Ehmcke J, Schlatt S, Schöler H, Kliesch S, Gromoll J. A combined approach facilitates the reliable detection of human spermatogonia in vitro. Hum Reprod 2013; 28:3012-25. [PMID: 24001715 DOI: 10.1093/humrep/det336] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Does a combined approach allow for the unequivocal detection of human germ cells and particularly of spermatogonia in vitro? SUMMARY ANSWER Based on our findings, we conclude that an approach comprising: (i) the detailed characterization of patients and tissue samples prior to the selection of biopsies, (ii) the use of unambiguous markers for the characterization of cultures and (iii) the use of biopsies lacking the germ cell population as a negative control is the prerequisite for the establishment of human germ cell cultures. WHAT IS KNOWN ALREADY The use of non-specific marker genes and the failure to assess the presence of testicular somatic cell types in germ cell cultures may have led to a misinterpretation of results and the erroneous description of germ cells in previous studies. STUDY DESIGN, SIZE, DURATION Testicular biopsies were selected from a pool of 264 consecutively obtained biopsies. Based on the histological diagnosis, biopsies with distinct histological phenotypes were selected (n = 35) to analyze the expression of germ cell and somatic cell markers. For germ cell culture experiments, gonadotrophin levels and clinical data were used as selection criteria resulting in the following two groups: (i) biopsies with qualitatively intact spermatogenesis (n = 4) and (ii) biopsies from Klinefelter syndrome Klinefelter patients lacking the germ cell population (n = 3). PARTICIPANTS/MATERIALS, SETTING, METHODS Quantitative real-time PCR analyses were performed to evaluate the specificity of 18 selected germ cell and 3 somatic marker genes. Cell specificity of individual markers was subsequently validated using immunohistochemistry. Finally, testicular cell cultures were established and were analyzed after 10 days for the expression of germ cell- (UTF1, FGFR3, MAGE A4, DDX4) and somatic cell-specific markers (SMA, VIM, LHCGR) at the RNA and the protein levels. MAIN RESULTS AND THE ROLE OF CHANCE Interestingly, only 9 out of 18 marker genes reflected the presence of germ cells and cell specificity could be validated using immunohistochemistry. Furthermore, VIM, SMA and LHCGR were found to reflect the presence of testicular somatic cells at the RNA and the protein levels. Using this validated marker panel and biopsies lacking the germ cell population (n = 3) as a negative control, we demonstrated that germ cell cultures containing spermatogonia can be established from biopsies with normal spermatogenesis (n = 4) and that these cultures can be maintained for the period of 10 days. However, marker profiling has to be performed at regular time points as the composition of testicular cell types may continuously change under longer term culture conditions. LIMITATIONS, REASONS FOR CAUTION There are significant differences regarding the spermatogonial stem cell (SSC) system and spermatogenesis between rodents and primates. It is therefore possible that marker genes that do not reflect the presence of spermatogonia in the human are specific for spermatogonia in other animal models. WIDER IMPLICATIONS OF THE FINDINGS While some studies have reported that human SSCs can be maintained in vitro and show characteristics of pluripotency, the germ cell origin and the differentiation potential of these cells were subsequently called into question. This study provides critical insights into possible sources for the misinterpretation of results regarding the presence of germ cells in human testicular cell cultures and our findings can therefore help to avoid conflicting reports in the future. STUDY FUNDING/COMPETING INTEREST(S) This project was supported by the Stem Cell Network North Rhine-Westphalia and the Innovative Medical Research of the University of Münster Medical School (Grant KO111014). In addition, it was funded by the DFG-Research Unit FOR 1041 Germ Cell Potential (GR 1547/11-1 and SCHL 394/11-2), the BMBF (01GN0809/10) and the IZKF (CRA 03/09). The authors declare that there is no conflict of interest. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- N Kossack
- Institute for Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus 1 (D11), Münster 48149, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Aeckerle N, Dressel R, Behr R. Grafting of Neonatal Marmoset Monkey Testicular Single-Cell Suspensions into Immunodeficient Mice Leads to ex situ Testicular Cord Neomorphogenesis. Cells Tissues Organs 2013; 198:209-20. [DOI: 10.1159/000355339] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2013] [Indexed: 11/19/2022] Open
|
34
|
Costa GMJ, Avelar GF, Rezende-Neto JV, Campos-Junior PHA, Lacerda SMSN, Andrade BSC, Thomé RG, Hofmann MC, Franca LR. Spermatogonial stem cell markers and niche in equids. PLoS One 2012; 7:e44091. [PMID: 22937157 PMCID: PMC3429436 DOI: 10.1371/journal.pone.0044091] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/01/2012] [Indexed: 01/15/2023] Open
Abstract
Spermatogonial stem cells (SSCs) are the foundation of spermatogenesis and are located in a highly dynamic microenvironment called "niche" that influences all aspects of stem cell function, including homing, self-renewal and differentiation. Several studies have recently identified specific proteins that regulate the fate of SSCs. These studies also aimed at identifying surface markers that would facilitate the isolation of these cells in different vertebrate species. The present study is the first to investigate SSC physiology and niche in stallions and to offer a comparative evaluation of undifferentiated type A spermatogonia (Aund) markers (GFRA1, PLZF and CSF1R) in three different domestic equid species (stallions, donkeys, and mules). Aund were first characterized according to their morphology and expression of the GFRA1 receptor. Our findings strongly suggest that in stallions these cells were preferentially located in the areas facing the interstitium, particularly those nearby blood vessels. This distribution is similar to what has been observed in other vertebrate species. In addition, all three Aund markers were expressed in the equid species evaluated in this study. These markers have been well characterized in other mammalian species, which suggests that the molecular mechanisms that maintain the niche and Aund/SSCs physiology are conserved among mammals. We hope that our findings will help future studies needing isolation and cryopreservation of equids SSCs. In addition, our data will be very useful for studies that aim at preserving the germplasm of valuable animals, and involve germ cell transplantation or xenografts of equids testis fragments/germ cells suspensions.
Collapse
Affiliation(s)
- Guilherme M. J. Costa
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gleide F. Avelar
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - José V. Rezende-Neto
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paulo Henrique A. Campos-Junior
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Samyra M. S. N. Lacerda
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno S. C. Andrade
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ralph Gruppi Thomé
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marie-Claude Hofmann
- Unit 1105, Department of Endocrine Neoplasia and Hormonal Disorders, MD Anderson Cancer Center, University of Texas, Houston, Texas, United States of America
| | - Luiz R. Franca
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|