1
|
AbuMadighem A, Rubin E, Arazi E, Lunenfeld E, Huleihel M. Adrenocorticotropic hormone and its receptor as a novel testicular system involves in the development of spermatogenesis. Life Sci 2025; 368:123480. [PMID: 39978588 DOI: 10.1016/j.lfs.2025.123480] [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: 10/25/2024] [Revised: 01/17/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
AIMS To identify functional membrane-associate-specific SSC markers and examine the development of these cells under in vitro conditions. MATERIALS AND METHODS Cells were enzymatically isolated from seminiferous tubules (STs) of immature mice. Spermatogonial cells (Thy1, alpha-6-integrin, and C-KIT) were sorted by FACS. RNA was extracted from these cells for RNAseq analysis. The effect of adrenocorticotropic hormone (ACTH) - the ligand of MC2R- on the development of mouse spermatogonial cells was performed in vitro using a methylcellulose culture system (MCS). Immunofluorescence staining was used to localize MC2R-positive cells in the testes of immature and adult humans and mice and testes of busulfan-treated immature mice. KEY FINDINGS Our RNAseq analysis revealed a high expression of melanocortin receptor 2 (MC2R) in Thy1-positive sorted cells. MC2R-positive cells were localized in the periphery of the STs of humans (prepubertal and adults) and mice at immature and adult ages (normal and busulfan-treated mice). MC2R was doubled stained with PLZF and CDH1 (SSC markers). ACTH was localized in mouse testicular germ cells (pre-meiotic, meiotic, and post-meiotic cells) and somatic cells (Sertoli, Leydig, and peritubular cells). The addition of ACTH to isolated cells from mouse STs in MCS significantly increased the development of pre-meiotic and meiotic/post-meiotic cells in vitro. SIGNIFICANCE We were able to identify, for the first time, a novel membrane-associated and functional SSC marker (MC2R) with relation to ACTH. This marker can be used in future male fertility preservation strategies. Furthermore, we explored a novel testicular system (ACTH system) that regulates the development of spermatogenesis.
Collapse
Affiliation(s)
- Ali AbuMadighem
- The Shraga Segal Dept. Microbiology, Immunology and Genetics, Israel; The Center of Advanced Research and Education in Reproduction (CARER), Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eitan Rubin
- The Shraga Segal Dept. Microbiology, Immunology and Genetics, Israel; The Center of Advanced Research and Education in Reproduction (CARER), Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eden Arazi
- The Shraga Segal Dept. Microbiology, Immunology and Genetics, Israel; The Center of Advanced Research and Education in Reproduction (CARER), Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Mahmoud Huleihel
- The Shraga Segal Dept. Microbiology, Immunology and Genetics, Israel; The Center of Advanced Research and Education in Reproduction (CARER), Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
2
|
Moura AO, Silva TCF, Caetano AR, Kussano NR, Dode MAN, Franco MM. Characterization of DNA methylation profile of the entire CpG island spanning the 5' untranslated region to intron 1 of the Oct4/POU5F1 gene in bovine gametes, embryos, and somatic cells. Anim Genet 2025; 56:e70002. [PMID: 39953930 DOI: 10.1111/age.70002] [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: 07/29/2024] [Revised: 01/14/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
Stem cells are undifferentiated cells that exhibit a bivalent chromatin state that determines their fate. These cells have potential applications in human and animal health and livestock production. Somatic cell nuclear transfer or cloning is currently being used to produce genetically edited animals. A highly differentiated genome is the main obstacle to correcting epigenetic reprogramming by enucleated oocytes during cloning. Activation of pluripotency genes in the somatic genome is a promising strategy to contribute to more efficient epigenetic reprogramming, improving this technique. Recently, epigenome editing has emerged as a new generation of clustered regularly interspaced short palindromic repeats-clustered regularly interspaced short palindromic repeats-associated protein 9 technology with the aim of modifying the cellular epigenome to turn genes on or off without modifying DNA. Here, we characterize the DNA methylation profile of the CpG island spanning the 5' untranslated region to intron 1 of the bovine octamer-binding transcription factor (Oct4) gene in gametes, embryos, and fibroblasts. DNA methylation patterns were categorized into three levels: low (0%-20%), moderate (21%-50%), and high (51%-100%). Sperm and embryos showed a hypomethylation pattern, whereas oocytes exhibited a hypo- to moderate methylation pattern. Fetal and adult skin fibroblasts were hypomethylated and moderately methylated, respectively. These results are essential for future studies aimed at manipulating the expression of Oct4. Thus, epigenome editing can be used to turn on the Oct4 in somatic cells to generate induced pluripotent stem cells. This strategy could potentially convert a fully differentiated cell into a cell with certain degree of pluripotency, facilitating nuclear reprogramming by the enucleated oocyte and improving cloning success rates.
Collapse
Affiliation(s)
- Amanda Oliveira Moura
- School of Veterinary Medicine, Federal University of Uberlândia (FAMEV-UFU), Uberlândia, Minas Gerais, Brazil
- EMBRAPA Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil
| | | | | | | | | | - Maurício Machaim Franco
- School of Veterinary Medicine, Federal University of Uberlândia (FAMEV-UFU), Uberlândia, Minas Gerais, Brazil
- EMBRAPA Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil
| |
Collapse
|
3
|
Wei YL, Fan XJ, Lin XC, Zhang HT, Huang YL, Wang XR. Expression pattern and functional analysis of kinesin-14 KIFC1 in spermatogenesis of Macaca mulatta. Gene 2025; 933:148949. [PMID: 39278374 DOI: 10.1016/j.gene.2024.148949] [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: 04/09/2024] [Revised: 08/15/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
C-terminal kinesin motor KIFC1 is increasingly concerned with an essential role in germ cell development. During the spermatogenesis of mice, rats, and crustaceans, KIFC1 functions in regulating meiotic chromosome separation, acrosome vesicle transportation, and nuclear morphology maintenance. The expression pattern of KIFC1 is conservatively concentrated at the acrosome and nucleus of haploid sperm cells. However, whether KIFC1 has similar functions in non-human primates remains unknown. In this study, we constructed the testis-specific cDNA library and cloned different transcripts of KIFC1 based on the genomic sequence. New variants of KIFC1 were identified, and showed different functional domains from the predicted isoforms. The spatio-temporal expression of KIFC1 proteins in seminiferous tubules of rhesus monkeys showed an obvious nuclear localization, specifically expressed in the spermatocytes and early haploid spermatids. The transcripts of KIFC1 also exhibited considerable expression in the nucleus of rhesus LLC-MK2 cells. Besides, we demonstrated that KIFC1 located at the acrosome and microtubule flagella of the mature sperm, and KIFC1 inhibition resulted in sperm tail deformation as well as increased the instability of head-to-tail connection. In summary, this study filled a gap in the reproductive research of the KIFC1 gene in non-human primates.
Collapse
Affiliation(s)
- Ya-Lan Wei
- Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China
| | - Xiao-Jing Fan
- Medical Research Center, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, Fujian 350011, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China
| | - Xin-Chen Lin
- Medical Research Center, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, Fujian 350011, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China
| | - Hai-Tao Zhang
- Medical Research Center, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, Fujian 350011, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China
| | - Yi-Lan Huang
- Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China.
| | - Xin-Rui Wang
- Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China.
| |
Collapse
|
4
|
Bashiri Z, Hosseini SJ, Salem M, Koruji M. In vivo and in vitro sperm production: an overview of the challenges and advances in male fertility restoration. Clin Exp Reprod Med 2024; 51:171-180. [PMID: 38525520 PMCID: PMC11372308 DOI: 10.5653/cerm.2023.06569] [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: 10/03/2023] [Accepted: 12/14/2023] [Indexed: 03/26/2024] Open
Abstract
Male infertility can be caused by genetic anomalies, endocrine disorders, inflammation, and exposure to toxic chemicals or gonadotoxic treatments. Therefore, several recent studies have concentrated on the preservation and restoration of fertility to enhance the quality of life for affected individuals. It is currently recommended to biobank the tissue extracted from testicular biopsies to provide a later source of spermatogonial stem cells (SSCs). Another successful approach has been the in vitro production of haploid male germ cells. The capacity of SSCs to transform into sperm, as in testicular tissue transplantation, SSC therapy, and in vitro or ex vivo spermatogenesis, makes them ideal candidates for in vivo fertility restoration. The transplantation of SSCs or testicular tissue to regenerate spermatogenesis and create embryos has been achieved in nonhuman mammal species. Although the outcomes of human trials have yet to be released, this method may soon be approved for clinical use in humans. Furthermore, regenerative medicine techniques that develop tissue or cells on organic or synthetic scaffolds enriched with bioactive molecules have also gained traction. All of these methods are now in different stages of experimentation and clinical trials. However, thanks to rigorous studies on the safety and effectiveness of SSC-based reproductive treatments, some of these techniques may be clinically available in upcoming decades.
Collapse
Affiliation(s)
- Zahra Bashiri
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility and Infertility Clinic, Hamedan, Iran
| | - Seyed Jamal Hosseini
- Biomedical Engineering Department, Amirkabir University of Technology, Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Salem
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Liu S, Wu J, Zhao X, Yu M, Taniguchi M, Bao H, Kang K. Recent Progress of Induced Spermatogenesis In Vitro. Int J Mol Sci 2024; 25:8524. [PMID: 39126092 PMCID: PMC11313507 DOI: 10.3390/ijms25158524] [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: 05/29/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Sperm, a crucial gamete for reproduction in sexual reproduction, is generated through the proliferation, differentiation, and morphological transformations of spermatogonial stem cells within the specialized microenvironment of the testes. Replicating this environment artificially presents challenges. However, interdisciplinary advancements in physics, materials science, and cell engineering have facilitated the utilization of innovative materials, technologies, and structures for inducing in vitro sperm production. This article offers a comprehensive overview of research progress on inducing in vitro sperm production by categorizing techniques into two major systems based on matrix-based and non-matrix-based approaches, respectively. Detailed discussions are provided for both types of technology systems through comparisons of their similarities and differences, as well as research advancements. The aim is to provide researchers in this field with a comprehensive panoramic view while presenting our own perspectives and prospects.
Collapse
Affiliation(s)
- Siqi Liu
- College of Coastal Agriculture Science, Guangdong Ocean University, Zhanjiang 524088, China; (S.L.); (J.W.)
| | - Jiang Wu
- College of Coastal Agriculture Science, Guangdong Ocean University, Zhanjiang 524088, China; (S.L.); (J.W.)
| | - Xin Zhao
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
| | - Meng Yu
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
| | - Masayasu Taniguchi
- Department of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-0841, Japan
| | - Huimingda Bao
- College of Coastal Agriculture Science, Guangdong Ocean University, Zhanjiang 524088, China; (S.L.); (J.W.)
| | - Kai Kang
- College of Coastal Agriculture Science, Guangdong Ocean University, Zhanjiang 524088, China; (S.L.); (J.W.)
| |
Collapse
|
6
|
Rahbar M, Asadpour R, Mazaheri Z. The effect of epididymosomes on the development of frozen-thawed mouse spermatogonial stem cells after culture in a decellularized testicular scaffold and transplantation into azoospermic mice. J Assist Reprod Genet 2024; 41:2079-2098. [PMID: 38839698 PMCID: PMC11339233 DOI: 10.1007/s10815-024-03157-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
PURPOSE This study examined SSC proliferation on an epididymosome-enriched decellularized testicular matrix (DTM) hydrogel and spermatogenesis induction in azoospermic mice. METHODS Epididymosomes were extracted and characterized using SEM and western blotting. After cryopreservation, thawed SSCs were cultured in a hydrogel-based three-dimensional (3D) culture containing 10 ng/mL GDNF or 20 µg/mL epididymosomes. SSCs were assessed using the MTT assay, flow cytometry, and qRT-PCR after two weeks of culture. The isolated SSCs were microinjected into the efferent ducts of busulfan-treated mice. DiI-labeled SSCs were followed, and cell homing was assessed after two weeks. After 8 weeks, the testes were evaluated using morphometric studies and immunohistochemistry. RESULTS The expression of PLZF, TGF-β, and miR-10b did not increase statistically significantly in the 3D + GDNF and 3D + epididymosome groups compared to the 3D group. Among the groups, the GDNF-treated group exhibited the highest expression of miR-21 (*P < 0.05). Caspase-3 expression was lower in the epididymosome-treated group than in the other groups (***P < 0.001). Compared to the 3D and negative control groups, the 3D + epididymosomes and 3D + GDNF groups showed an increase in spermatogenic cells. Immunohistochemical results confirmed the growth and differentiation of spermatogonial cells into spermatids in the treatment groups. CONCLUSION The DTM hydrogel containing 20 µg/mL epididymosomes or 10 ng/mL GDNF is a novel and safe culture system that can support SSC proliferation in vitro to obtain adequate SSCs for transplantation success. It could be a novel therapeutic agent that could recover deregulated SSCs in azoospermic patients.
Collapse
Affiliation(s)
- Maryam Rahbar
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Reza Asadpour
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Zohreh Mazaheri
- Basic Medical Science Research Center, Histogenotech Company, Tehran, Iran
| |
Collapse
|
7
|
Asgari F, Asgari H, Najafi M, Hajiaghalou S, Pirhajati-Mahabadi V, Mohammadi A, Gholipourmalekabadi M, Koruji M. In vitro proliferation and differentiation of mouse spermatogonial stem cells in decellularized human placenta matrix. J Biomed Mater Res B Appl Biomater 2024; 112:e35414. [PMID: 38733611 DOI: 10.1002/jbm.b.35414] [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: 04/25/2023] [Revised: 01/15/2024] [Accepted: 02/18/2024] [Indexed: 05/13/2024]
Abstract
Utilizing natural scaffold production derived from extracellular matrix components presents a promising strategy for advancing in vitro spermatogenesis. In this study, we employed decellularized human placental tissue as a scaffold, upon which neonatal mouse spermatogonial cells (SCs) were cultured three-dimensional (3D) configuration. To assess cellular proliferation, we examined the expression of key markers (Id4 and Gfrα1) at both 1 and 14 days into the culture. Our quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis revealed a notable increase in Gfrα1 gene expression, with the 3D culture group exhibiting the highest levels. Furthermore, the relative frequency of Gfrα1-positive cells significantly rose from 38.1% in isolated SCs to 46.13% and 76.93% in the two-dimensional (2D) and 3D culture systems, respectively. Moving forward to days 14 and 35 of the culture period, we evaluated the expression of differentiating markers (Sycp3, acrosin, and Protamine 1). Sycp3 and Prm1 gene expression levels were upregulated in both 2D and 3D cultures, with the 3D group displaying the highest expression. Additionally, acrosin gene expression increased notably within the 3D culture. Notably, at the 35-day mark, the percentage of Prm1-positive cells in the 3D group (36.4%) significantly surpassed that in the 2D group (10.96%). This study suggests that the utilization of placental scaffolds holds significant promise as a bio-scaffold for enhancing mouse in vitro spermatogenesis.
Collapse
Affiliation(s)
- Fatemeh Asgari
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Avicenna Infertility Clinic, Avicenna Research Institute, ACECR, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Asgari
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Hajiaghalou
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | | | - Amirhossein Mohammadi
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Gholipourmalekabadi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Segunda MN, Díaz C, Torres CG, Parraguez VH, De Los Reyes M, Peralta OA. Bovine Peripheral Blood-Derived Mesenchymal Stem Cells (PB-MSCs) and Spermatogonial Stem Cells (SSCs) Display Contrasting Expression Patterns of Pluripotency and Germ Cell Markers under the Effect of Sertoli Cell Conditioned Medium. Animals (Basel) 2024; 14:803. [PMID: 38473188 DOI: 10.3390/ani14050803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
In vitro gamete derivation has been proposed as an interesting strategy for treatment of infertility, improvement of genetic traits, and conservation of endangered animals. Spermatogonial stem cells (SSCs) are primary candidates for in vitro gamete derivation; however, recently, mesenchymal stem cells (MSCs) have also been proposed as candidates for germ cell (GCs) differentiation mainly due to their transdifferentiating capacity. The objective of the present study was to compare the potential for GC differentiation of bovine peripheral blood-derived MSCs (PB-MSCs) and SSCs under the effect of conditioned medium (CM) derived from Sertoli cells (SCs/CM). Samples were collected every 7 days for 21 days and analyzed for pluripotent, GC, and MSC marker expression. The absence of OCT4 and the increased (p < 0.05) expression of NANOG seems to play a role in SSC differentiation, whereas the absence of NANOG and the increased expression (p < 0.05) of OCT4 may be required for PB-MSC differentiation into GCs. SSCs cultured with SCs/CM increased (p < 0.05) the expression of PIWIL2 and DAZL, while PB-MSCs cultured under the same condition only increased (p < 0.05) the expression of DAZL. Overall, the patterns of markers expression suggest that PB-MSCs and SSCs activate different signaling pathways after exposure to SCs/CM and during differentiation into GCs.
Collapse
Affiliation(s)
- Moisés N Segunda
- Faculty of Veterinary and Animal Sciences, University of Chile, Santiago 8820808, Chile
- Doctorate Program of Forestry, Agriculture, and Veterinary Sciences (DCSAV), University of Chile, Santiago 8820808, Chile
- Faculdade de Medicina Veterinária, Universidade José Eduardo dos Santos, Bairro Santo António-Avenida Nuno Alvarez, Huambo 555, Angola
| | - Carlos Díaz
- Doctorate Program in Sciences, UNED, Bravo Murillo 38, 28015 Madrid, Spain
| | - Cristian G Torres
- Faculty of Veterinary and Animal Sciences, University of Chile, Santiago 8820808, Chile
| | - Víctor H Parraguez
- Faculty of Veterinary and Animal Sciences, University of Chile, Santiago 8820808, Chile
| | - Mónica De Los Reyes
- Faculty of Veterinary and Animal Sciences, University of Chile, Santiago 8820808, Chile
| | - Oscar A Peralta
- Faculty of Veterinary and Animal Sciences, University of Chile, Santiago 8820808, Chile
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 7820436, Chile
| |
Collapse
|
9
|
Jorban A, Lunenfeld E, Huleihel M. Effect of Temperature on the Development of Stages of Spermatogenesis and the Functionality of Sertoli Cells In Vitro. Int J Mol Sci 2024; 25:2160. [PMID: 38396838 PMCID: PMC10889116 DOI: 10.3390/ijms25042160] [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: 12/26/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Spermatogenesis is the process of proliferation and differentiation of spermatogonial cells to meiotic and post-meiotic stages and sperm generation. Normal spermatogenesis occurs in vivo at 34 °C to 35 °C, and high temperatures are known to cause male infertility. The aim of the present study was to examine the effect of temperature (35 °C compared to 37 °C) on the viability/apoptosis of developed cells, on the development of different stages of spermatogenesis in 3D in vitro culture conditions, and the functionality of Sertoli cells under these conditions. We used isolated cells from seminiferous tubules of sexually immature mice. The cells were cultured in methylcellulose (as a three-dimensional (3D) in vitro culture system) and incubated in a CO2 incubator at 35 °C or 37 °C. After two to six weeks, the developed cells and organoids were collected and examined for cell viability and apoptosis markers. The development of different stages of spermatogenesis was evaluated by immunofluorescence staining or qPCR analysis using specific antibodies or primers, respectively, for cells at each stage. Factors that indicate the functionality of Sertoli cells were assessed by qPCR analysis. The developed organoids were examined by a confocal microscope. Our results show that the percentages and/or the expression levels of the developed pre-meiotic, meiotic, and post-meiotic cells were significantly higher at 35 °C compared to those at 37 °C, including the expression levels of the androgen receptor, the FSH receptor, transferrin, the androgen-binding protein (ABP), and the glial-derived nerve growth factor (GDNF) which were similarly significantly higher at 35 °C than at 37 °C. The percentages of apoptotic cells (according to acridine orange staining) and the expression levels of BAX, FAS, and CASPAS 3 were significantly higher in cultures incubated at 37 °C compared to those incubated at 35 °C. These findings support the in vivo results regarding the negative effect of high temperatures on the process of spermatogenesis and suggest a possible effect of high temperatures on the viability/apoptosis of spermatogenic cells. In addition, increasing the temperature in vitro also impaired the functionality of Sertoli cells. These findings may deepen our understanding of the mechanisms behind optimal conditions for normal spermatogenesis in vivo and in vitro.
Collapse
Affiliation(s)
- Areej Jorban
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Eitan Lunenfeld
- Adelson School of Medicine, Ariel University, Ariel 4076414, Israel;
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| |
Collapse
|
10
|
Bashiri Z, Movahedin M, Pirhajati V, Asgari H, Koruji M. Ultrastructural study: in vitro and in vivo differentiation of mice spermatogonial stem cells. ZYGOTE 2024; 32:87-95. [PMID: 38149356 DOI: 10.1017/s096719942300062x] [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] [Indexed: 12/28/2023]
Abstract
Mouse testicular tissue is composed of seminiferous tubules and interstitial tissue. Mammalian spermatogenesis is divided into three stages: spermatocytogenesis (mitotic divisions) in which spermatogonial stem cells (SSCs) turn into spermatocytes, followed by two consecutive meiotic divisions in which spermatocytes form spermatids. Spermatids differentiate into spermatozoa during spermiogenesis. Various factors affect the process of spermatogenesis and the organization of cells in the testis. Any disorder in different stages of spermatogenesis will have negative effects on male fertility. The aim of the current study was to compare the in vitro and in vivo spermatogenesis processes before and after transplantation to azoospermic mice using ultrastructural techniques. In this study, mice were irradiated with single doses of 14 Gy 60Co radiation. SSCs isolated from neonatal mice were cultured in vitro for 1 week and were injected into the seminiferous tubule recipient's mice. Testicular cells of neonatal mice were cultured in the four groups on extracellular matrix-based 3D printing scaffolds. The transplanted testes (8 weeks after transplantation) and cultured testicular cells in vitro (after 3 weeks) were then processed for transmission electron microscopy studies. Our study's findings revealed that the morphology and ultrastructure of testicular cells after transplantation and in vitro culture are similar to those of in vivo spermatogenesis, indicating that spermatogenic cell nature is unaltered in vitro.
Collapse
Affiliation(s)
- Zahra Bashiri
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility & Infertility Clinic, Hamedan, Iran
| | - Mansoureh Movahedin
- Department of Anatomical Sciences, Medical Sciences Faculty, Tarbiat Modares University, Tehran, Iran
| | - Vahid Pirhajati
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Asgari
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Bashiri Z, Gholipourmalekabadi M, Khadivi F, Salem M, Afzali A, Cham TC, Koruji M. In vitro spermatogenesis in artificial testis: current knowledge and clinical implications for male infertility. Cell Tissue Res 2023; 394:393-421. [PMID: 37721632 DOI: 10.1007/s00441-023-03824-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 08/14/2023] [Indexed: 09/19/2023]
Abstract
Men's reproductive health exclusively depends on the appropriate maturation of certain germ cells known as sperm. Certain illnesses, such as Klinefelter syndrome, cryptorchidism, and syndrome of androgen insensitivity or absence of testis maturation in men, resulting in the loss of germ cells and the removal of essential genes on the Y chromosome, can cause non-obstructive azoospermia. According to laboratory research, preserving, proliferating, differentiating, and transplanting spermatogonial stem cells or testicular tissue could be future methods for preserving the fertility of children with cancer and men with azoospermia. Therefore, new advances in stem cell research may lead to promising therapies for treating male infertility. The rate of progression and breakthrough in the area of in vitro spermatogenesis is lower than that of SSC transplantation, but newer methods are also being developed. In this regard, tissue and cell culture, supplements, and 3D scaffolds have opened new horizons in the differentiation of stem cells in vitro, which could improve the outcomes of male infertility. Various 3D methods have been developed to produce cellular aggregates and mimic the organization and function of the testis. The production of an artificial reproductive organ that supports SSCs differentiation will certainly be a main step in male infertility treatment.
Collapse
Affiliation(s)
- Zahra Bashiri
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Shahid Hemmat Highway, Tehran, 1449614535, Iran.
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Shahid Hemmat Highway, Tehran, 1449614535, Iran.
- Omid Fertility & Infertility Clinic, Hamedan, Iran.
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farnaz Khadivi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Maryam Salem
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Azita Afzali
- Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Tat-Chuan Cham
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Morteza Koruji
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Shahid Hemmat Highway, Tehran, 1449614535, Iran.
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Shahid Hemmat Highway, Tehran, 1449614535, Iran.
| |
Collapse
|
12
|
Moeinzadeh A, Ashtari B, Garcia H, Koruji M, Velazquez CA, Bagher Z, Barati M, Shabani R, Davachi SM. The Effect of Chitosan/Alginate/Graphene Oxide Nanocomposites on Proliferation of Mouse Spermatogonial Stem Cells. J Funct Biomater 2023; 14:556. [PMID: 38132810 PMCID: PMC10744091 DOI: 10.3390/jfb14120556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
Male survivors of childhood cancer have been known to be afflicted with azoospermia. To combat this, the isolation and purification of spermatogonial stem cells (SSCs) are crucial. Implementing scaffolds that emulate the extracellular matrix environment is vital for promoting the regeneration and proliferation of SSCs. This research aimed to evaluate the efficiency of nanocomposite scaffolds based on alginate, chitosan, and graphene oxide (GO) in facilitating SSCs proliferation. To analyze the cytotoxicity of the scaffolds, an MTT assay was conducted at 1, 3, and 7 days, and the sample containing 30 µg/mL of GO (ALGCS/GO30) exhibited the most favorable results, indicating its optimal performance. The identity of the cells was confirmed using flow cytometry with C-Kit and GFRα1 markers. The scaffolds were subjected to various analyses to characterize their properties. FTIR was employed to assess the chemical structure, XRD to examine crystallinity, and SEM to visualize the morphology of the scaffolds. To evaluate the proliferation of SSCs, qRT-PCR was used. The study's results demonstrated that the ALGCS/GO30 nanocomposite scaffold exhibited biocompatibility and facilitated the attachment and proliferation of SSCs. Notably, the scaffold displayed a significant increase in proliferation markers compared to the control group, indicating its ability to support SSC growth. The expression level of the PLZF protein was assessed using the Immunocytochemistry method. The observations confirmed the qRT-PCR results, which indicated that the nanocomposite scaffolds had higher levels of PLZF protein expression than scaffolds without GO. The biocompatible ALGCS/GO30 is a promising alternative for promoting SSC proliferation in in vitro applications.
Collapse
Affiliation(s)
- Alaa Moeinzadeh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Ashtari
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Heriberto Garcia
- Department of Biology and Chemistry, Texas A&M International University, Laredo, TX 78041, USA
| | - Morteza Koruji
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Carlo Alberto Velazquez
- Department of Biology and Chemistry, Texas A&M International University, Laredo, TX 78041, USA
| | - Zohreh Bagher
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- ENT and Head & Neck Research Center and Department, The Five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Barati
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Davachi
- Department of Biology and Chemistry, Texas A&M International University, Laredo, TX 78041, USA
| |
Collapse
|
13
|
Wu J, Kang K, Liu S, Ma Y, Yu M, Zhao X. Recent Progress of In Vitro 3D Culture of Male Germ Stem Cells. J Funct Biomater 2023; 14:543. [PMID: 37998112 PMCID: PMC10672244 DOI: 10.3390/jfb14110543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023] Open
Abstract
Male germline stem cells (mGSCs), also known as spermatogonial stem cells (SSCs), are the fundamental seed cells of male animal reproductive physiology. However, environmental influences, drugs, and harmful substances often pose challenges to SSCs, such as population reduction and quality decline. With advancements in bioengineering technology and biomaterial technology, an increasing number of novel cell culture methods and techniques have been employed for studying the proliferation and differentiation of SSCs in vitro. This paper provides a review on recent progress in 3D culture techniques for SSCs in vitro; we summarize the microenvironment of SSCs and spermatocyte development, with a focus on scaffold-based culture methods and 3D printing cell culture techniques for SSCs. Additionally, decellularized testicular matrix (DTM) and other biological substrates are utilized through various combinations and approaches to construct an in vitro culture microenvironment suitable for SSC growth. Finally, we present some perspectives on current research trends and potential opportunities within three areas: the 3D printing niche environment, alternative options to DTM utilization, and advancement of the in vitro SSC culture technology system.
Collapse
Affiliation(s)
- Jiang Wu
- Coastal Agricultural College, Guangdong Ocean University, Zhanjiang 524000, China; (J.W.)
| | - Kai Kang
- Coastal Agricultural College, Guangdong Ocean University, Zhanjiang 524000, China; (J.W.)
| | - Siqi Liu
- Coastal Agricultural College, Guangdong Ocean University, Zhanjiang 524000, China; (J.W.)
| | - Yaodan Ma
- Coastal Agricultural College, Guangdong Ocean University, Zhanjiang 524000, China; (J.W.)
| | - Meng Yu
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xin Zhao
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
14
|
Salem M, Khadivi F, Javanbakht P, Mojaverrostami S, Abbasi M, Feizollahi N, Abbasi Y, Heidarian E, Rezaei Yazdi F. Advances of three-dimensional (3D) culture systems for in vitro spermatogenesis. Stem Cell Res Ther 2023; 14:262. [PMID: 37735437 PMCID: PMC10512562 DOI: 10.1186/s13287-023-03466-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 08/22/2023] [Indexed: 09/23/2023] Open
Abstract
The loss of germ cells and spermatogenic failure in non-obstructive azoospermia are believed to be the main causes of male infertility. Laboratory studies have used in vitro testicular models and different 3-dimensional (3D) culture systems for preservation, proliferation and differentiation of spermatogonial stem cells (SSCs) in recent decades. The establishment of testis-like structures would facilitate the study of drug and toxicity screening, pathological mechanisms and in vitro differentiation of SSCs which resulted in possible treatment of male infertility. The different culture systems using cellular aggregation with self-assembling capability, the use of different natural and synthetic biomaterials and various methods for scaffold fabrication provided a suitable 3D niche for testicular cells development. Recently, 3D culture models have noticeably used in research for their architectural and functional similarities to native microenvironment. In this review article, we briefly investigated the recent 3D culture systems that provided a suitable platform for male fertility preservation through organ culture of testis fragments, proliferation and differentiation of SSCs.
Collapse
Affiliation(s)
- Maryam Salem
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Farnaz Khadivi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Parinaz Javanbakht
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Mehdi Abbasi
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Narjes Feizollahi
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Yasaman Abbasi
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Heidarian
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Farzane Rezaei Yazdi
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
15
|
Bashiri Z, Moghaddaszadeh A, Falak R, Khadivi F, Afzali A, Abbasi M, Sharifi AM, Asgari HR, Ghanbari F, Koruji M. Generation of Haploid Spermatids on Silk Fibroin-Alginate-Laminin-Based Porous 3D Scaffolds. Macromol Biosci 2023; 23:e2200574. [PMID: 37116215 DOI: 10.1002/mabi.202200574] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/03/2023] [Indexed: 04/30/2023]
Abstract
In vitro production of sperm is a desirable idea for fertility preservation in azoospermic men and prepubertal boys suffering from cancer. In this study, a biocompatible porous scaffold based on a triad mixture of silk fibroin (SF), alginate (Alg), and laminin (LM) is developed to facilitate the differentiation of mouse spermatogonia stem cells (SSCs). Following SF extraction, the content is analyzed by SDS-PAGE and stable porous 3D scaffolds are successfully prepared by merely Alg, SF, and a combination of Alg-SF, or Alg-SF-LM through freeze-drying. Then, the biomimetic scaffolds are characterized regarding the structural and biological properties, water absorption capacity, biocompatibility, biodegradability, and mechanical behavior. Neonatal mice testicular cells are seeded on three-dimensional scaffolds and their differentiation efficiency is evaluated using real-time PCR, flow cytometry, immunohistochemistry. Blend matrices showed uniform porous microstructures with interconnected networks, which maintained long-term stability and mechanical properties better than homogenous structures. Molecular analysis of the cells after 21 days of culture showed that the expression of differentiation-related proteins in cells that are developed in composite scaffolds is significantly higher than in other groups. The application of a composite system can lead to the differentiation of SSCs, paving the way for a novel infertility treatment landscape in the future.
Collapse
Affiliation(s)
- Zahra Bashiri
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Omid Fertility & Infertility Clinic, Hamedan, 6516796198, Iran
| | - Ali Moghaddaszadeh
- Departement of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran
| | - Reza Falak
- Immunology Research Center (IRC), Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Farnaz Khadivi
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, 8815713471, Iran
| | - Azita Afzali
- Hajar hospital, Shahrekord University of Medical Sciences, Shahrekord, 8816854633, Iran
| | - Mehdi Abbasi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, 1417653761, Iran
| | - Ali Mohammad Sharifi
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Tissue Engineering Group (NOCERAL), Department of Orthopedics Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Hamid Reza Asgari
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Farid Ghanbari
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Morteza Koruji
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| |
Collapse
|
16
|
Chen L, Dong Z, Chen X. Fertility preservation in pediatric healthcare: a review. Front Endocrinol (Lausanne) 2023; 14:1147898. [PMID: 37206440 PMCID: PMC10189781 DOI: 10.3389/fendo.2023.1147898] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Survival rates for children and adolescents diagnosed with malignancy have been steadily increasing due to advances in oncology treatments. These treatments can have a toxic effect on the gonads. Currently, oocyte and sperm cryopreservation are recognized as well-established and successful strategies for fertility preservation for pubertal patients, while the use of gonadotropin-releasing hormone agonists for ovarian protection is controversial. For prepubertal girls, ovarian tissue cryopreservation is the sole option. However, the endocrinological and reproductive outcomes after ovarian tissue transplantation are highly heterogeneous. On the other hand, immature testicular tissue cryopreservation remains the only alternative for prepubertal boys, yet it is still experimental. Although there are several published guidelines for navigating fertility preservation for pediatric and adolescent patients as well as transgender populations, it is still restricted in clinical practice. This review aims to discuss the indications and clinical outcomes of fertility preservation. We also discuss the probably effective and efficient workflow to facilitate fertility preservation.
Collapse
Affiliation(s)
- Lin Chen
- Reproductive Medical Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zirui Dong
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
- The Fertility Preservation Research Center, Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
Jabari A, Gholami K, Khadivi F, Koruji M, Amidi F, Gilani MAS, Mahabadi VP, Nikmahzar A, Salem M, Movassagh SA, Feizollahi N, Abbasi M. In vitro complete differentiation of human spermatogonial stem cells to morphologic spermatozoa using a hybrid hydrogel of agarose and laminin. Int J Biol Macromol 2023; 235:123801. [PMID: 36842740 DOI: 10.1016/j.ijbiomac.2023.123801] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/08/2023] [Accepted: 02/18/2023] [Indexed: 02/28/2023]
Abstract
Spermatogenesis refers to the differentiation of the spermatogonial stem cells (SSCs) located in the base seminiferous tubules into haploid spermatozoa. Prerequisites for in vitro spermatogenesis include an extracellular matrix (ECM), paracrine factors, and testicular somatic cells which play a supporting role for SSCs. Thus, the present study evaluated the potential of co-culturing Sertoli cells and SSCs embedded in a hybrid hydrogel of agarose and laminin, the main components of the ECM. Following the three-week conventional culture of human testicular cells, the cells were cultured in agarose hydrogel or agarose/laminin one (hybrid) for 74 days. Then, immunocytochemistry, real-time PCR, electron microscopy, and morphological staining methods were applied to analyze the presence of SSCs, as well as the other cells of the different stages of spermatogenesis. Based on the results, the colonies with positive spermatogenesis markers were observed in both culture systems. The existence of the cells of all three phases of spermatogenesis (spermatogonia, meiosis, and spermiogenesis) was confirmed in the two groups, while morphological spermatozoa were detected only in the hybrid hydrogel group. Finally, a biologically improved 3D matrix can support all the physiological activities of SSCs such as survival, proliferation, and differentiation.
Collapse
Affiliation(s)
- Ayob Jabari
- Department of Obstetrics and Gynecology, Molud Infertility Center, Zahedan University of Medical Sciences, Zahedan, Iran; Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Science in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Keykavos Gholami
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Khadivi
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Morteza Koruji
- Cellular and Molecular Research Center & Department of Anatomical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fardin Amidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Aghbibi Nikmahzar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Salem
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Narjes Feizollahi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Abbasi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Segunda MN, Díaz C, Torres CG, Parraguez VH, De los Reyes M, Peralta OA. Comparative Analysis of the Potential for Germ Cell (GC) Differentiation of Bovine Peripheral Blood Derived-Mesenchymal Stem Cells (PB-MSC) and Spermatogonial Stem Cells (SSC) in Co-Culture System with Sertoli Cells (SC). Animals (Basel) 2023; 13:ani13020318. [PMID: 36670859 PMCID: PMC9854759 DOI: 10.3390/ani13020318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/28/2022] [Accepted: 12/31/2022] [Indexed: 01/18/2023] Open
Abstract
Although spermatogonial stem cells (SSC) constitute primary candidates for in vitro germ cell (GC) derivation, they are scarce and difficult to maintain in an undifferentiated state. Alternatively, mesenchymal stem cells (MSC) are also candidates for GC derivation due to their simplicity for culture and multipotential for transdifferentiation. The aim of the present study was to compare the GC differentiation potentials of bull peripheral blood-derived MSC (PB-MSC) and SSC using an in vitro 3D co-culture system with Sertoli cells (SC). Samples of PB-MSC or SSC co-cultures with SC were collected on days 0, 7, 14 and 21 and analyzed for pluripotency, GC and mesenchymal marker expression. Co-culture of PB-MSC+SC resulted in down-regulation of NANOG and up-regulation of OCT4 at day 7. In comparison, co-culture of SSC+SC resulted in consistent expression of NANOG, OCT4 and SOX2 at day 14. During co-culture, SSC+SC increased the expression of DAZL, PIWIL2, FRAGILIS and STELLA and activated the expression of STRA8, whereas co-culture of PB-MSC+SC only increased the expression of DAZL and PIWIL2. Thus, co-culture of bull PB-MSC+SC and SSC+SC in 3D SACS results in differential expression of pluripotency and GC markers, where bull SSC display a more robust GC differentiation profile compared to PB-MSC.
Collapse
Affiliation(s)
- Moisés N. Segunda
- Department of Animal Production Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, Santiago 8820808, Chile
- Doctorate Program of Forestry, Agriculture, and Veterinary Sciences (DCSAV), University of Chile, Santa Rosa 11315, Santiago 8820808, Chile
- Faculdade de Medicina Veterinária, Universidade José Eduardo dos Santos, Bairro Santo António-Avenida Nuno Alvarez, Huambo 555, Angola
| | - Carlos Díaz
- Doctorate Program in Sciences, UNED, Bravo Murillo 38, 28015 Madrid, Spain
| | - Cristian G. Torres
- Department of Clinical Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, Santiago 8820808, Chile
| | - Víctor H. Parraguez
- Department of Biological Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, Santiago 8820808, Chile
| | - Mónica De los Reyes
- Department of Animal Production Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, Santiago 8820808, Chile
| | - Oscar A. Peralta
- Department of Animal Production Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, Santiago 8820808, Chile
- Correspondence:
| |
Collapse
|
19
|
Rahbar M, Asadpour R, Azami M, Mazaheri Z, Hamali H. Improving the process of spermatogenesis in azoospermic mice using spermatogonial stem cells co-cultured with epididymosomes in three-dimensional culture system. Life Sci 2022; 310:121057. [DOI: 10.1016/j.lfs.2022.121057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/09/2022]
|
20
|
Abofoul‐Azab M, Lunenfeld E, Kleiman S, Barda S, Hauser R, Huleihel M. Determining the expression levels of CSF-1 and OCT4, CREM-1, and protamine in testicular biopsies of adult Klinefelter patients: Their possible correlation with spermatogenesis. Andrologia 2022; 54:e14558. [PMID: 36177809 PMCID: PMC9786270 DOI: 10.1111/and.14558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 06/24/2022] [Accepted: 08/01/2022] [Indexed: 12/30/2022] Open
Abstract
Klinefelter syndrome (KS) is the most prevalent genetic disorder of infertile males. This study aimed to determine in Klinefelter patients (KS) the expression levels of spermatogenic markers and testicular growth factors that might predict spermatogenesis based on conventional testicular sperm extraction (TESE). The expression levels of the pre-meiotic (OCT4, CD9, GFR-α1, α-6-INTEGRIN, SALL4, C-KIT), meiotic (CREM-1), and post-meiotic (protamine) markers, as well as the colony stimulating factor-1 (CSF-1) were examined in testicular biopsies with and without mature sperm of KS and normal karyotype of azoospermic patients (AZO) with complete spermatogenesis. In the biopsies of AZO, the expression levels (fold of expression compared to the PPI of the same sample) of OCT4 were 9.68± 7.93, CREM 42.78± 28.22, CSF-1 3.07 ± 3.19, and protamine 78498.12 ± 73214.40. Biopsies from KS included 7 with sperm and 17 without sperm. Among the biopsies with sperm, the expression levels of OCT4 were 7.27± 9.29, CREM 3.13± 7.89, CSF-1 35.5 ± 48.01, and protamine 902.97 ± 2365.92. In 14 biopsies without sperm, we found low expression levels of OCT4, CREM and CSF-1, and no expression of protamine. However, in three of the biopsies without sperm that highly expressed OCT4 and CSF-1, the expression levels of CREM-1 and protamine were high. These results may be used for further consulting with patients considering repeating conventional TESE or micro TESE and cryopreservation for possible future in-vitro spermatogenesis.
Collapse
Affiliation(s)
- Maram Abofoul‐Azab
- The Shraga Segal Department of Microbiology, Immunology, and GeneticsBen‐Gurion University of the NegevBeer‐ShevaIsrael,The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael,Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | | | - Sandra Kleiman
- Male Fertility Clinic and Sperm BankLis Maternity HospitalTel AvivIsrael,Sourasky Medical CenterTel‐AvivIsrael,Sackler School of Medicine Tel Aviv UniversityTel AvivIsrael
| | - Shimi Barda
- Male Fertility Clinic and Sperm BankLis Maternity HospitalTel AvivIsrael
| | - Ron Hauser
- Male Fertility Clinic and Sperm BankLis Maternity HospitalTel AvivIsrael,Sourasky Medical CenterTel‐AvivIsrael,Sackler School of Medicine Tel Aviv UniversityTel AvivIsrael
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology, and GeneticsBen‐Gurion University of the NegevBeer‐ShevaIsrael,The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael,Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| |
Collapse
|
21
|
Noghani AE, Asadpour R, Saberivand A, Mazaheri Z, Rodriguez-Wallberg KA, Hamidian G. Differentiation of neonate mouse spermatogonia on two-dimensional and three-dimensional culture systems supplemented with d-Serine and Dizocilpine (MK-801). Theriogenology 2022; 191:168-178. [PMID: 35998400 DOI: 10.1016/j.theriogenology.2022.08.004] [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: 01/22/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022]
Abstract
N-methyl-d-aspartate (NMDA) modulates the spermatogenesis process through stimulating the steroid hormone biosynthesis. The aim of this study was to evaluate the effects of NMDA receptors agonists (d-Serine) and antagonists (MK801) on spermatogonia differentiation on decellularization testicular matrix (DTM) hydrogel scaffold. Four treatment groups were planned: 2D + D-Serine, 3D + D-Serine, 2D + MK801, and 3D + MK801. Results showed that cell viability was significantly decreased after 48 h in the 3D + D-Serine group and after 24 and 48 h in the 3D + MK801 group compared to the controls. The spermatogonia proliferation after two, four, and eight weeks was significantly increased in the 3D + D-Serine culture, while it was significantly reduced in the 2D + MK801 and 3D + MK801 groups after four and eight weeks. Real-time PCR results demonstrated that pre-meiotic gene (Plzf) expression was significantly increased only in the 3D + D-Serine culture compared to the control groups after four weeks of culture. The meiotic gene (Sycp3) expression was significantly increased in the 2D + D-Serine and 3D + D-Serine compared to the 2D controls after four and eight weeks. The post-meiotic gene (Tnp1) level in the 3D + D-Serine was significantly higher than the other groups. Flow-cytometry results indicated that the protein expression of Plzf (after four and eight weeks), Sycp3 (after eight weeks), and Tnp1 (after eight weeks) in the d-Serine-treated groups was significantly increased compared with the 2D control groups. There were not any significant changes in the gene expression of spermatogenic-related markers in MK801 culture media. However, a significant decrease in the protein levels of Plzf after eight weeks and Sycp3 after four and eight weeks was observed. In conclusion, the addition of NMDARs agonists (d-Serine) could be used to regulate the differentiation of spermatogonia in the 3D culture system.
Collapse
Affiliation(s)
- Amirhessam Eskafi Noghani
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Reza Asadpour
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Adel Saberivand
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Zohreh Mazaheri
- Basic Medical Science Research Center, Histogenotech Company, Tehran, Iran.
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Novumhuset Plan 4, SE-141 86, Stockholm, Sweden.
| | - Gholamreza Hamidian
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
22
|
Lawlor M, Zigo M, Kerns K, Cho IK, Easley IV CA, Sutovsky P. Spermatozoan Metabolism as a Non-Traditional Model for the Study of Huntington’s Disease. Int J Mol Sci 2022; 23:ijms23137163. [PMID: 35806166 PMCID: PMC9266437 DOI: 10.3390/ijms23137163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Huntington’s Disease (HD) is a fatal autosomal dominant neurodegenerative disease manifested through motor dysfunction and cognitive deficits. Decreased fertility is also observed in HD animal models and HD male patients, due to altered spermatogenesis and sperm function, thus resulting in reduced fertilization potential. Although some pharmaceuticals are currently utilized to mitigate HD symptoms, an effective treatment that remedies the pathogenesis of the disease is yet to be approved by the FDA. Identification of genes and relevant diagnostic biomarkers and therapeutic target pathways including glycolysis and mitochondrial complex-I-dependent respiration may be advantageous for early diagnosis, management, and treatment of the disease. This review addresses the HD pathway in neuronal and sperm metabolism, including relevant gene and protein expression in both neurons and spermatozoa, indicated in the pathogenesis of HD. Furthermore, zinc-containing and zinc-interacting proteins regulate and/or are regulated by zinc ion homeostasis in both neurons and spermatozoa. Therefore, this review also aims to explore the comparative role of zinc in both neuronal and sperm function. Ongoing studies aim to characterize the products of genes implicated in HD pathogenesis that are expressed in both neurons and spermatozoa to facilitate studies of future treatment avenues in HD and HD-related male infertility. The emerging link between zinc homeostasis and the HD pathway could lead to new treatments and diagnostic methods linking genetic sperm defects with somatic comorbidities.
Collapse
Affiliation(s)
- Meghan Lawlor
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA; (M.L.); (M.Z.); (K.K.)
| | - Michal Zigo
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA; (M.L.); (M.Z.); (K.K.)
| | - Karl Kerns
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA; (M.L.); (M.Z.); (K.K.)
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - In Ki Cho
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA; (I.K.C.); (C.A.E.IV)
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Charles A. Easley IV
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA; (I.K.C.); (C.A.E.IV)
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Peter Sutovsky
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA; (M.L.); (M.Z.); (K.K.)
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO 65211, USA
- Correspondence: ; Tel.: +1-(573)-882-3329
| |
Collapse
|
23
|
Understanding the Underlying Molecular Mechanisms of Meiotic Arrest during In Vitro Spermatogenesis in Rat Prepubertal Testicular Tissue. Int J Mol Sci 2022; 23:ijms23115893. [PMID: 35682573 PMCID: PMC9180380 DOI: 10.3390/ijms23115893] [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: 04/21/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 12/10/2022] Open
Abstract
In vitro spermatogenesis appears to be a promising approach to restore the fertility of childhood cancer survivors. The rat model has proven to be challenging, since germ cell maturation is arrested in organotypic cultures. Here, we report that, despite a meiotic entry, abnormal synaptonemal complexes were found in spermatocytes, and in vitro matured rat prepubertal testicular tissues displayed an immature phenotype. RNA-sequencing analyses highlighted up to 600 differentially expressed genes between in vitro and in vivo conditions, including genes involved in blood-testis barrier (BTB) formation and steroidogenesis. BTB integrity, the expression of two steroidogenic enzymes, and androgen receptors were indeed altered in vitro. Moreover, most of the top 10 predicted upstream regulators of deregulated genes were involved in inflammatory processes or immune cell recruitment. However, none of the three anti-inflammatory molecules tested in this study promoted meiotic progression. By analysing for the first time in vitro matured rat prepubertal testicular tissues at the molecular level, we uncovered the deregulation of several genes and revealed that defective BTB function, altered steroidogenic pathway, and probably inflammation, could be at the origin of meiotic arrest.
Collapse
|
24
|
Shuchat S, Yossifon G, Huleihel M. Perfusion in Organ-on-Chip Models and Its Applicability to the Replication of Spermatogenesis In Vitro. Int J Mol Sci 2022; 23:5402. [PMID: 35628214 PMCID: PMC9141186 DOI: 10.3390/ijms23105402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
Organ/organoid-on-a-chip (OoC) technologies aim to replicate aspects of the in vivo environment in vitro, at the scale of microns. Mimicking the spatial in vivo structure is important and can provide a deeper understanding of the cell-cell interactions and the mechanisms that lead to normal/abnormal function of a given organ. It is also important for disease models and drug/toxin testing. Incorporating active fluid flow in chip models enables many more possibilities. Active flow can provide physical cues, improve intercellular communication, and allow for the dynamic control of the environment, by enabling the efficient introduction of biological factors, drugs, or toxins. All of this is in addition to the fundamental role of flow in supplying nutrition and removing waste metabolites. This review presents an overview of the different types of fluid flow and how they are incorporated in various OoC models. The review then describes various methods and techniques of incorporating perfusion networks into OoC models, including self-assembly, bioprinting techniques, and utilizing sacrificial gels. The second part of the review focuses on the replication of spermatogenesis in vitro; the complex process whereby spermatogonial stem cells differentiate into mature sperm. A general overview is given of the various approaches that have been used. The few studies that incorporated microfluidics or vasculature are also described. Finally, a future perspective is given on elements from perfusion-based models that are currently used in models of other organs and can be applied to the field of in vitro spermatogenesis. For example, adopting tubular blood vessel models to mimic the morphology of the seminiferous tubules and incorporating vasculature in testis-on-a-chip models. Improving these models would improve our understanding of the process of spermatogenesis. It may also potentially provide novel therapeutic strategies for pre-pubertal cancer patients who need aggressive chemotherapy that can render them sterile, as well asfor a subset of non-obstructive azoospermic patients with maturation arrest, whose testes do not produce sperm but still contain some of the progenitor cells.
Collapse
Affiliation(s)
- Sholom Shuchat
- Faculty of Mechanical Engineering, Technion–Israel Institute of Technology, Haifa 3200003, Israel; (S.S.); (G.Y.)
| | - Gilad Yossifon
- Faculty of Mechanical Engineering, Technion–Israel Institute of Technology, Haifa 3200003, Israel; (S.S.); (G.Y.)
- School of Mechanical Engineering, University of Tel Aviv, Tel Aviv 6997801, Israel
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
- The Center of Advanced Research and Education in Reproduction (CARER), Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| |
Collapse
|
25
|
Robinson M, Bedford E, Witherspoon L, Willerth SM, Flannigan R. Using clinically derived human tissue to 3-dimensionally bioprint personalized testicular tubules for in vitro culturing: first report. F&S SCIENCE 2022; 3:130-139. [PMID: 35560010 DOI: 10.1016/j.xfss.2022.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To study the feasibility and spermatogenic potential of 3-dimensional (3D) bioprinting personalized human testicular cells derived from a patient with nonobstructive azoospermia (NOA). DESIGN A human testicular biopsy from a single donor with NOA was dissociated into single cells, expanded in vitro, and 3D bioprinted into tubular structures akin to the seminiferous tubule using AGC-10 bioink and an RX1 bioprinter with a CENTRA coaxial microfluidic printhead from Aspect Biosystems. Three-dimensional organoid cultures were used as a nonbioprinted in vitro control. SETTING Academic medical center. PATIENT(S) A 31-year-old man with NOA with testis biopsy demonstrating Sertoli cell-only syndrome. INTERVENTION(S) Three-dimensional bioprinting and in vitro culturing of patient-derived testis cells. MAIN OUTCOME MEASURE(S) Cellular viability after printing was determined, along with the expression of phenotypic and spermatogenic functional genetic markers after 12 days of in vitro culture. RESULT(S) Testicular cultures were expandable in vitro and generated sufficiently large numbers for 3D bioprinting at 35 million cells per mL of bioink. Viability 24 hours after printing was determined to be 93.4% ± 2.4%. Immunofluorescence staining for the phenotype markers SRY-Box transcription factor 9, insulin-like 3, actin alpha 2 smooth muscle, and synaptonemal complex protein 3 after 12 days was positive, confirming the presence of Sertoli, Leydig, peritubular myoid, and meiotic germ cells. Reverse transcription qualitative polymerase chain reaction analysis showed that after 12 days in spermatogenic media, the bioprints substantially up-regulated spermatogenic gene expression on par with nonbioprinted controls and showed a particularly significant improvement in genes involved in spermatogonial stem cell maintenance: inhibitor of deoxyribonucleic acid binding 4 by 365-fold; fibroblast growth factor 3 by 94,152-fold; stem cell growth factor receptor KIT by twofold; stimulated by retinoic acid 8 by 125-fold; deleted in azoospermia-like by 114-fold; synaptonemal complex protein 3 by sevenfold; zona pellucida binding protein by twofold; transition protein 1 by 2,908-fold; and protamine 2 by 11-fold. CONCLUSION(S) This study demonstrates for the first time the feasibility of 3D bioprinting adult human testicular cells. We show that the bioprinting process is compatible with high testicular cell viability and without loss of the main somatic phenotypes within the testis tissue. We demonstrate an increase in germ cell markers in the 3D bioprinted tubules after 12 days of in vitro culture. This platform may carry future potential for disease modeling and regenerative opportunities in a personalized medicine framework.
Collapse
Affiliation(s)
- Meghan Robinson
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Erin Bedford
- Aspect Biosystems, Vancouver, British Columbia, Canada
| | - Luke Witherspoon
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Department of Urology, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Stephanie M Willerth
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ryan Flannigan
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Department of Urology, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
26
|
Tran KTD, Valli-Pulaski H, Colvin A, Orwig KE. Male fertility preservation and restoration strategies for patients undergoing gonadotoxic therapies†. Biol Reprod 2022; 107:382-405. [PMID: 35403667 PMCID: PMC9382377 DOI: 10.1093/biolre/ioac072] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 11/22/2022] Open
Abstract
Medical treatments for cancers or other conditions can lead to permanent infertility. Infertility is an insidious disease that impacts not only the ability to have a biological child but also the emotional well-being of the infertile individuals, relationships, finances, and overall health. Therefore, all patients should be educated about the effects of their medical treatments on future fertility and about fertility preservation options. The standard fertility preservation option for adolescent and adult men is sperm cryopreservation. Sperms can be frozen and stored for a long period, thawed at a later date, and used to achieve pregnancy with existing assisted reproductive technologies. However, sperm cryopreservation is not applicable for prepubertal patients who do not yet produce sperm. The only fertility preservation option available to prepubertal boys is testicular tissue cryopreservation. Next-generation technologies are being developed to mature those testicular cells or tissues to produce fertilization-competent sperms. When sperm and testicular tissues are not available for fertility preservation, inducing pluripotent stem cells derived from somatic cells, such as blood or skin, may provide an alternative path to produce sperms through a process call in vitro gametogenesis. This review describes standard and experimental options to preserve male fertility as well as the experimental options to produce functional spermatids or sperms from immature cryopreserved testicular tissues or somatic cells.
Collapse
Affiliation(s)
- Kien T D Tran
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Hanna Valli-Pulaski
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Amanda Colvin
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Kyle E Orwig
- Correspondence: Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA 15213, USA. Tel: 412-641-2460; E-mail:
| |
Collapse
|
27
|
Zhang W, Nie R, Cai Y, Xie W, Zou K. Progress in germline stem cell transplantation in mammals and the potential usage. Reprod Biol Endocrinol 2022; 20:59. [PMID: 35361229 PMCID: PMC8969385 DOI: 10.1186/s12958-022-00930-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/10/2022] [Indexed: 11/10/2022] Open
Abstract
Germline stem cells (GSCs) are germ cells with the capacities of self-renewal and differentiation into functional gametes, and are able to migrate to their niche and reconstitute the fertility of recipients after transplantation. Therefore, GSCs transplantation is a promising technique for fertility recovery in the clinic, protection of rare animals and livestock breeding. Though this novel technique faces tremendous challenges, numerous achievements have been made after several decades' endeavor. This review summarizes the current knowledge of GSCs transplantation and its utilization in mammals, and discusses the application prospect in reproductive medicine and animal science.
Collapse
Affiliation(s)
- Wen Zhang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ruotian Nie
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yihui Cai
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wenhai Xie
- School of Life Sciences, Shandong University of Technology, NO. 266 Xincun Road, Zibo, 255000, Shandong, China.
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
28
|
AbuMadighem A, Shuchat S, Lunenfeld E, Yossifon G, Huleihel M. Testis on a chip - a microfluidic 3-dimensional culture system for the development of spermatogenesis in-vitro. Biofabrication 2022; 14. [PMID: 35334473 DOI: 10.1088/1758-5090/ac6126] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/25/2022] [Indexed: 11/12/2022]
Abstract
This research presents a novel Testis-on-a Chip- platform. Testicular cells are enzymatically isolated from the seminiferous tubules of sexually immature mice, seeded in a methylcellulose gel and cultured in a microfluidic chip. The unique design sandwiches the soft methylcellulose between stiffer agar support gels. The cells develop into organoids continuing to proliferate and differentiate. After seven weeks of culture the cells have over 95% viability. Confocal microscopy of the developed organoids reveals a structure containing the various stages of spermatogenesis up to and including meiosis II: premeiotic, meiotic and post-meiotic germ cells. The organoid structure also contains the supporting Sertoli and peritubular cells. The responsiveness of the system to the addition of testosterone and retinoic acid to the culture medium during the experiment are also investigated. As a benchmark, the Testis-on-a-Chip is compared to a conventional three-dimensional methylcellulose cell culture in a well plate. Analysis via FACS (Fluorescence-activated cell sorting) shows more haploid cells in the chip as compared to the plates. Immunofluorescence staining after seven weeks of culture shows more differentiated cells in the chip as compared to the well plate. This demonstrates the feasibility of our platform as well as its advantages. This research opens new horizons for the study and realization of spermatogenesis in-vitro. It can also enable the implementation of microfluidic technologies in future therapeutic strategies for pre pubertal male fertility preservation and adults with maturation arrest. Lastly, it can serve as a platform for drug and toxin testing.
Collapse
Affiliation(s)
- Ali AbuMadighem
- Ben-Gurion University of the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Southern, 84105, ISRAEL
| | - Sholom Shuchat
- Technion Israel Institute of Technology, Faculty of Mechanical Engineering, Haifa, Haifa, 3200003, ISRAEL
| | - Eitan Lunenfeld
- Ben-Gurion University of the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Beer-Sheva, Southern, 84105, ISRAEL
| | - Gilad Yossifon
- Technion Israel Institute of Technology, Faculty of Mechanical Engineering, Haifa, Haifa, 3200003, ISRAEL
| | - Mahmoud Huleihel
- Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, 84105, ISRAEL
| |
Collapse
|
29
|
Noghani AE, Asadpour R, Saberivand A, Mazaheri Z, Hamidian G. Effect of NMDA receptor agonist and antagonist on spermatogonial stem cells proliferation in 2- and 3- dimensional culture systems. Mol Biol Rep 2022; 49:2197-2207. [PMID: 35000063 DOI: 10.1007/s11033-021-07041-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/01/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The main purpose of this study was to investigate the effect of D-serine (DS) and Dizocilpine (MK-801) on the proliferation of spermatogonial stem cells (SSCs) in two-dimensional (2D) and three-dimensional (3D) culture systems. METHODS AND RESULTS The SSCs of male NMRI mice were isolated by enzymatic digestion and cultured for two weeks. Then, the identity of SSCs was validated by anti-Plzf and anti-GFR-α1 antibodies via immunocytochemistry (ICC). The proliferation capacity of SSCs was evaluated by their culture on a layer of the decellularized testicular matrix (DTM) prepared from mouse testis, as well as two-dimensional (2D) with different mediums. After two weeks of the initiation of proliferation culture on 3D and 2D medium, the pre-meiotic at the mRNA and protein levels were evaluated via qRT-PCR and flow cytometry methods, respectively. The results showed that the proliferation rate of SSCs in 3D culture with 50 mM glutamic acid and 20 mM D-serine was significantly different from other groups after 14 days treatment. mRNA expression levels of promyelocytic leukemia zinc finger (Plzf) in 3D cultures supplemented by 20 mM D-serine and 50 mM glutamic acid were considerably higher than the 3D control group (p < 0.001). The flow cytometry analysis revealed that the amount of Plzf in the 2D-culture groups of SSCs with 20 mM MK-801 was considerably lower compared to the 2D-culture control group (p < 0.001). CONCLUSIONS This study indicated that decellularized testicular matrix supplemented with D-serine and glutamic acid could be considered a promising vehicle to support cells and provide an appropriate niche for the proliferation of SSCs.
Collapse
Affiliation(s)
| | - Reza Asadpour
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Adel Saberivand
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Zohreh Mazaheri
- Basic Medical Science Research Center, Histogenotech Company, Tehran, Iran
| | - Gholamreza Hamidian
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
30
|
Gholami K, Solhjoo S, Aghamir SMK. Application of Tissue-Specific Extracellular Matrix in Tissue Engineering: Focus on Male Fertility Preservation. Reprod Sci 2022; 29:3091-3099. [PMID: 35028926 DOI: 10.1007/s43032-021-00823-9] [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: 07/01/2021] [Accepted: 12/03/2021] [Indexed: 11/28/2022]
Abstract
In vitro spermatogenesis and xenotransplantation of the immature testicular tissues (ITT) are the experimental approaches that have been developed for creating seminiferous tubules-like functional structures in vitro and keeping the integrity of the ITTs in vivo, respectively. These strategies are rapidly developing in response to the growing prevalence of infertility in adolescent boys undergoing cancer treatment, by the logic that there is no sperm cryopreservation option for them. Recently, with the advances made in the field of tissue engineering and biomaterials, these methods have achieved promising results for fertility preservation. Due to the importance of extracellular matrix for the formation of vascular bed around the grafted ITTs and also the creation of spatial arrangements between Sertoli cells and germ cells, today it is clear that the scaffold plays a very important role in the success of these methods. Decellularized extracellular matrix (dECM) as a biocompatible, functionally graded, and biodegradable scaffold with having tissue-specific components and growth factors can support reorganization and physiologic processes of originated cells. This review discusses the common protocols for the tissue decellularization, sterilization, and hydrogel formation of the decellularized and lyophilized tissues as well as in vitro and in vivo studies on the use of the testis-derived dECM for testicular organoids.
Collapse
Affiliation(s)
- Keykavos Gholami
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Solhjoo
- Department of Anatomy, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
31
|
Hu X, Wang H, Tian GG, Hou C, Xu B, Zhao X, Zhao Y, Fang Q, Li X, He L, Chen X, Li S, Wu J. Offspring production of haploid spermatid-like cells derived from mouse female germline stem cells with chromatin condensation. Cell Biosci 2022; 12:5. [PMID: 34983631 PMCID: PMC8729121 DOI: 10.1186/s13578-021-00697-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/06/2021] [Indexed: 12/04/2022] Open
Abstract
Background During male meiosis, the Y chromosome can form perfect pairing with the X chromosome. However, it is unclear whether mammalian Female germline stem cells (FGSCs) without a Y chromosome can transdifferentiate into functional haploid spermatid-like cells (SLCs). Results We found that spermatogenesis was restarted by transplanting FGSCs into Kitw/wv mutant testes. Complete meiosis and formation of SLCs was induced in vitro by testicular cells of Kitw/wv mutant mice, cytokines and retinoic acid. Healthy offspring were produced by sperm and SLCs derived from the in vivo and in vitro transdifferentiation of FGSCs, respectively. Furthermore, high-throughput chromosome conformation capture sequencing(Hi-C-seq) and “bivalent” (H3K4me3-H3K27me3) micro chromatin immunoprecipitation sequencing (μChIP-seq) experiments showed that stimulated by retinoic acid gene 8 (STRA8)/protamine 1 (PRM1)-positive transdifferentiated germ cells (tGCs) and male germ cells (mGCs) display similar chromatin dynamics and chromatin condensation during in vitro spermatogenesis. Conclusion This study demonstrates that sperm can be produced from FGSCs without a Y chromosome. This suggests a strategy for dairy cattle breeding to produce only female offspring with a high-quality genetic background. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00697-z.
Collapse
Affiliation(s)
- Xiaopeng Hu
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Hu Wang
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Geng G Tian
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Changliang Hou
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Bo Xu
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Xinyan Zhao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Yongqiang Zhao
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Qian Fang
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Xinyue Li
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Lin He
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Xuejin Chen
- Department of Laboratory Animal Science, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Shangang Li
- State Key Laboratory of Primate Biomedicine Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China. .,Yunnan Key Laboratory of Primate Biomedicine Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Ji Wu
- Bio-X Institutes, Shanghai Jiao Tong University, No. 800. Dongchuan Road, Minhang District, Shanghai, 200240, China. .,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China. .,Shanghai Key Laboratory of Reproductive Medicine, Shanghai, 200025, China.
| |
Collapse
|
32
|
Use of alginate hydrogel to improve long-term 3D culture of spermatogonial stem cells: stemness gene expression and structural features. ZYGOTE 2021; 30:312-318. [PMID: 34641993 DOI: 10.1017/s0967199421000551] [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] [Indexed: 11/07/2022]
Abstract
The quality and quantity of a spermatogonial stem-cell (SSC) culture can be measured in less time using a 3D culture in a scaffold. The present study investigated stemness gene expression and the morphological and structural characterization of SSCs encapsulated in alginate. SSCs were harvested from BALB/c neonatal mice testes through two-step mechanical and enzymatic digestion. The spermatogonial populations were separated using magnetic-activated cell sorting (MACS) using an anti-Thy1 antibody and c-Kit. The SSCs then were encapsulated in alginate hydrogel. After 2 months of SSC culturing, the alginate microbeads were extracted and stained to evaluate their histological properties. Real-time polymerase chain reaction (PCR) was performed to determine the stemness gene expression. Scanning electron microscopy (SEM) was performed to evaluate the SSC morphology, density and scaffold structure. The results showed that encapsulated SSCs had decreased expression of Oct4, Sox2 and Nanos2 genes, but the expression of Nanog, Bcl6b and Plzf genes was not significantly altered. Histological examination showed that SSCs with pale nuclei and numerous nucleolus formed colonies. SEM evaluation revealed that the alginate scaffold structure preserved the SSC morphology and density for more than 60 days. Cultivation of SSCs on alginate hydrogel can affect Oct4, Sox2 and Nanos2 expression.
Collapse
|
33
|
Pediatric and Adolescent Oncofertility in Male Patients-From Alpha to Omega. Genes (Basel) 2021; 12:genes12050701. [PMID: 34066795 PMCID: PMC8150386 DOI: 10.3390/genes12050701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 01/15/2023] Open
Abstract
This article reviews the latest information about preserving reproductive potential that can offer enhanced prospects for future conception in the pediatric male population with cancer, whose fertility is threatened because of the gonadotoxic effects of chemotherapy and radiation. An estimated 400,000 children and adolescents aged 0–19 years will be diagnosed with cancer each year. Fertility is compromised in one-third of adult male survivors of childhood cancer. We present the latest approaches and techniques for fertility preservation, starting with fertility preservation counselling, a clinical practice guideline used around the world and finishing with recent advances in basic science and translational research. Improving strategies for the maturation of germ cells in vitro combined with new molecular techniques for gene editing could be the next scientific keystone to eradicate genetic diseases such as cancer related mutations in the offspring of cancer survivors.
Collapse
|
34
|
Wei YL, She ZY, Huang T, Zhang HT, Wang XR. Male reproductive systems of Macaca mulatta: Gonadal development, spermatogenesis and applications in spermatogonia stem cell transplantation. Res Vet Sci 2021; 137:127-137. [PMID: 33965833 DOI: 10.1016/j.rvsc.2021.04.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 04/16/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Rhesus macaque (Macaca mulatta) is widely applied in animal model construction of infertility, spermatogonia stem cell transplantation and male reproductive diseases. In this review, we describe the seasonal changes of the reproductive system in rhesus macaques, the regular pattern of spermatogenesis and spermatozoa maturation, and the differentiation of spermatogonia and spermatocytes. The duration of the M. mulatta spermatogenesis is approximately 10 days and seminiferous epithelium cycles mainly consist of 12 stages, which provide a suitable model for reproductive studies in non-human primates. Here, we summarize the features of gonadal development and sperm maturation in the rhesus monkeys, which provide important information in the studies of reproductive biology. Rhesus macaque is an excellent animal model in spermatogonia stem cell transplantation. We discuss the applications and progresses of assisted reproductive technologies in sperm liquefaction, semen cryopreservation and spermatogonia stem cell transplantation of rhesus macaques. Besides, we sort out recent proteomic analyses of male reproductive systems and semen samples in rhesus macaques. This review mainly focuses on male reproductive biology and application studies using M. mulatta, which would promote the development of new therapeutic interventions on assisted reproduction and reproductive disease studies in the future.
Collapse
Affiliation(s)
- Ya-Lan Wei
- Fujian Obstetrics and Gynecology Hospital, Fuzhou, Fujian 350011, China; Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, China; Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, National Health Commission, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350013, China.
| | - Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Tao Huang
- Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, China; Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, National Health Commission, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350013, China
| | - Hai-Tao Zhang
- Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, China; Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, National Health Commission, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350013, China
| | - Xin-Rui Wang
- Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, China; Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, National Health Commission, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350013, China.
| |
Collapse
|
35
|
Is It Possible to Treat Infertility with Stem Cells? Reprod Sci 2021; 28:1733-1745. [PMID: 33834375 DOI: 10.1007/s43032-021-00566-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023]
Abstract
Infertility is a major health problem, and despite improved treatments over the years, there are still some conditions that cannot be treated successfully using a conventional approach. Therefore, new options are being considered and one of them is cell therapy using stem cells. Stem cell treatments for infertility can be divided into two major groups, the first one being direct transplantation of stem cells or their paracrine factors into reproductive organs and the second one being in vitro differentiation into germ cells or gametes. In animal models, all of these approaches were able to improve the reproductive potential of tested animals, although in humans there is still too little evidence to suggest successful use. The reasons for lack of evidence are unavailability of proper material, the complexity of explored biological processes, and ethical considerations. Despite all of the above-mentioned hurdles, researchers were able to show that in women, it seems to be possible to improve some conditions, but in men, no similar clinically important improvement was achieved. To conclude, the data presented in this review suggest that the treatment of infertility with stem cells seems plausible, because some types of treatments have already been tested in humans, achieving live births, while others show great potential only in animal studies, for now.
Collapse
|
36
|
Sawaied A, Arazi E, AbuElhija A, Lunenfeld E, Huleihel M. The Presence of Colony-Stimulating Factor-1 and Its Receptor in Different Cells of the Testis; It Involved in the Development of Spermatogenesis In Vitro. Int J Mol Sci 2021; 22:ijms22052325. [PMID: 33652607 PMCID: PMC7956383 DOI: 10.3390/ijms22052325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/12/2021] [Accepted: 02/21/2021] [Indexed: 11/25/2022] Open
Abstract
Spermatogenesis is a complex process, in which spermatogonial cells proliferate and differentiate in the seminiferous tubules of the testis to generate sperm. This process is under the regulation of endocrine and testicular paracrine/autocrine factors. In the present study, we demonstrated that colony stimulating factor-1 (CSF-1) is produced by mouse testicular macrophages, Leydig, Sertoli, peritubular cells and spermatogonial cells (such as CDH1-positively stained cells; a marker of spermatogonial cells). In addition, we demonstrated the presence of CSF-1 and its receptor (CSF-1R) in testicular macrophages, Leydig, Sertoli, peritubular cells and spermatogonial cells of human testis. We also show that the protein levels of CSF-1 were the highest in testis of 1-week-old mice and significantly decreased with age (2–12-week-old). However, the transcriptome levels of CSF-1 significantly increased in 2–3-week-old compared to 1-week-old, and thereafter significantly decreased with age. On the other hand, the transcriptome levels of CSF-1R was significantly higher in mouse testicular tissue of all examined ages (2–12-week-old) compared to 1-week-old. Our results demonstrate the involvement of CSF-1 in the induction the proliferation and differentiation of spermatogonial cells to meiotic and postmeiotic stages (BOULE- and ACROSIN-positive cells) under in vitro culture conditions, using methylcellulose culture system (MCS). Thus, it is possible to suggest that CSF-1 system, as a testicular paracrine/autocrine system, is involved in the development of different stages of spermatogenesis and may be used in the development of future therapeutic strategies for treatment of male infertility.
Collapse
Affiliation(s)
- Alaa Sawaied
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (A.S.); (E.A.); (A.A.)
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
| | - Eden Arazi
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (A.S.); (E.A.); (A.A.)
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
| | - Ahmad AbuElhija
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (A.S.); (E.A.); (A.A.)
| | - Eitan Lunenfeld
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- Department of OB/GYN, Soroka Medical Center, Beer Sheva 8410501, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (A.S.); (E.A.); (A.A.)
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- Correspondence: ; Tel.: +972-8-6479959
| |
Collapse
|
37
|
Cham TC, Chen X, Honaramooz A. Current progress, challenges, and future prospects of testis organoids†. Biol Reprod 2021; 104:942-961. [PMID: 33550399 DOI: 10.1093/biolre/ioab014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/24/2020] [Accepted: 01/26/2021] [Indexed: 12/29/2022] Open
Abstract
Spermatogenic failure is believed to be a major cause of male infertility. The establishment of a testis organoid model would facilitate the study of such pathological mechanisms and open the possibility of male fertility preservation. Because of the complex structures and cellular events occurring within the testis, the establishment of a compartmentalized testis organoid with a complete spermatogenic cycle remains a challenge in all species. Since the late 20th century, a great variety of scaffold-based and scaffold-free testis cell culture systems have been established to recapitulate de novo testis organogenesis and in vitro spermatogenesis. The utilization of the hydrogel scaffolds provides a 3D microenvironment for testis cell growth and development, facilitating the reconstruction of de novo testis tissue-like structures and spermatogenic differentiation. Using a combination of different strategies, including the use of various scaffolding biomaterials, the incorporation of the living cells with high self-assembling capacity, and the integration of the advanced fabrication techniques, a scaffold-based testis organoid with a compartmentalized structure that supports in vitro spermatogenesis may be achieved. This article briefly reviews the current progress in the development of scaffold-based testis organoids while focusing on the scaffolding biomaterials (hydrogels), cell sources, and scaffolding approaches. Key challenges in current organoid studies are also discussed along with recommendations for future research.
Collapse
Affiliation(s)
- Tat-Chuan Cham
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Xiongbiao Chen
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Ali Honaramooz
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| |
Collapse
|
38
|
Involvement of Cytokines and Hormones in the Development of Spermatogenesis In Vitro from Spermatogonial Cells of Cyclophosphamide-Treated Immature Mice. Int J Mol Sci 2021; 22:ijms22041672. [PMID: 33562323 PMCID: PMC7914946 DOI: 10.3390/ijms22041672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
Aggressive chemotherapy treatment may lead to male infertility. Prepubertal boys do not produce sperm at this age, however, they have spermatogonial stem cells in their testes. Here, we examined the effect of intraperitoneal injection of cyclophosphamide (CP) on the capacity of immature mice (IM) to develop spermatogenesis in vivo and in vitro [using methylcellulose culture system (MCS)]. Our results show a significant decrease in testicular weight, total number of testicular cells, and the number of Sertoli, peritubular, premeiotic, and meiotic/post-meiotic cells, but an increase in the percentages of damaged seminiferous tubules in CP-treated IM compared to control. The functionality of Sertoli cells was significantly affected. The addition of testosterone to isolated cells from seminiferous tubules of CP-treated IM significantly increased the percentages of premeiotic (CD9-positive cells) and meiotic/post-meiotic cells (ACROSIN-positive cells) developed in MCS compared to control. The addition of FSH did not affect developed cells in MCS compared to control, but in combination with testosterone, it significantly decreased the percentages of CD9-positive cells and ACROSIN-positive cells. The addition of IL-1 did not affect developed cells in MCS compared to control, but in combination with testosterone, it significantly increased the percentages of VASA-positive cells and BOULE-positive cells compared to IL-1 or testosterone. Addition of TNF significantly increased only CD9-positive cells in MCS compared to control, but in combination with testosterone, it significantly decreased ACROSIN-positive cells compared to testosterone. Our results show a significant impairment of spermatogenesis in the testes of CP-treated IM, and that spermatogonial cells from these mice proliferate and differentiate to meiotic/post-meiotic cells under in vitro culture conditions.
Collapse
|
39
|
Bagdadi N, Sawaied A, AbuMadighem A, Lunenfeld E, Huleihel M. The Expression Levels and Cellular Localization of Pigment Epithelium Derived Factor (PEDF) in Mouse Testis: Its Possible Involvement in the Differentiation of Spermatogonial Cells. Int J Mol Sci 2021; 22:1147. [PMID: 33498962 PMCID: PMC7865766 DOI: 10.3390/ijms22031147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 11/24/2022] Open
Abstract
Pigment epithelium derived factor (PEDF) is a multifunctional secretory soluble glycoprotein that belongs to the serine protease inhibitor (serpin) family. It was reported to have neurotrophic, anti-angiogenic and anti-tumorigenic activity. Recently, PEDF was found in testicular peritubular cells and it was assumed to be involved in the avascular nature of seminiferous tubules. The aim of this study was to determine the cellular origin, expression levels and target cells of PEDF in testicular tissue of immature and adult mice under physiological conditions, and to explore its possible role in the process of spermatogenesis in vitro. Using immunofluorescence staining, we showed that PEDF was localized in spermatogenic cells at different stages of development as well as in the somatic cells of the testis. Its protein levels in testicular homogenates and Sertoli cells supernatant showed a significant decrease with age. PEDF receptor (PEDF-R) was localized within the seminiferous tubule cells and in the interstitial cells compartment. Its RNA expression levels showed an increase with age until 8 weeks followed by a decrease. RNA levels of PEDF-R showed the opposite trend of the protein. Addition of PEDF to cultures of isolated cells from the seminiferous tubules did not changed their proliferation rate, however, a significant increase was observed in number of meiotic/post meiotic cells at 1000 ng/mL of PEDF; indicating an in vitro differentiation effect. This study may suggest a role for PEDF in the process of spermatogenesis.
Collapse
Affiliation(s)
- Noy Bagdadi
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (N.B.); (A.S.); (A.A.)
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
| | - Alaa Sawaied
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (N.B.); (A.S.); (A.A.)
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
| | - Ali AbuMadighem
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (N.B.); (A.S.); (A.A.)
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
| | - Eitan Lunenfeld
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- Department of OB/GYN, Soroka Medical Center, Beer Sheva 8410501, Israel
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (N.B.); (A.S.); (A.A.)
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
| |
Collapse
|
40
|
Saulnier J, Oblette A, Delessard M, Dumont L, Rives A, Rives N, Rondanino C. Improving Freezing Protocols and Organotypic Culture: A Histological Study on Rat Prepubertal Testicular Tissue. Ann Biomed Eng 2021; 49:203-218. [PMID: 32440757 DOI: 10.1007/s10439-020-02535-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/13/2020] [Indexed: 10/24/2022]
Abstract
Testicular tissue freezing before gonadotoxic treatments allows the preservation of fertility for children suffering from cancer. Recently, the testis organ culture method was presented as a relevant method to restore the fertility of these patients. However, the yield of spermatozoa production is low in the mouse model and no gamete has been obtained in vitro in the rat model. Here, we assess different cryopreservation protocols and culture conditions to improve the efficiency of in vitro maturation of rat prepubertal testes. Testes from male rats aged 5 or 8 days post-partum were cultured onto agarose gels of different percentages. After determining the best culture conditions, different cryopreservation protocols were assessed. Finally, testicular tissues were cultured with media of various compositions and analyzed at different time points. Our results show that the cryopreservation protocols allow the preservation of tissue architecture, cell proliferation, with no or moderate increase of cell death. In vitro spermatogenesis did not proceed beyond the pachytene spermatocyte stage. Only 2 of the 6 tested media allowed the survival of differentiated germ cells over the 45-day culture period. In conclusion, this study highlights the necessity to further improve the organ culture method before applying it into the clinics.
Collapse
Affiliation(s)
- Justine Saulnier
- Department of Reproductive Biology-CECOS, Rouen University Hospital, EA 4308 "Gametogenesis and Gamete Quality", UNIROUEN, Normandie Univ, 22 Boulevard Gambetta, 76000, Rouen, France
| | - Antoine Oblette
- Department of Reproductive Biology-CECOS, Rouen University Hospital, EA 4308 "Gametogenesis and Gamete Quality", UNIROUEN, Normandie Univ, 22 Boulevard Gambetta, 76000, Rouen, France
| | - Marion Delessard
- Department of Reproductive Biology-CECOS, Rouen University Hospital, EA 4308 "Gametogenesis and Gamete Quality", UNIROUEN, Normandie Univ, 22 Boulevard Gambetta, 76000, Rouen, France
| | - Ludovic Dumont
- Department of Reproductive Biology-CECOS, Rouen University Hospital, EA 4308 "Gametogenesis and Gamete Quality", UNIROUEN, Normandie Univ, 22 Boulevard Gambetta, 76000, Rouen, France
| | - Aurélie Rives
- Department of Reproductive Biology-CECOS, Rouen University Hospital, EA 4308 "Gametogenesis and Gamete Quality", UNIROUEN, Normandie Univ, 22 Boulevard Gambetta, 76000, Rouen, France
| | - Nathalie Rives
- Department of Reproductive Biology-CECOS, Rouen University Hospital, EA 4308 "Gametogenesis and Gamete Quality", UNIROUEN, Normandie Univ, 22 Boulevard Gambetta, 76000, Rouen, France
| | - Christine Rondanino
- Department of Reproductive Biology-CECOS, Rouen University Hospital, EA 4308 "Gametogenesis and Gamete Quality", UNIROUEN, Normandie Univ, 22 Boulevard Gambetta, 76000, Rouen, France.
| |
Collapse
|
41
|
Wyns C, Kanbar M, Giudice MG, Poels J. Fertility preservation for prepubertal boys: lessons learned from the past and update on remaining challenges towards clinical translation. Hum Reprod Update 2020; 27:433-459. [PMID: 33326572 DOI: 10.1093/humupd/dmaa050] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/25/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Childhood cancer incidence and survivorship are both on the rise. However, many lifesaving treatments threaten the prepubertal testis. Cryopreservation of immature testicular tissue (ITT), containing spermatogonial stem cells (SSCs), as a fertility preservation (FP) option for this population is increasingly proposed worldwide. Recent achievements notably the birth of non-human primate (NHP) progeny using sperm developed in frozen-thawed ITT autografts has given proof of principle of the reproductive potential of banked ITT. Outlining the current state of the art on FP for prepubertal boys is crucial as some of the boys who have cryopreserved ITT since the early 2000s are now in their reproductive age and are already seeking answers with regards to their fertility. OBJECTIVE AND RATIONALE In the light of past decade achievements and observations, this review aims to provide insight into relevant questions for clinicians involved in FP programmes. Have the indications for FP for prepubertal boys changed over time? What is key for patient counselling and ITT sampling based on the latest achievements in animals and research performed with human ITT? How far are we from clinical application of methods to restore reproductive capacity with cryostored ITT? SEARCH METHODS An extensive search for articles published in English or French since January 2010 to June 2020 using keywords relevant to the topic of FP for prepubertal boys was made in the MEDLINE database through PubMed. Original articles on fertility preservation with emphasis on those involving prepubertal testicular tissue, as well as comprehensive and systematic reviews were included. Papers with redundancy of information or with an absence of a relevant link for future clinical application were excluded. Papers on alternative sources of stem cells besides SSCs were excluded. OUTCOMES Preliminary follow-up data indicate that around 27% of boys who have undergone testicular sampling as an FP measure have proved azoospermic and must therefore solely rely on their cryostored ITT to ensure biologic parenthood. Auto-transplantation of ITT appears to be the first technique that could enter pilot clinical trials but should be restricted to tissue free of malignant cells. While in vitro spermatogenesis circumvents the risk linked to cancer cell contamination and has led to offspring in mice, complete spermatogenesis has not been achieved with human ITT. However, generation of haploid germ cells paves the way to further studies aimed at completing the final maturation of germ cells and increasing the efficiency of the processes. WIDER IMPLICATIONS Despite all the research done to date, FP for prepubertal boys remains a relatively young field and is often challenging to healthcare providers, patients and parents. As cryopreservation of ITT is now likely to expand further, it is important not only to acknowledge some of the research questions raised on the topic, e.g. the epigenetic and genetic integrity of gametes derived from strategies to restore fertility with banked ITT but also to provide healthcare professionals worldwide with updated knowledge to launch proper multicollaborative care pathways in the field and address clinical issues that will come-up when aiming for the child's best interest.
Collapse
Affiliation(s)
- Christine Wyns
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Maria Grazia Giudice
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jonathan Poels
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
42
|
Isolation, identification and differentiation of human spermatogonial cells on three-dimensional decellularized sheep testis. Acta Histochem 2020; 122:151623. [PMID: 32992121 DOI: 10.1016/j.acthis.2020.151623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/19/2022]
Abstract
Improvement of in vitro culture methods of Spermatogonial Stem Cells (SSCs) is known to be an effective procedure for further study of the process of spermatogenesis and can offer effective therapeutic modality for male infertility. Tissue decellularization by providing natural 3D and extracellular matrix (ECM) conditions for cell growth can be an alternative procedure to enhance in vitro culture conditions. In the present study, the testicular tissues were taken from brain death donors. After enzymatic digestion, the tissue cells were isolated and cultured for four weeks. Then the identity of the SSCs was confirmed using anti-GFRα1 and anti-PLZF antibodies via immunocytochemistry (ICC). The differentiation capacity of SSCs were evaluated by culture of them on a layer of decellularized testicular matrix (DTM) prepared from sheep testis, as well as under two-dimensional (2D) culture with differentiation medium. After four and six weeks of the initiation of differentiation culture, the pre-meiotic, meiotic and post- meiotic genes at the mRNA and protein levels was examined via qPCR and ICC methods, respectively. The results showed that pre-meiotic, meiotic and post-meiotic genes expressions were significantly higher in the cells cultured in DTM substrate (P ≤ 0.01).The present study indicated that, the natural structure of ECM prepare the suitable conditions for further study of the spermatogenesis process in the in vitro and contributes to the maintenance and treatment of male infertility.
Collapse
|
43
|
Application of platelet-rich plasma (PRP) improves self-renewal of human spermatogonial stem cells in two-dimensional and three-dimensional culture systems. Acta Histochem 2020; 122:151627. [PMID: 33002788 DOI: 10.1016/j.acthis.2020.151627] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/17/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
Spermatogonial stem cells (SSCs) are very sensitive to chemotherapy and radiotherapy, so male infertility is a great challenge for prepubertal cancer survivors. Cryoconservation of testicular cells before cancer treatment can preserve SSCs from treatment side effects. Different two-dimensional (2D) and three-dimensional (3D) culture systems of SSCs have been used in many species as a useful technique to in vitro spermatogenesis. We evaluated the proliferation of SSCs in 2D and 3D culture systems of platelet-rich plasma (PRP). testicular cells of four brain-dead patients cultivated in 2D pre-culture system, characterization of SSCs performed by RT-PCR, flow cytometry, immunocytochemistry and their functionality assessed by xenotransplantation to azoospermia mice. PRP prepared and dosimetry carried out to determine the optimized dose of PRP. After preparation of PRP scaffold, cytotoxic and histological evaluation performed and SSCs cultivated into three groups: control, 2D culture by optimized dose of PRP and PRP scaffold. The diameter and number of colonies measured and relative expression of GFRa1 and c-KIT evaluated by real-time PCR. Results indicated the expression of PLZF, VASA, OCT4, GFRa1 and vimentin in colonies after 2D pre-culture, xenotransplantation demonstrated proliferated SSCs have proper functionality to homing in mouse testes. The relative expression of c-KIT showed a significant increase as compared to the control group (*: p < 0.05) in PRP- 2D group, expression of GFRa1 and c-KIT in PRP scaffold group revealed a significant increase as compared to other groups (***: p < 0.001). The number and diameter of colonies in the PRP-2D group showed a considerable increase (p < 0.01) as compared to the control group. In PRP- scaffold group, a significant increase (p < 0.01) was seen only in the number of colonies related to the control group. Our results suggested that PRP scaffold can reconstruct a suitable structure to the in vitro proliferation of SSCs.
Collapse
|
44
|
Interleukin-34, a Novel Paracrine/Autocrine Factor in Mouse Testis, and Its Possible Role in the Development of Spermatogonial Cells In Vitro. Int J Mol Sci 2020; 21:ijms21218143. [PMID: 33143373 PMCID: PMC7662511 DOI: 10.3390/ijms21218143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/23/2020] [Accepted: 10/26/2020] [Indexed: 01/15/2023] Open
Abstract
Spermatogenesis is the process of spermatogonial stem cell (SSC) proliferation and differentiation to generate sperm. This process is regulated by cell–cell interactions between Sertoli cells and developing SSCs by autocrine/paracrine and endocrine factors. It is also affected by cells in the interstitial compartment, such as Leydig cells and peritubular cells. Here, we demonstrate, for the first time, the presence of interleukin-34 (IL-34) in Leydig, Sertoli, and peritubular cells and in the premeiotic, meiotic, and postmeiotic cells. Its receptor, colony-stimulating factor-1 (CSF-1), has already been demonstrated in Leydig, Sertoli, premeiotic, and meiotic cells. IL-34 was detected in testicular homogenates and Sertoli cell-conditioned media, and was affected by mouse age. We showed that the addition of IL-34 in vitro to isolated cells from the seminiferous tubules of 7-day-old mice, using the methylcellulose culture system (MCS), increased the percentages and expression of the premeiotic cells (VASA), the meiotic cells (BOULE), and the meiotic/postmeiotic cells (ACROSIN) after four weeks of culture, when examined by immunofluorescence staining (IF) and qPCR analysis. It is possible to suggest that IL-34 is a novel paracrine/autocrine factor involved in the development of spermatogenesis. This factor may be used in future therapeutic strategies for the treatment of male infertility.
Collapse
|
45
|
Liu HC, Xie Y, Deng CH, Liu GH. Stem cell-based therapies for fertility preservation in males: Current status and future prospects. World J Stem Cells 2020; 12:1097-1112. [PMID: 33178394 PMCID: PMC7596443 DOI: 10.4252/wjsc.v12.i10.1097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/13/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
With the decline in male fertility in recent years, strategies for male fertility preservation have received increasing attention. In this study, by reviewing current treatments and recent publications, we describe research progress in and the future directions of stem cell-based therapies for male fertility preservation, focusing on the use of spermatogonial stem cells (SSCs), SSC niches, SSC-based testicular organoids, other stem cell types such as mesenchymal stem cells, and stem cell-derived extracellular vesicles. In conclusion, a more comprehensive understanding of the germ cell microenvironment, stem cell-derived extracellular vesicles, and testicular organoids will play an important role in achieving male fertility preservation.
Collapse
Affiliation(s)
- Han-Chao Liu
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Yun Xie
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Chun-Hua Deng
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Gui-Hua Liu
- Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| |
Collapse
|
46
|
Deebel NA, Galdon G, Zarandi NP, Stogner-Underwood K, Howards S, Lovato J, Kogan S, Atala A, Lue Y, Sadri-Ardekani H. Age-related presence of spermatogonia in patients with Klinefelter syndrome: a systematic review and meta-analysis. Hum Reprod Update 2020; 26:58-72. [PMID: 31822886 DOI: 10.1093/humupd/dmz038] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/15/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Klinefelter syndrome (KS) has been defined by sex chromosome aneuploidies (classically 47, XXY) in the male patient. The peripubertal timeframe in KS patients has been associated with the initiation of progressive testicular fibrosis, loss of spermatogonial stem cells (SSC), hypogonadism and impaired fertility. Less than half of KS patients are positive for spermatozoa in the ejaculate or testis via semen analysis or testicular sperm extraction, respectively. However, the chance of finding spermatogonia including a sub-population of SSCs in KS testes has not been well defined. Given the recent demonstration of successful cell culture for mouse and human SSCs, it could be feasible to isolate and propagate SSCs and transplant the cells back to the patient or to differentiate them in vitro to haploid cells. OBJECTIVE AND RATIONALE The main objective of this study was to meta-analyse the currently available data from KS patients to identify the prevalence of KS patients with spermatogonia on testicular biopsy across four age groups (year): fetal/infantile (age ≤ 1), prepubertal (age 1 ≤ x ≤ 10), peripubertal/adolescent (age 10 < x < 18) and adult (age ≥ 18) ages. Additionally, the association of endocrine parameters with presence or absence of spermatogonia was tested to obtain a more powered analysis of whether FSH, LH, testosterone and inhibin B can serve as predictive markers for successful spermatogonia retrieval. SEARCH METHODS A thorough Medline/PubMed search was conducted using the following search terms: 'Klinefelter, germ cells, spermatogenesis and spermatogonia', yielding results from 1 October 1965 to 3 February 2019. Relevant articles were added from the bibliographies of selected articles. Exclusion criteria included non-English language, abstracts only, non-human data and review papers. OUTCOMES A total of 751 papers were identified with independent review returning 36 papers with relevant information for meta-analysis on 386 patients. For the most part, articles were case reports, case-controlled series and cohort studies (level IV-VI evidence). Spermatogonial cells were present in all of the fetal/infantile and 83% of the prepubertal patients' testes, and in 42.7% and 48.5% of the peripubertal and adult groups, respectively were positive for spermatogonia. Additionally, 26 of the 56 (46.4%) peripubertal/adolescent and 37 of the 152 (24.3%) adult patients negative for spermatozoa were positive for spermatogonia (P < 0.05). In peripubertal/adolescent patients, the mean ± SEM level for FSH was 12.88 ± 3.13 IU/L for spermatogonia positive patients and 30.42 ± 4.05 IU/L for spermatogonia negative patients (P = 0.001); the mean ± SEM level LH levels were 4.36 ± 1.31 and 11.43 ± 1.68 IU/L for spermatogonia positive and negative, respectively (P < 0.01); the mean ± SEM level for testosterone levels were 5.04 ± 1.37 and 9.05 ± 0.94 nmol/L (equal to 145 ± 40 and 261 ± 27 and ng/dl) for the spermatogonia positive and negative groups, respectively (P < 0.05), while the difference in means for inhibin B was not statistically significant (P > 0.05). A similar analysis in the adult group showed the FSH levels in spermatogonia positive and negative patients to be 25.77 ± 2.78 and 36.12 ± 2.90 IU/L, respectively (mean ± SEM level, P < 0.05). All other hormone measurements were not statistically significantly different between groups. WIDER IMPLICATIONS While azoospermia is a common finding in the KS patient population, many patients are positive for spermatogonia. Recent advances in SSC in vitro propagation, transplantation and differentiation open new avenues for these patients for fertility preservation. This would offer a new subset of KS patients a chance of biological paternity. Data surrounding the hormonal profiles of KS patients and their relation to fertility should be interpreted with caution as a paucity of adequately powered data exists. Future work is needed to clarify the utility of FSH, LH, testosterone and inhibin B as biomarkers for successful retrieval of spermatogonia.
Collapse
Affiliation(s)
- Nicholas A Deebel
- Department of Urology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Guillermo Galdon
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Nima Pourhabibi Zarandi
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | - Stuart Howards
- Department of Urology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - James Lovato
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Stanley Kogan
- Department of Urology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Anthony Atala
- Department of Urology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Yanhe Lue
- Division of Endocrinology, Department of Medicine, Los Angeles Biomedical Research Institute and Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Hooman Sadri-Ardekani
- Department of Urology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
47
|
Approaches and Technologies in Male Fertility Preservation. Int J Mol Sci 2020; 21:ijms21155471. [PMID: 32751826 PMCID: PMC7432867 DOI: 10.3390/ijms21155471] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Male fertility preservation is required when treatment with an aggressive chemo-/-radiotherapy, which may lead to irreversible sterility. Due to new and efficient protocols of cancer treatments, surviving rates are more than 80%. Thus, these patients are looking forward to family life and fathering their own biological children after treatments. Whereas adult men can cryopreserve their sperm for future use in assistance reproductive technologies (ART), this is not an option in prepubertal boys who cannot produce sperm at this age. In this review, we summarize the different technologies for male fertility preservation with emphasize on prepubertal, which have already been examined and/or demonstrated in vivo and/or in vitro using animal models and, in some cases, using human tissues. We discuss the limitation of these technologies for use in human fertility preservation. This update review can assist physicians and patients who are scheduled for aggressive chemo-/radiotherapy, specifically prepubertal males and their parents who need to know about the risks of the treatment on their future fertility and the possible present option of fertility preservation.
Collapse
|
48
|
Jabari A, Sadighi Gilani MA, Koruji M, Gholami K, Mohsenzadeh M, Rastegar T, Khadivi F, Ghanami Gashti N, Nikmahzar A, Mojaverrostami S, Talebi A, Ashouri Movassagh S, Rezaie MJ, Abbasi M. Three-dimensional co-culture of human spermatogonial stem cells with Sertoli cells in soft agar culture system supplemented by growth factors and Laminin. Acta Histochem 2020; 122:151572. [PMID: 32622422 DOI: 10.1016/j.acthis.2020.151572] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022]
Abstract
Application of a three-dimensional (3D) culture system for in vitro proliferation and differentiation of human spermatogonial stem cells (SSCs) is a useful tool for the investigation of the spermatogenesis process and the management of male infertility particularly in prepubertal cancer patients. The main purpose of this study was to investigate the proliferation of human SSCs co-cultured with Sertoli cells in soft agar culture system (SACS) supplemented by Laminin and growth factors. Testicular cells were isolated from testes of brain-dead patients and cultured in two-dimensional (2D) culture system for 3 weeks. After 3 weeks, functional SSCs were evaluated by xenotransplantation and also identification of cells was assessed by immunocytochemistry, flow cytometry, and RT-PCR. Then, SSCs and Sertoli cells were transferred to the upper layer of SACS for 3 weeks. After 3 weeks, the number of colonies and the expression of specific SSCs and Sertoli cell markers, as well as apoptotic genes were evaluated. Our results showed that transplanted SSCs, migrated into the basement membrane of seminiferous tubules of recipient mice. The expression of PLZF, α6-Integrin, and Vimentin proteins in SSCs and Sertoli cells were observed in 2D and 3D culture systems. The expression rate of PLZF, α6-Integrin, Bcl2, and colony number in SACS supplemented by Laminin and growth factors group were significantly higher than non-supplemented groups (P ≤ 0.01), but the expression rate of c-kit and Bax in supplemented group were significantly lower than non-supplemented groups (P ≤ 0.05). This 3D co-culture system decreased apoptosis and increased propagation of human SSCs. Therefore, this designed system can be utilized to increase the proliferation of human SSCs in prepubertal male cancer and azoospermic men to obtain an adequate SSCs number to outotransplant success and in vitro spermatogenesis.
Collapse
Affiliation(s)
- Ayob Jabari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Morteza Koruji
- Cellular and Molecular Research Center & Department of Anatomical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Keykavos Gholami
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mojtaba Mohsenzadeh
- Iranian Tissue Bank and Research Center of Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Rastegar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Khadivi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Ghanami Gashti
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aghbibi Nikmahzar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Talebi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Sepideh Ashouri Movassagh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Mohammad Jafar Rezaie
- Department of Embryology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mehdi Abbasi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Wormser O, Levy Y, Bakhrat A, Bonaccorsi S, Graziadio L, Gatti M, AbuMadighem A, McKenney RJ, Okada K, El Riati S, Har-Vardi I, Huleihel M, Levitas E, Birk OS, Abdu U. Absence of SCAPER causes male infertility in humans and Drosophila by modulating microtubule dynamics during meiosis. J Med Genet 2020; 58:254-263. [PMID: 32527956 PMCID: PMC10405349 DOI: 10.1136/jmedgenet-2020-106946] [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: 02/23/2020] [Revised: 04/22/2020] [Accepted: 04/25/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Mutation in S-phase cyclin A-associated protein rin the endoplasmic reticulum (SCAPER) have been found across ethnicities and have been shown to cause variable penetrance of an array of pathological traits, including intellectual disability, retinitis pigmentosa and ciliopathies. METHODS Human clinical phenotyping, surgical testicular sperm extraction and testicular tissue staining. Generation and analysis of short spindle 3 (ssp3) (SCAPER orthologue) Drosophila CAS9-knockout lines. In vitro microtubule (MT) binding assayed by total internal reflection fluorescence microscopy. RESULTS We show that patients homozygous for a SCAPER mutation lack SCAPER expression in spermatogonia (SPG) and are azoospermic due to early defects in spermatogenesis, leading to the complete absence of meiotic cells. Interestingly, Drosophila null mutants for the ubiquitously expressed ssp3 gene are viable and female fertile but male sterile. We further show that male sterility in ssp3 null mutants is due to failure in both chromosome segregation and cytokinesis. In cells undergoing male meiosis, the MTs emanating from the centrosomes do not appear to interact properly with the chromosomes, which remain dispersed within dividing spermatocytes (SPCs). In addition, mutant SPCs are unable to assemble a normal central spindle and undergo cytokinesis. Consistent with these results, an in vitro assay demonstrated that both SCAPER and Ssp3 directly bind MTs. CONCLUSIONS Our results show that SCAPER null mutations block the entry into meiosis of SPG, causing azoospermia. Null mutations in ssp3 specifically disrupt MT dynamics during male meiosis, leading to sterility. Moreover, both SCAPER and Ssp3 bind MTs in vitro. These results raise the intriguing possibility of a common feature between human and Drosophila meiosis.
Collapse
Affiliation(s)
- Ohad Wormser
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ygal Levy
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anna Bakhrat
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Silvia Bonaccorsi
- Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Lucia Graziadio
- Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Maurizio Gatti
- Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza University of Rome, Rome, Italy.,Istituto di Biologia e Patologia Molecolari Consiglio Nazionale delle Ricerche, Roma, Italy
| | - Ali AbuMadighem
- Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev Faculty of Health Sciences, Beer-Sheva, Israel.,The Center of Advanced Research and Education in Reproduction (CARER), Ben-Gurion University of the Negev Faculty of Health Sciences, Beer-Sheva, Israel
| | - Richard J McKenney
- Department of Molecular and Cellular Biology, UC Davis, Davis, California, USA
| | - Kyoko Okada
- Department of Molecular and Cellular Biology, UC Davis, Davis, California, USA
| | - Saad El Riati
- Southern District, Clalit Health Services, Beer-Sheva, Israel
| | - Iris Har-Vardi
- The Center of Advanced Research and Education in Reproduction (CARER), Ben-Gurion University of the Negev Faculty of Health Sciences, Beer-Sheva, Israel.,Fertility and IVF Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Mahmoud Huleihel
- Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev Faculty of Health Sciences, Beer-Sheva, Israel.,The Center of Advanced Research and Education in Reproduction (CARER), Ben-Gurion University of the Negev Faculty of Health Sciences, Beer-Sheva, Israel
| | - Eliahu Levitas
- The Center of Advanced Research and Education in Reproduction (CARER), Ben-Gurion University of the Negev Faculty of Health Sciences, Beer-Sheva, Israel.,Fertility and IVF Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Ohad S Birk
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel .,Genetics Institute, Soroka University Medical Center, Beer-Sheva, Israel
| | - Uri Abdu
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
50
|
Gao H, Liu C, Wu B, Cui H, Zhao Y, Duan Y, Gao F, Gu Q, Wang H, Li W. Effects of Different Biomaterials and Cellular Status on Testicular Cell Self-Organization. ACTA ACUST UNITED AC 2020; 4:e1900292. [PMID: 32453509 DOI: 10.1002/adbi.201900292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/03/2020] [Indexed: 01/12/2023]
Abstract
A multicellular organism's development is coupled with cellular self-organization, which is regulated by cell-cell interactions and cell-extracellular matrix (ECM) crosstalk. Testicular cells from different species such as mouse, rat, and porcine can self-organize into seminiferous tubules both in vitro and in vivo, but the understanding of the functional role of the ECM during this process is limited. Here, it is shown that mouse testicular cells encapsulated with the biomaterial Matrigel can self-organize into seminiferous tubules with blood-testis barrier (BTB) formation and Leydig cell differentiation. By varying the encapsulation method, a combination of sodium alginate and collagen is used to promote reorganization of seminiferous tubules, which resemble those in vivo. In addition, the self-organization ability of testicular cells declines with advanced cell age, and those germ cells play a pivotal role in this process. These findings will be helpful to understand the self-organization process of testicular cells and provide insights for the reconstruction of testes.
Collapse
Affiliation(s)
- Hui Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Bingbing Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hang Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Yan Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yongchao Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Stem cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|