1
|
Kawamura W, Yazawa R, Takeuchi Y, Kamio S, Ichida K, Hattori RS, Morita T, Hayashi M, Yoshizaki G. Bluefin tuna sperm production is hastened by surrogacy in small Euthynnus. Nat Commun 2024; 15:8128. [PMID: 39362867 PMCID: PMC11450220 DOI: 10.1038/s41467-024-52393-4] [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: 01/15/2024] [Accepted: 09/04/2024] [Indexed: 10/05/2024] Open
Abstract
Pacific bluefin tuna (Thunnus orientalis) remains heavily depleted due to overexploitation. Aquaculture and stock enhancement based on artificial seedlings could be effective solutions to this problem. However, widespread adoption of seedling production is limited because spawning in captivity of bluefin tuna, a large pelagic top predator, requires much space, time, cost, and labour. To downsize and shorten their generation time, here, we show that xenogeneic germ cell transplantation enables a surrogate production of functional bluefin tuna sperm in hybrid little tuna (genus: Euthynnus), which attain sexual maturity at a smaller body size and shorter time. Intraperitonially transplanted bluefin tuna germ cells migrate towards and are incorporated into the gonads of little tuna larvae. These recipients produce functional donor-derived bluefin tuna sperm at a mere 8 months of age and 1 kg of body size. This result represents an advance toward compact, low-cost, and time-efficient seedling production that will improve the efficiency of bluefin tuna aquaculture.
Collapse
Affiliation(s)
- Wataru Kawamura
- Institute for Reproductive Biotechnology for Aquatic Species, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan
| | - Ryosuke Yazawa
- Institute for Reproductive Biotechnology for Aquatic Species, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan
| | - Yutaka Takeuchi
- Faculty of Biological Science and Technology, Kanazawa University, Kakuma, Kanazawa, Ishikawa, 920-1192, Japan
| | - Shigeharu Kamio
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan
| | - Kensuke Ichida
- Institute for Reproductive Biotechnology for Aquatic Species, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan
| | - Ricardo Shohei Hattori
- Salmonid Experimental Station at Campos do Jordão, Unidade de Pesquisa e Desenvolvimento-Campos do Jordão, Agência Paulista de Tecnologia dos Agronegócios, Secretaria de Agricultura, São Paulo, 12460-000, Brazil
| | - Tetsuro Morita
- Institute for Reproductive Biotechnology for Aquatic Species, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan
| | - Makoto Hayashi
- Institute for Reproductive Biotechnology for Aquatic Species, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan
| | - Goro Yoshizaki
- Institute for Reproductive Biotechnology for Aquatic Species, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan.
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan.
| |
Collapse
|
2
|
Sriram S, Macedo T, Mavinkurve‐Groothuis A, van de Wetering M, Looijenga LHJ. Alkylating agents-induced gonadotoxicity in prepubertal males: Insights on the clinical and preclinical front. Clin Transl Sci 2024; 17:e13866. [PMID: 38965809 PMCID: PMC11224131 DOI: 10.1111/cts.13866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
Rising cure rates in pediatric cancer patients warrants an increased attention toward the long-term consequences of the diagnosis and treatment in survivors. Chemotherapeutic agents can be gonadotoxic, rendering them at risk for infertility post-survival. While semen cryopreservation is an option that can be provided for most (post)pubertal boys before treatment, this is unfortunately not an option prepubertal in age, simply due to the lack of spermatogenesis. Over the last couple of years, studies have thus focused on better understanding the testis niche in response to various chemotherapeutic agents that are commonly administered and their direct and indirect impact on the germ cell populations. These are generally compounds that have a high risk of infertility and have been classified into risk categories in curated fertility guidelines. However, with it comes the lack of evidence and the challenge of using informative models and conditions most reflective of the physiological scenario, in short, the appropriate study designs for clinically relevant outcomes. Besides, the exact mechanism(s) of action for many of these "risk" compounds as well as other agents is unclear. Understanding their behavior and effect on the testis niche will pave the way for incorporating new strategies to ultimately combat infertility. Of the various drug classes, alkylating agents pose the highest risk of gonadotoxicity as per previously established studies as well as risk stratification guidelines. Therefore, this review will summarize the findings in the field of male fertility concerning gonadotoxicity of akylating agents as a result of chemotherapy exposure.
Collapse
Affiliation(s)
- Sruthi Sriram
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Tiago Macedo
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | | |
Collapse
|
3
|
Blank MH, Kawaoku AJT, Rui BR, Carreira ACO, Hamilton TRDS, Goissis MD, Pereira RJG. Successful xenotransplantation of testicular cells following fractionated chemotherapy of recipient birds. Sci Rep 2024; 14:3085. [PMID: 38321093 PMCID: PMC10847125 DOI: 10.1038/s41598-023-45019-0] [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: 05/19/2023] [Accepted: 10/14/2023] [Indexed: 02/08/2024] Open
Abstract
An essential step in the success of germ cell transplantation is the preparation of the recipient's testicular environment to increase the availability of stem cell niches. However, most methods for this purpose in birds face serious limitations such as partial germ cell depletion, high toxicity and mortality, or the need to use expensive technologies. Here, we validated a simple and practical technique of transferring quail testicular cells into chicken testes depleted of endogenous spermatozoa by fractioned chemotherapy (20 mg/kg/week busulfan for 5 weeks). This protocol resulted in a very low mortality of the treated day-old chicks and, despite maintenance of androgenic activity, sperm production was decreased by 84.3% at 25 weeks of age. NANOG immunostaining revealed that very few to no germ cells were present following treatment with 20 and 40 mg/kg, respectively. RT-qPCR data also showed that c-MYC and NANOG expression declined in these treatments, but GRFα1 and BID expressions remained unaltered among groups. After xenotransplantation, quail germ cells were immunodetected in chicken testes using a species-specific antibody (QCPN), and quail ovalbumin DNA was found in seminal samples collected from chicken recipients. Together, these data confirm that fractionated administration of busulfan in hatchlings is a practical, effective, and safe protocol to prepare recipient male birds capable of supporting xenogeneic spermatogenesis.
Collapse
Affiliation(s)
- Marcel Henrique Blank
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil.
| | | | - Bruno Rogério Rui
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil
| | - Ana Claudia Oliveira Carreira
- Cell and Molecular Therapy Center (NUCEL), Medical School, University of Sao Paulo, Rua Pangaré 100, São Paulo, 05360-130, Brazil
| | - Thais Rose Dos Santos Hamilton
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil
| | - Marcelo Demarchi Goissis
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil
| | - Ricardo José Garcia Pereira
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil.
| |
Collapse
|
4
|
Song Y, Zhang X, Desmarais JA, Nagano M. Postnatal development of mouse spermatogonial stem cells as determined by immunophenotype, regenerative capacity, and long-term culture-initiating ability: a model for practical applications. Sci Rep 2024; 14:2299. [PMID: 38280889 PMCID: PMC10821885 DOI: 10.1038/s41598-024-52824-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/24/2024] [Indexed: 01/29/2024] Open
Abstract
Spermatogonial stem cells (SSCs) are the foundation of life-long spermatogenesis. While SSC research has advanced greatly over the past two decades, characterization of SSCs during postnatal development has not been well documented. Using the mouse as a model, in this study, we defined the immunophenotypic profiles of testis cells during the course of postnatal development using multi-parameter flow cytometry with up to five cell-surface antigens. We found that the profiles progress over time in a manner specific to developmental stages. We then isolated multiple cell fractions at different developmental stages using fluorescent-activated cell sorting (FACS) and identified specific cell populations with prominent capacities to regenerate spermatogenesis upon transplantation and to initiate long-term SSC culture. The data indicated that the cell fraction with the highest level of regeneration capacity exhibited the most prominent potential to initiate SSC culture, regardless of age. Interestingly, refinement of cell fractionation using GFRA1 and KIT did not lead to further enrichment of regenerative and culture-initiating stem cells, suggesting that when a high degree of SSC enrichment is achieved, standard markers of SSC self-renewal or commitment may lose their effectiveness to distinguish cells at the stem cell state from committed progenitors. This study provides a significant information resource for future studies and practical applications of mammalian SSCs.
Collapse
Affiliation(s)
- Youngmin Song
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
| | - Xiangfan Zhang
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
| | - Joëlle A Desmarais
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
- JEFO Nutrition Inc, 5020 Avenue Jefo, Saint-Hyachinthe, Quebec, J2R 2E7, Canada
| | - Makoto Nagano
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
5
|
Lara NLM, Goldsmith T, Rodriguez-Villamil P, Ongaratto F, Solin S, Webster D, Ganbaatar U, Hodgson S, Corbière SMAS, Bondareva A, Carlson DF, Dobrinski I. DAZL Knockout Pigs as Recipients for Spermatogonial Stem Cell Transplantation. Cells 2023; 12:2582. [PMID: 37947660 PMCID: PMC10649044 DOI: 10.3390/cells12212582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Spermatogonial stem cell (SSC) transplantation into the testis of a germ cell (GC)-depleted surrogate allows transmission of donor genotype via donor-derived sperm produced by the recipient. Transplantation of gene-edited SSCs provides an approach to propagate gene-edited large animal models. DAZL is a conserved RNA-binding protein important for GC development, and DAZL knockout (KO) causes defects in GC commitment and differentiation. We characterized DAZL-KO pigs as SSC transplantation recipients. While there were GCs in 1-week-old (wko) KO, complete GC depletion was observed by 10 wko. Donor GCs were transplanted into 18 DAZL-KO recipients at 10-13 wko. At sexual maturity, semen and testes were evaluated for transplantation efficiency and spermatogenesis. Approximately 22% of recipient seminiferous tubules contained GCs, including elongated spermatids and proliferating spermatogonia. The ejaculate of 89% of recipients contained sperm, exclusively from donor origin. However, sperm concentration was lower than the wild-type range. Testicular protein expression and serum hormonal levels were comparable between DAZL-KO and wild-type. Intratesticular testosterone and Leydig cell volume were increased, and Leydig cell number decreased in transplanted DAZL-KO testis compared to wild-type. In summary, DAZL-KO pigs support donor-derived spermatogenesis following SSC transplantation, but low spermatogenic efficiency currently limits their use for the production of offspring.
Collapse
Affiliation(s)
- Nathalia L. M. Lara
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N4N1, Canada; (N.L.M.L.); (A.B.)
| | - Taylor Goldsmith
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | | | - Felipe Ongaratto
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Staci Solin
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Dennis Webster
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Uyanga Ganbaatar
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Shane Hodgson
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | | | - Alla Bondareva
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N4N1, Canada; (N.L.M.L.); (A.B.)
| | - Daniel F. Carlson
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Ina Dobrinski
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N4N1, Canada; (N.L.M.L.); (A.B.)
| |
Collapse
|
6
|
Reifarth L, Körber H, Packeiser EM, Goericke-Pesch S. Detection of spermatogonial stem cells in testicular tissue of dogs with chronic asymptomatic orchitis. Front Vet Sci 2023; 10:1205064. [PMID: 37396999 PMCID: PMC10311113 DOI: 10.3389/fvets.2023.1205064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
Chronic asymptomatic idiopathic orchitis (CAO) is an important but neglected cause of acquired infertility due to non-obstructive azoospermia (NOA) in male dogs. The similarity of the pathophysiology in infertile dogs and men supports the dog's suitability as a possible animal model for studying human diseases causing disruption of spermatogenesis and evaluating the role of spermatogonial stem cells (SSCs) as a new therapeutic approach to restore or recover fertility in cases of CAO. To investigate the survival of resilient stem cells, the expression of the protein gene product (PGP9.5), deleted in azoospermia like (DAZL), foxo transcription factor 1 (FOXO1) and tyrosine-kinase receptor (C-Kit) were evaluated in healthy and CAO-affected canine testes. Our data confirmed the presence of all investigated germ cell markers at mRNA and protein levels. In addition, we postulate a specific expression pattern of FOXO1 and C-Kit in undifferentiated and differentiating spermatogonia, respectively, whereas DAZL and PGP9.5 expressions were confirmed in the entire spermatogonial population. Furthermore, this is the first study revealing a significant reduction of PGP9.5, DAZL, and FOXO1 in CAO at protein and/or gene expression level indicating a severe disruption of spermatogenesis. This means that chronic asymptomatic inflammatory changes in CAO testis are accompanied by a significant loss of SSCs. Notwithstanding, our data confirm the survival of putative stem cells with the potential of self-renewal and differentiation and lay the groundwork for further research into stem cell-based therapeutic options to reinitialize spermatogenesis in canine CAO-affected patients.
Collapse
Affiliation(s)
| | | | | | - Sandra Goericke-Pesch
- Reproductive Unit – Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
7
|
Huang L, Zhang J, Zhang P, Huang X, Yang W, Liu R, Sun Q, Lu Y, Zhang M, Fu Q. Single-cell RNA sequencing uncovers dynamic roadmap and cell-cell communication during buffalo spermatogenesis. iScience 2022; 26:105733. [PMID: 36582818 PMCID: PMC9793287 DOI: 10.1016/j.isci.2022.105733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Spermatogenesis carries the task of precise intergenerational transmission of genetic information from the paternal genome and involves complex developmental processes regulated by the testicular microenvironment. Studies performed mainly in mouse models have established the theoretical basis for spermatogenesis, yet the wide interspecies differences preclude direct translation of the findings, and farm animal studies are progressing slowly. More than 32,000 cells from prepubertal (3-month-old) and pubertal (24-month-old) buffalo testes were analyzed by using single-cell RNA sequencing (scRNA-seq), and dynamic gene expression roadmaps of germ and somatic cell development were generated. In addition to identifying the dynamic processes of sequential cell fate transitions, the global cell-cell communication essential to maintain regular spermatogenesis in the buffalo testicular microenvironment was uncovered. The findings provide the theoretical basis for establishing buffalo germline stem cells in vitro or culturing organoids and facilitating the expansion of superior livestock breeding.
Collapse
Affiliation(s)
- Liangfeng Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Junjun Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Pengfei Zhang
- Institute of Medical and Health, Guangxi Academy of Sciences, Nanning 530007, China
| | - Xingchen Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Weihan Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Runfeng Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Qinqiang Sun
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| | - Qiang Fu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| |
Collapse
|
8
|
Thiageswaran S, Steele H, Voigt AL, Dobrinski I. A Role for Exchange of Extracellular Vesicles in Porcine Spermatogonial Co-Culture. Int J Mol Sci 2022; 23:ijms23094535. [PMID: 35562927 PMCID: PMC9103065 DOI: 10.3390/ijms23094535] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 12/22/2022] Open
Abstract
Spermatogonial stem cells (SSCs) provide the basis for lifelong male fertility through self-renewal and differentiation. Prepubertal male cancer patients may be rendered infertile by gonadotoxic chemotherapy and, unlike sexually mature men, cannot store sperm. Alternatively, testicular biopsies taken prior to treatment may be used to restore fertility in adulthood. Testicular SSC populations are limited, and in vitro culture systems are required to increase numbers of SSCs for treatment, demanding culture systems for SSC propagation. Using the pig as a non-rodent model, we developed culture systems to expand spermatogonia from immature testis tissue, comparing different feeders (Sertoli cells, peritubular myoid cells (PMCs) and pig fetal fibroblasts (PFFs)). Spermatogonia co-cultured with Sertoli cells, PMCs and PFFs had comparable rates of proliferation and apoptosis. To elucidate the mechanism behind the beneficial nature of feeder layers, we investigated the role of extracellular vesicles in crosstalk between spermatogonia and feeder cells. Sertoli cell-released exosomes are incorporated by spermatogonia, and inhibition of exosomal release reduces spermatogonial proliferation. Together, these results show that PMCs, PFFs and Sertoli cells promote spermatogonial proliferation in co-culture, with exosomal exchange representing one possible mechanism. Further characterization of exosomal cargo may ultimately allow the development of feeder-free culture systems for clinical use.
Collapse
Affiliation(s)
- Shiama Thiageswaran
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Heather Steele
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (H.S.); (A.L.V.)
| | - Anna Laura Voigt
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (H.S.); (A.L.V.)
| | - Ina Dobrinski
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (H.S.); (A.L.V.)
- Correspondence: ; Tel.: +1-403-210-6532
| |
Collapse
|
9
|
Fayaz MA, Ibtisham F, Cham TC, Honaramooz A. Culture supplementation of bFGF, GDNF, and LIF alters in vitro proliferation, colony formation, and pluripotency of neonatal porcine germ cells. Cell Tissue Res 2022; 388:195-210. [PMID: 35102441 DOI: 10.1007/s00441-022-03583-3] [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: 06/11/2021] [Accepted: 01/12/2022] [Indexed: 11/25/2022]
Abstract
Gonocytes in the neonatal testis have male germline stem cell properties and as such have important potential applications in fertility preservation and regenerative medicine. Such applications require further studies aimed at increasing gonocyte numbers and evaluating their pluripotency in vitro. The objective of the present study was to test the effects of basic fibroblast growth factor (bFGF), glial cell line-derived neurotrophic factor (GDNF), and leukemia inhibitory factor (LIF) on in vitro propagation, colony formation, and expression of pluripotency markers of neonatal porcine gonocytes. Testis cells from 1-week-old piglets were cultured in basic media (DMEM + 15% FBS), supplemented with various concentrations of bFGF, GDNF, and LIF, either individually or in combinations, in a stepwise experimental design. Gonocytes and/or their colonies were evaluated every 7 days and the gonocyte- (DBA) and pluripotency-specific markers (POU5F1, SSEA-1, E-cadherin, and NANOG) assessed on day 28. Greatest gonocyte numbers and largest colonies were found in media supplemented with 10 ng/mL bFGF and 10 ng/mL bFGF + 100 ng/mL GDNF + 1500 U/mL LIF, respectively. The resultant gonocytes and colonies expressed both germ cell- and pluripotency-specific markers. These results shed light on the growth hormone requirements of porcine gonocytes for in vitro proliferation and colony formation.
Collapse
Affiliation(s)
- Mohammad Amin Fayaz
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, S7N 5B4, Canada
| | - Fahar Ibtisham
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, S7N 5B4, Canada
| | - Tat-Chuan Cham
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, S7N 5B4, Canada
| | - Ali Honaramooz
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, S7N 5B4, Canada.
| |
Collapse
|
10
|
Long Noncoding RNA Mediated Regulation in Human Embryogenesis, Pluripotency, and Reproduction. Stem Cells Int 2022; 2022:8051717. [PMID: 35103065 PMCID: PMC8800634 DOI: 10.1155/2022/8051717] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022] Open
Abstract
Long noncoding RNAs (lncRNAs), a class of noncoding RNAs with more than 200 bp in length, are produced by pervasive transcription in mammalian genomes and regulate gene expression through various action mechanisms. Accumulating data indicate that lncRNAs mediate essential biological functions in human development, including early embryogenesis, induction of pluripotency, and germ cell development. Comprehensive analysis of sequencing data highlights that lncRNAs are expressed in a stage-specific and human/primate-specific pattern during early human development. They contribute to cell fate determination through interacting with almost all classes of cellular biomolecules, including proteins, DNA, mRNAs, and microRNAs. Furthermore, the expression of a few of lncRNAs is highly associated with the pathogenesis and progression of many reproductive diseases, suggesting that they could serve as candidate biomarkers for diagnosis or novel targets for treatment. Here, we review research on lncRNAs and their roles in embryogenesis, pluripotency, and reproduction. We aim to identify the underlying molecular mechanisms essential for human development and provide novel insight into the causes and treatments of human reproductive diseases.
Collapse
|
11
|
Implications of testicular ACE2 and the renin-angiotensin system for SARS-CoV-2 on testis function. Nat Rev Urol 2022; 19:116-127. [PMID: 34837081 PMCID: PMC8622117 DOI: 10.1038/s41585-021-00542-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2021] [Indexed: 12/16/2022]
Abstract
Although many studies have focused on SARS-CoV-2 infection in the lungs, comparatively little is known about the potential effects of the virus on male fertility. SARS-CoV-2 infection of target cells requires the presence of furin, angiotensin-converting enzyme 2 (ACE2) receptors, and transmembrane protease serine 2 (TMPRSS2). Thus, cells in the body that express these proteins might be highly susceptible to viral entry and downstream effects. Currently, reports regarding the expression of the viral entry proteins in the testes are conflicting; however, other members of the SARS-CoV family of viruses - such as SARS-CoV - have been suspected to cause testicular dysfunction and/or orchitis. SARS-CoV-2, which displays many similarities to SARS-CoV, could potentially cause similar adverse effects. Commonalities between SARS family members, taken in combination with sparse reports of testicular discomfort and altered hormone levels in patients with SARS-CoV-2, might indicate possible testicular dysfunction. Thus, SARS-CoV-2 infection has the potential for effects on testis somatic and germline cells and experimental approaches might be required to help identify potential short-term and long-term effects of SARS-CoV-2 on male fertility.
Collapse
|
12
|
Long-Term Ex Vivo Expansion of Murine Spermatogonial Stem Cells in a Simple Serum-Free Medium. Methods Mol Biol 2021. [PMID: 32474876 DOI: 10.1007/978-1-0716-0655-1_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Spermatogonial stem cells (SSCs) possess both self-renewal and differentiation abilities to sustain lifelong production of enormous numbers of spermatozoa in males. SSCs hold a unique position among tissue-specific stem cells in adults because of their ability to transmit the genetic information to subsequent generations. Ex vivo expansion of SSCs in conjunction with their transplantation is highly invaluable to study SSCs and develop new reproductive technologies for therapeutic applications. In this chapter, we describe a culture system involving a simple serum-free medium for mouse SSCs. Elimination of the serum from the culture is important to enhance the effects of exogenous factors, which are rather masked by the serum, and to avert the serum-induced inflammatory responses of testicular mesenchymal cells, which cause adverse effects on SSC proliferation. Consequently, using this culture system has proven for the first time that glial cell line-derived neurotrophic factor (GDNF) was found to be the key factor to drive the self-renewing proliferation of SSCs, and fibroblast growth factor 2 enhanced the GDNF-dependent proliferation of SSCs. Besides determining these two key cytokines, the simplicity of the system enabled individual modification of its components to develop long-term cultures of rat and rabbit SSCs. The basics of these culture systems will enable development of the culture conditions for human and other mammalian SSCs in the near future.
Collapse
|
13
|
Sakib S, Uchida A, Valenzuela-Leon P, Yu Y, Valli-Pulaski H, Orwig K, Ungrin M, Dobrinski I. Formation of organotypic testicular organoids in microwell culture†. Biol Reprod 2020; 100:1648-1660. [PMID: 30927418 DOI: 10.1093/biolre/ioz053] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/29/2019] [Indexed: 01/15/2023] Open
Abstract
Three-dimensional (3D) organoids can serve as an in vitro platform to study cell-cell interactions, tissue development, and toxicology. Development of organoids with tissue architecture similar to testis in vivo has remained a challenge. Here, we present a microwell aggregation approach to establish multicellular 3D testicular organoids from pig, mouse, macaque, and human. The organoids consist of germ cells, Sertoli cells, Leydig cells, and peritubular myoid cells forming a distinct seminiferous epithelium and interstitial compartment separated by a basement membrane. Sertoli cells in the organoids express tight junction proteins claudin 11 and occludin. Germ cells in organoids showed an attenuated response to retinoic acid compared to germ cells in 2D culture indicating that the tissue architecture of the organoid modulates response to retinoic acid similar to in vivo. Germ cells maintaining physiological cell-cell interactions in organoids also had lower levels of autophagy indicating lower levels of cellular stress. When organoids were treated with mono(2-ethylhexyl) phthalate (MEHP), levels of germ cell autophagy increased in a dose-dependent manner, indicating the utility of the organoids for toxicity screening. Ablation of primary cilia on testicular somatic cells inhibited the formation of organoids demonstrating an application to screen for factors affecting testicular morphogenesis. Organoids can be generated from cryopreserved testis cells and preserved by vitrification. Taken together, the testicular organoid system recapitulates the 3D organization of the mammalian testis and provides an in vitro platform for studying germ cell function, testicular development, and drug toxicity in a cellular context representative of the testis in vivo.
Collapse
Affiliation(s)
- Sadman Sakib
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Aya Uchida
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada
| | - Paula Valenzuela-Leon
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada
| | - Yang Yu
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Hanna Valli-Pulaski
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kyle Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mark Ungrin
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ina Dobrinski
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
14
|
Zhang P, Li F, Zhang L, Lei P, Zheng Y, Zeng W. Stage-specific embryonic antigen 4 is a membrane marker for enrichment of porcine spermatogonial stem cells. Andrology 2020; 8:1923-1934. [PMID: 32691968 DOI: 10.1111/andr.12870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Spermatogonial stem cells (SSCs), as tissue-specific stem cells, are capable of both self-renewal and differentiation and supporting the continual and robust spermatogenesis for male fertility. As a rare sub-fraction of undifferentiated spermatogonia, SSCs share most molecular markers with the progenitor spermatogonia. Thus, the heterogeneity of the progenitor cells often obscures the characteristics of stem cells. Distinguishing SSCs from the progenitors is of paramount importance to understand the regulatory mechanisms governing their actions. OBJECTIVES The present study was designed to reveal that SSEA4 can be a marker for putative porcine SSCs that distinguished it from the progenitors and to build a sorting program for efficient enrichment of porcine SSCs. METHODS To explore expression of SSEA4 within the undifferentiated spermatogonial population, we performed co-immunofluorescent staining for SSEA4 and common molecular markers (VASA, DBA, PLZF, c-KIT, and SOX9) in the 7-, 90-, and 150-day-old porcine testicular tissues. SSEA4-positive cells were isolated from the 90-day-old porcine testes by flow cytometry. Immunofluorescent, RNA-sequencing, and transplantation analysis were used to reveal that SSEA4-positive fraction holds the stem cell capacity. RESULTS We found that SSEA4 was expressed in a rare sub-fraction of porcine undifferentiated spermatogonia, and RNA-sequencing analysis revealed that the genes for stem cell maintenance and SSC-specific markers (ID4 and PAX7) were up-regulated in the SSEA4-sorted fraction, compared with undifferentiated spermatogonia. In addition, germ cell transplantation assay demonstrated that SSEA4-positive spermatogonia colonized in the recipient testicular tubules. Sorting of the undifferentiated spermatogonia with anti-SSEA4 antibody resulted in a 2.5-fold enrichment of SSCs compared with the germ cell gate group, and 21-fold enrichment of SSCs compared with the SSEA4-negative spermatogonia group. CONCLUSIONS Our findings revealed that SSEA4 is a new surface marker for porcine undifferentiated spermatogonia. This finding helps to elucidate the characteristics of porcine SSCs and facilitates the culture and manipulation of SSCs.
Collapse
Affiliation(s)
- Pengfei Zhang
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Fuyuan Li
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Lingkai Zhang
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Peipei Lei
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yi Zheng
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Wenxian Zeng
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
15
|
The study and manipulation of spermatogonial stem cells using animal models. Cell Tissue Res 2020; 380:393-414. [PMID: 32337615 DOI: 10.1007/s00441-020-03212-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023]
Abstract
Spermatogonial stem cells (SSCs) are a rare group of cells in the testis that undergo self-renewal and complex sequences of differentiation to initiate and sustain spermatogenesis, to ensure the continuity of sperm production throughout adulthood. The difficulty of unequivocal identification of SSCs and complexity of replicating their differentiation properties in vitro have prompted the introduction of novel in vivo models such as germ cell transplantation (GCT), testis tissue xenografting (TTX), and testis cell aggregate implantation (TCAI). Owing to these unique animal models, our ability to study and manipulate SSCs has dramatically increased, which complements the availability of other advanced assisted reproductive technologies and various genome editing tools. These animal models can advance our knowledge of SSCs, testis tissue morphogenesis and development, germ-somatic cell interactions, and mechanisms that control spermatogenesis. Equally important, these animal models can have a wide range of experimental and potential clinical applications in fertility preservation of prepubertal cancer patients, and genetic conservation of endangered species. Moreover, these models allow experimentations that are otherwise difficult or impossible to be performed directly in the target species. Examples include proof-of-principle manipulation of germ cells for correction of genetic disorders or investigation of potential toxicants or new drugs on human testis formation or function. The primary focus of this review is to highlight the importance, methodology, current and potential future applications, as well as limitations of using these novel animal models in the study and manipulation of male germline stem cells.
Collapse
|
16
|
Sakib S, Goldsmith T, Voigt A, Dobrinski I. Testicular organoids to study cell-cell interactions in the mammalian testis. Andrology 2019; 8:835-841. [PMID: 31328437 DOI: 10.1111/andr.12680] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/03/2019] [Accepted: 06/19/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Over the last ten years, three-dimensional organoid culture has garnered renewed interest, as organoids generated from primary cells or stem cells with cell associations and functions similar to organs in vivo can be a powerful tool to study tissue-specific cell-cell interactions in vitro. Very recently, a few interesting approaches have been put forth for generating testicular organoids for studying the germ cell niche microenvironment. AIM To review different model systems that have been employed to study germ cell biology and testicular cell-cell interactions and discuss how the organoid approach can address some of the shortcomings of those systems. RESULTS AND CONCLUSION Testicular organoids that bear architectural and functional similarities to their in vivo counterparts are a powerful model system to study cell-cell interactions in the germ cell niche. Organoids enable studying samples in humans and other large animals where in vivo experiments are not possible, allow modeling of testicular disease and malignancies and may provide a platform to design more precise therapeutic interventions.
Collapse
Affiliation(s)
- S Sakib
- Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AL, Canada.,Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AL, Canada
| | - T Goldsmith
- Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AL, Canada.,Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AL, Canada
| | - A Voigt
- Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AL, Canada
| | - I Dobrinski
- Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AL, Canada.,Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AL, Canada
| |
Collapse
|
17
|
Kubota H, Brinster RL. Spermatogonial stem cells. Biol Reprod 2019; 99:52-74. [PMID: 29617903 DOI: 10.1093/biolre/ioy077] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/29/2018] [Indexed: 12/19/2022] Open
Abstract
Spermatogonial stem cells (SSCs) are the most primitive spermatogonia in the testis and have an essential role to maintain highly productive spermatogenesis by self-renewal and continuous generation of daughter spermatogonia that differentiate into spermatozoa, transmitting genetic information to the next generation. Since the 1950s, many experimental methods, including histology, immunostaining, whole-mount analyses, and pulse-chase labeling, had been used in attempts to identify SSCs, but without success. In 1994, a spermatogonial transplantation method was reported that established a quantitative functional assay to identify SSCs by evaluating their ability to both self-renew and differentiate to spermatozoa. The system was originally developed using mice and subsequently extended to nonrodents, including domestic animals and humans. Availability of the functional assay for SSCs has made it possible to develop culture systems for their ex vivo expansion, which dramatically advanced germ cell biology and allowed medical and agricultural applications. In coming years, SSCs will be increasingly used to understand their regulation, as well as in germline modification, including gene correction, enhancement of male fertility, and conversion of somatic cells to biologically competent male germline cells.
Collapse
Affiliation(s)
- Hiroshi Kubota
- Laboratory of Cell and Molecular Biology, Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Ralph L Brinster
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Sakib S, Voigt A, Goldsmith T, Dobrinski I. Three-dimensional testicular organoids as novel in vitro models of testicular biology and toxicology. ENVIRONMENTAL EPIGENETICS 2019; 5:dvz011. [PMID: 31463083 PMCID: PMC6705190 DOI: 10.1093/eep/dvz011] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/06/2019] [Accepted: 07/03/2019] [Indexed: 05/05/2023]
Abstract
Organoids are three dimensional structures consisting of multiple cell types that recapitulate the cellular architecture and functionality of native organs. Over the last decade, the advent of organoid research has opened up many avenues for basic and translational studies. Following suit of other disciplines, research groups working in the field of male reproductive biology have started establishing and characterizing testicular organoids. The three-dimensional architectural and functional similarities of organoids to their tissue of origin facilitate study of complex cell interactions, tissue development and establishment of representative, scalable models for drug and toxicity screening. In this review, we discuss the current state of testicular organoid research, their advantages over conventional monolayer culture and their potential applications in the field of reproductive biology and toxicology.
Collapse
Affiliation(s)
- Sadman Sakib
- Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada
| | - Anna Voigt
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada
| | - Taylor Goldsmith
- Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada
| | - Ina Dobrinski
- Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada
- Correspondence address. Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Room 404, Heritage Medical Research Building, 3300 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada. Tel: 4032106523; Fax: 4032108821; E-mail:
| |
Collapse
|
19
|
Savvulidi F, Ptacek M, Savvulidi Vargova K, Stadnik L. Manipulation of spermatogonial stem cells in livestock species. J Anim Sci Biotechnol 2019; 10:46. [PMID: 31205688 PMCID: PMC6560896 DOI: 10.1186/s40104-019-0355-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
We are entering an exciting epoch in livestock biotechnology during which the fundamental approaches (such as transgenesis, spermatozoa cryopreservation and artificial insemination) will be enhanced based on the modern understanding of the biology of spermatogonial stem cells (SSCs) combined with the outstanding recent advances in genomic editing technologies and in vitro cell culture systems. The general aim of this review is to outline comprehensively the promising applications of SSC manipulation that could in the nearest future find practical application in livestock breeding. Here, we will focus on 1) the basics of mammalian SSC biology; 2) the approaches for SSC isolation and purification; 3) the available in vitro systems for the stable expansion of isolated SSCs; 4) a discussion of how the manipulation of SSCs can accelerate livestock transgenesis; 5) a thorough overview of the techniques of SSC transplantation in livestock species (including the preparation of recipients for SSC transplantation, the ultrasonographic-guided SSC transplantation technique in large farm animals, and the perspectives to improve further the SSC transplantation efficiency), and finally, 6) why SSC transplantation is valuable to extend the techniques of spermatozoa cryopreservation and/or artificial insemination. For situations where no reliable data have yet been obtained for a particular livestock species, we will rely on the data obtained from studies conducted in rodents because the knowledge gained from rodent research is translatable to livestock species to a great extent. On the other hand, we will draw special attention to situations where such translation is not possible.
Collapse
Affiliation(s)
- Filipp Savvulidi
- Department of Animal Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Suchdol Czech Republic
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 128 53 Prague, Czech Republic
| | - Martin Ptacek
- Department of Animal Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Suchdol Czech Republic
| | - Karina Savvulidi Vargova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 128 53 Prague, Czech Republic
| | - Ludek Stadnik
- Department of Animal Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Suchdol Czech Republic
| |
Collapse
|
20
|
Kakiuchi K, Taniguchi K, Kubota H. Conserved and non-conserved characteristics of porcine glial cell line-derived neurotrophic factor expressed in the testis. Sci Rep 2018; 8:7656. [PMID: 29769589 PMCID: PMC5955883 DOI: 10.1038/s41598-018-25924-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 05/02/2018] [Indexed: 12/22/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is essential for the self-renewal and proliferation of spermatogonial stem cells (SSCs) in mice, rats, and rabbits. Although the key extrinsic factors essential for spermatogonial proliferation in other mammals have not been determined, GDNF is one of the potential candidates. In this study, we isolated porcine GDNF (pGDNF) cDNAs from neonatal testis and generated recombinant pGDNF to investigate its biological activity on gonocytes/undifferentiated spermatogonia, including SSCs. In porcine testis, long and short forms of GDNF transcripts, the counterparts of pre-(α)pro and pre-(β)pro GDNF identified in humans and rodents, were expressed. The two transcripts encode identical mature proteins. Recombinant pGDNF supported proliferation of murine SSCs in culture, and their stem cell activity was confirmed by a transplantation assay. Subsequently, porcine gonocytes/undifferentiated spermatogonia were cultured with pGDNF; however, pGDNF did not affect their proliferation. Furthermore, GDNF expression was localised to the vascular smooth muscle cells, and its cognate receptor GFRA1 expression was negligible during spermatogonial proliferation in the testes. These results indicate that although pGDNF retains structural similarity with those of other mammals and conserves the biological activity on the self-renewal of murine SSCs, porcine SSCs likely require extrinsic factors other than GDNF for their proliferation.
Collapse
Affiliation(s)
- Kazue Kakiuchi
- Laboratory of Cell and Molecular Biology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Kazumi Taniguchi
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Hiroshi Kubota
- Laboratory of Cell and Molecular Biology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan.
| |
Collapse
|
21
|
Tang L, Bondareva A, González R, Rodriguez-Sosa JR, Carlson DF, Webster D, Fahrenkrug S, Dobrinski I. TALEN-mediated gene targeting in porcine spermatogonia. Mol Reprod Dev 2018; 85:250-261. [PMID: 29393557 DOI: 10.1002/mrd.22961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/22/2018] [Accepted: 01/25/2018] [Indexed: 01/05/2023]
Abstract
Spermatogonia represent a diploid germ cell population that includes spermatogonial stem cells. In this report, we describe new methods for isolation of highly enriched porcine spermatogonia based on light scatter properties, and for targeted mutagenesis in porcine spermatogonia using nucleofection and TALENs. We optimized a nucleofection protocol to deliver TALENs specifically targeting the DMD locus in porcine spermatogonia. We also validated specific sorting of porcine spermatogonia based on light scatter properties. We were able to obtain a highly enriched germ cell population with over 90% of cells being UCH-L1 positive undifferentiated spermatogonia. After gene targeting in porcine spermatogonia, indel (insertion or deletion) mutations as a result of non-homologous end joining (NHEJ) were detected in up to 18% of transfected cells. Our report demonstrates for the first time an approach to obtain a live cell population highly enriched in undifferentiated spermatogonia from immature porcine testes, and that gene targeting can be achieved in porcine spermatogonia which will enable germ line modification.
Collapse
Affiliation(s)
- Lin Tang
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Alla Bondareva
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Raquel González
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Jose R Rodriguez-Sosa
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | | | | | | | - Ina Dobrinski
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| |
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: 0.7] [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
|
Ioannou D, Tempest HG. Does genome organization matter in spermatozoa? A refined hypothesis to awaken the silent vessel. Syst Biol Reprod Med 2018; 64:518-534. [DOI: 10.1080/19396368.2017.1421278] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Dimitrios Ioannou
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- IVF Florida Reproductive Associates, Margate, FL, USA
| | - Helen G. Tempest
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| |
Collapse
|
24
|
Zhang P, Qin Y, Zheng Y, Zeng W. Phospholipase D Family Member 6 Is a Surface Marker for Enrichment of Undifferentiated Spermatogonia in Prepubertal Boars. Stem Cells Dev 2017; 27:55-64. [PMID: 29113556 DOI: 10.1089/scd.2017.0140] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Spermatogonial stem cells (SSCs) have a functional ability to maintain self-renewal and sustain production of spermatozoa throughout the reproductive lifespan of a male. Studies on SSCs can thus better the understandings of spermatogenesis and unravel the mechanisms for self-renewal and differentiation of male germline stem cells. However, the rarity of SSCs in the testis and the lack of reliable surface markers obstruct the related study and further application of SSCs. This is especially the case in livestock animals. In this study, we identified that phospholipase D family member 6 (PLD6) is a surface marker for undifferentiated spermatogonia in boar testes. By magnetic-activated cell sorting, PLD6+ cell fraction comprises 84.45% ± 0.35% of undifferentiated spermatogonia (marked by PLZF). Xenotransplantation of PLD6+ cells into the recipient mouse testis revealed a ninefold increase of donor cell-derived colony formation compared with that in the unselected cell group, indicating the significant enrichment of SSCs. Furthermore, based on the sorted PLD6+ cells with a high SSC content, we established a feeder-free culture system that could maintain porcine undifferentiated spermatogonia for 4 weeks in vitro with the expression of typical markers throughout the culture period. In conclusion, this study demonstrates that PLD6 is a surface marker of undifferentiated spermatogonia in testes of prepubertal boars and could be utilized to unprecedentedly enrich porcine undifferentiated spermatogonia. These data provide the basis for future studies on the refinement of germ cell culture and manipulation of porcine undifferentiated spermatogonia.
Collapse
Affiliation(s)
- Pengfei Zhang
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Yuwei Qin
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Yi Zheng
- 2 Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam , Amsterdam, the Netherlands
| | - Wenxian Zeng
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| |
Collapse
|
25
|
Zak LJ, Gaustad AH, Bolarin A, Broekhuijse MLWJ, Walling GA, Knol EF. Genetic control of complex traits, with a focus on reproduction in pigs. Mol Reprod Dev 2017; 84:1004-1011. [DOI: 10.1002/mrd.22875] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 08/07/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Louisa J. Zak
- Topigs Norsvin Research Center; Beuningen The Netherlands
| | | | | | | | - Grant A. Walling
- JSR Genetics; Southburn; Driffield East Yorkshire United Kingdom
| | - Egbert F. Knol
- Topigs Norsvin Research Center; Beuningen The Netherlands
| |
Collapse
|
26
|
Ibtisham F, Wu J, Xiao M, An L, Banker Z, Nawab A, Zhao Y, Li G. Progress and future prospect of in vitro spermatogenesis. Oncotarget 2017; 8:66709-66727. [PMID: 29029549 PMCID: PMC5630449 DOI: 10.18632/oncotarget.19640] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/12/2017] [Indexed: 12/25/2022] Open
Abstract
Infertility has become a major health issue in the world. It affects the social life of couples and of all infertility cases; approximately 40–50% is due to “male factor” infertility. Male infertility could be due to genetic factors, environment or due to gonadotoxic treatment. Developments in reproductive biotechnology have made it possible to rescue fertility and uphold biological fatherhood. In vitro production of haploid male germ cell is a powerful tool, not only for the treatment of infertility including oligozoospermic or azoospermic patient, but also for the fertility preservation in pre-pubertal boys whose gonadal function is threatened by gonadotoxic therapies. Genomic editing of in-vitro cultured germ cells could also potentially cure flaws in spermatogenesis due to genomic mutation. Furthermore, this ex-vivo maturation technique with genomic editing may be used to prevent paternal transmission of genomic diseases. Here, we summarize the historical progress of in vitro spermatogenesis research by using organ and cell culture techniques and the future clinical application of in vitro spermatogenesis.
Collapse
Affiliation(s)
- Fahar Ibtisham
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Jiang Wu
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Mei Xiao
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Lilong An
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Zachary Banker
- Foreign Language College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Aamir Nawab
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yi Zhao
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Guanghui Li
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| |
Collapse
|
27
|
Lima AC, Jung M, Rusch J, Usmani A, Lopes AM, Conrad DF. Multispecies Purification of Testicular Germ Cells. Biol Reprod 2016; 95:85. [PMID: 27557646 PMCID: PMC5176363 DOI: 10.1095/biolreprod.116.140566] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022] Open
Abstract
Advanced methods of cellular purification are required to apply genome technology to the study of spermatogenesis. One approach, based on flow cytometry of murine testicular cells stained with Hoechst-33342 (Ho-FACS), has been extensively optimized and currently allows the isolation of nine germ cell types. This staining technique is straightforward to implement, is highly effective at purifying specific germ cell types, and yields sufficient cell numbers for high-throughput studies. Ho-FACS is a technique that does not require species-specific markers, but whose applicability to other species is largely unexplored. We hypothesized that, because of the similar cell physiology of spermatogenesis across mammals, Ho-FACS could be used to produce highly purified subpopulations of germ cells in mammals other than mouse. To test this hypothesis, we applied Ho-FACS to four mammalian species that are widely used in testis research: Rattus norvegicus, Cavia porcellus, Canis familiaris, and Sus scrofadomesticus. We successfully isolated four germ cell populations from these species with average purity of 79% for spermatocytes, 90% for spermatids, and 66% for spermatogonia. Additionally, we compare the performance of mechanical and chemical dissociation for each species, and propose an optimized gating strategy to better discriminate round and elongating spermatids in the mouse, which can potentially be applied to other species. Our work indicates that spermatogenesis may be uniquely accessible among mammalian developmental systems, as a single set of reagents may be sufficient to isolate germ cell populations from many different mammalian species, opening new avenues in the fields of development and male reproductive biology.
Collapse
Affiliation(s)
- Ana C. Lima
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
- Graduate Program in Areas of Basic and Applied Biology (GABBA), Abel Salazar Institute of Biomedical Sciences, University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal–IPATIMUP, Porto, Portugal
| | - Min Jung
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Jannette Rusch
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Abul Usmani
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Alexandra M. Lopes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal–IPATIMUP, Porto, Portugal
| | - Donald F. Conrad
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
- Correspondence: Donald F. Conrad, Department of Genetics, Washington University School of Medicine, Campus Box 8232, St. Louis, MO 63110. E-mail:
| |
Collapse
|
28
|
Mulder CL, Zheng Y, Jan SZ, Struijk RB, Repping S, Hamer G, van Pelt AMM. Spermatogonial stem cell autotransplantation and germline genomic editing: a future cure for spermatogenic failure and prevention of transmission of genomic diseases. Hum Reprod Update 2016; 22:561-73. [PMID: 27240817 PMCID: PMC5001497 DOI: 10.1093/humupd/dmw017] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/28/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Subfertility affects approximately 15% of all couples, and a severe male factor is identified in 17% of these couples. While the etiology of a severe male factor remains largely unknown, prior gonadotoxic treatment and genomic aberrations have been associated with this type of subfertility. Couples with a severe male factor can resort to ICSI, with either ejaculated spermatozoa (in case of oligozoospermia) or surgically retrieved testicular spermatozoa (in case of azoospermia) to generate their own biological children. Currently there is no direct treatment for azoospermia or oligozoospermia. Spermatogonial stem cell (SSC) autotransplantation (SSCT) is a promising novel clinical application currently under development to restore fertility in sterile childhood cancer survivors. Meanwhile, recent advances in genomic editing, especially the clustered regulatory interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9) system, are likely to enable genomic rectification of human SSCs in the near future. OBJECTIVE AND RATIONALE The objective of this review is to provide insights into the prospects of the potential clinical application of SSCT with or without genomic editing to cure spermatogenic failure and to prevent transmission of genetic diseases. SEARCH METHODS We performed a narrative review using the literature available on PubMed not restricted to any publishing year on topics of subfertility, fertility treatments, (molecular regulation of) spermatogenesis and SSCT, inherited (genetic) disorders, prenatal screening methods, genomic editing and germline editing. For germline editing, we focussed on the novel CRISPR-Cas9 system. We included papers written in English only. OUTCOMES Current techniques allow propagation of human SSCs in vitro, which is indispensable to successful transplantation. This technique is currently being developed in a preclinical setting for childhood cancer survivors who have stored a testis biopsy prior to cancer treatment. Similarly, SSCT could be used to restore fertility in sterile adult cancer survivors. In vitro propagation of SSCs might also be employed to enhance spermatogenesis in oligozoospermic men and in azoospermic men who still have functional SSCs albeit in insufficient numbers. The combination of SSCT with genomic editing techniques could potentially rectify defects in spermatogenesis caused by genomic mutations or, more broadly, prevent transmission of genomic diseases to the offspring. In spite of the promising prospects, SSCT and germline genomic editing are not yet clinically applicable and both techniques require optimization at various levels. WIDER IMPLICATIONS SSCT with or without genomic editing could potentially be used to restore fertility in cancer survivors to treat couples with a severe male factor and to prevent the paternal transmission of diseases. This will potentially allow these couples to have their own biological children. Technical development is progressing rapidly, and ethical reflection and societal debate on the use of SSCT with or without genomic editing is pressing.
Collapse
Affiliation(s)
- Callista L Mulder
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Yi Zheng
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Sabrina Z Jan
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Robert B Struijk
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Sjoerd Repping
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Geert Hamer
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Ans M M van Pelt
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
29
|
Saragusty J, Diecke S, Drukker M, Durrant B, Friedrich Ben-Nun I, Galli C, Göritz F, Hayashi K, Hermes R, Holtze S, Johnson S, Lazzari G, Loi P, Loring JF, Okita K, Renfree MB, Seet S, Voracek T, Stejskal J, Ryder OA, Hildebrandt TB. Rewinding the process of mammalian extinction. Zoo Biol 2016; 35:280-92. [PMID: 27142508 DOI: 10.1002/zoo.21284] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/11/2016] [Indexed: 12/15/2022]
Abstract
With only three living individuals left on this planet, the northern white rhinoceros (Ceratotherium simum cottoni) could be considered doomed for extinction. It might still be possible, however, to rescue the (sub)species by combining novel stem cell and assisted reproductive technologies. To discuss the various practical options available to us, we convened a multidisciplinary meeting under the name "Conservation by Cellular Technologies." The outcome of this meeting and the proposed road map that, if successfully implemented, would ultimately lead to a self-sustaining population of an extremely endangered species are outlined here. The ideas discussed here, while centered on the northern white rhinoceros, are equally applicable, after proper adjustments, to other mammals on the brink of extinction. Through implementation of these ideas we hope to establish the foundation for reversal of some of the effects of what has been termed the sixth mass extinction event in the history of Earth, and the first anthropogenic one. Zoo Biol. 35:280-292, 2016. © 2016 The Authors. Zoo Biology published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joseph Saragusty
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | | | - Micha Drukker
- Institute of Stem Cell Research, German Research Center for Environmental Health, Helmholtz Center Munich, Neuherberg, Germany
| | - Barbara Durrant
- San Diego Zoo Institute for Conservation Research, Escondido, California
| | - Inbar Friedrich Ben-Nun
- Department of Chemical Physiology, Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California
| | - Cesare Galli
- Avantea srl, Laboratorio di Tecnologie della Riproduzione, Cremona, Italy.,Dipartimento Scienze Mediche Veterinarie, Università di Bologna, Ozzano dell'Emilia, Italy.,Fondazione Avantea, Cremona, Italy
| | - Frank Göritz
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Katsuhiko Hayashi
- Faculty of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Robert Hermes
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Susanne Holtze
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | | | - Giovanna Lazzari
- Avantea srl, Laboratorio di Tecnologie della Riproduzione, Cremona, Italy.,Fondazione Avantea, Cremona, Italy
| | - Pasqualino Loi
- Faculty of Veterinary Medicine, Univeristy of Teramo, Campus Coste San Agostino, Teramo, Italy
| | - Jeanne F Loring
- Department of Chemical Physiology, Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California
| | - Keisuke Okita
- Center for iPS Cell Research and Application, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Steven Seet
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | | | - Jan Stejskal
- ZOO Dvůr Králové, Dvůr Králové nad Labem, Czech Republic
| | - Oliver A Ryder
- San Diego Zoo Institute for Conservation Research, Escondido, California
| | | |
Collapse
|
30
|
Silva MA, Costa GMJ, Lacerda SMSN, Brandão-Dias PFP, Kalapothakis E, Silva Júnior AF, Alvarenga ER, França LR. Successful xenogeneic germ cell transplantation from Jundia catfish (Rhamdia quelen) into adult Nile tilapia (Oreochromis niloticus) testes. Gen Comp Endocrinol 2016; 230-231:48-56. [PMID: 26972155 DOI: 10.1016/j.ygcen.2016.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 01/15/2023]
Abstract
Fish germ cell transplantation presents several important potential applications for aquaculture, including the preservation of germplasm from endangered fish species with high genetic and commercial values. Using this technique in studies developed in our laboratory with adult male Nile tilapias (Oreochromis niloticus), all the necessary procedures were successfully established, allowing the production of functional sperm and healthy progeny approximately 2months after allogeneic transplantation. In the present study, we evaluated the viability of the adult Nile tilapia testis to generate sperm after xenogeneic transplant of germ cells from sexually mature Jundia catfish (Rhamdia quelen) that belong to a different taxonomic order. Therefore, in order to investigate at different time-periods post-transplantation, the presence and development of donor PKH26 labeled catfish germ cells were followed in the tilapia seminiferous tubules. From 7 to 20days post-transplantation, only PKH26 labeled spermatogonia were observed, whereas spermatocytes at different stages of development were found at 70days. Germ cell transplantation success and progression of spermatogenesis were indicated by the presence of labeled PKH26 spermatids and sperm on days 90 and 120 post-transplantation, respectively. Confirming the presence of the catfish genetic material in the tilapia testis, all recipient tilapias evaluated (n=8) showed the genetic markers evaluated. Therefore, we demonstrated for the first time that the adult Nile tilapia testis offers the functional conditions for development of spermatogenesis with sperm production from a fish species belonging to a different order, which provides an important new venue for aquaculture advancement.
Collapse
Affiliation(s)
- M A Silva
- Laboratory of Cellular Biology (Dept. of Morphology), Federal University of Minas Gerais, MG 31270-901, Brazil
| | - G M J Costa
- Laboratory of Cellular Biology (Dept. of Morphology), Federal University of Minas Gerais, MG 31270-901, Brazil
| | - S M S N Lacerda
- Laboratory of Cellular Biology (Dept. of Morphology), Federal University of Minas Gerais, MG 31270-901, Brazil
| | - P F P Brandão-Dias
- Laboratory of Biotechnology and Molecular Markers (Dept. of General Biology), Federal University of Minas Gerais, MG 31270-901, Brazil
| | - E Kalapothakis
- Laboratory of Biotechnology and Molecular Markers (Dept. of General Biology), Federal University of Minas Gerais, MG 31270-901, Brazil
| | - A F Silva Júnior
- Laboratory of Aquaculture (Veterinary School), Federal University of Minas Gerais, MG 31270-901, Brazil
| | - E R Alvarenga
- Laboratory of Aquaculture (Veterinary School), Federal University of Minas Gerais, MG 31270-901, Brazil
| | - L R França
- Laboratory of Cellular Biology (Dept. of Morphology), Federal University of Minas Gerais, MG 31270-901, Brazil; National Institute for Amazonian Research, Manaus, AM 69067-375, Brazil.
| |
Collapse
|