1
|
Gizer M, Önen S, Korkusuz P. The Evolutionary Route of in vitro Human Spermatogenesis: What is the Next Destination? Stem Cell Rev Rep 2024; 20:1406-1419. [PMID: 38684571 PMCID: PMC11319530 DOI: 10.1007/s12015-024-10726-2] [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] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Malfunction in spermatogenesis due to genetic diseases, trauma, congenital disorders or gonadotoxic treatments results in infertility in approximately 7% of males. The behavior of spermatogonial stem cells (SSCs) within three-dimensional, multifactorial, and dynamic microenvironment implicates a niche that serves as a repository for fertility, since can serve as a source of mature and functional male germ cells. Current protocols enable reprogramming of mature somatic cells into induced pluripotent stem cells (iPSCs) and their limited differentiation to SSCs within the range of 0-5%. However, the resulting human iPSC-derived haploid spermatogenic germ cell yield in terms of number and functionality is currently insufficient for transfer to infertility clinic as a therapeutic tool. In this article, we reviewed the evolution of experimental culture platforms and introduced a novel iPSCs-based approach for in vitro spermatogenesis based on a niche perspective bearing cellular, chemical, and physical factors that provide the complex arrangement of testicular seminiferous tubules embedded within a vascularized stroma. We believe that bioengineered organoids supported by smart bio-printed tubules and microfluidic organ-on-a-chip systems offer efficient, precise, personalized platforms for autologous pluripotent stem cell sources to undergo the spermatogenetic cycle, presenting a promising tool for infertile male patients with complete testicular aplasia.
Collapse
Affiliation(s)
- Merve Gizer
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, 06100, Ankara, Turkey
- METU MEMS Center, 06530, Ankara, Turkey
| | | | - Petek Korkusuz
- METU MEMS Center, 06530, Ankara, Turkey.
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey.
| |
Collapse
|
2
|
Wu Y, Wang C, Fan X, Ma Y, Liu Z, Ye X, Shen C, Wu C. The impact of induced pluripotent stem cells in animal conservation. Vet Res Commun 2024; 48:649-663. [PMID: 38228922 DOI: 10.1007/s11259-024-10294-3] [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: 11/02/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
It is widely acknowledged that we are currently facing a critical tipping point with regards to global extinction, with human activities driving us perilously close to the brink of a devastating sixth mass extinction. As a promising option for safeguarding endangered species, induced pluripotent stem cells (iPSCs) hold great potential to aid in the preservation of threatened animal populations. For endangered species, such as the northern white rhinoceros (Ceratotherium simum cottoni), supply of embryos is often limited. After the death of the last male in 2019, only two females remained in the world. IPSC technology offers novel approaches and techniques for obtaining pluripotent stem cells (PSCs) from rare and endangered animal species. Successful generation of iPSCs circumvents several bottlenecks that impede the development of PSCs, including the challenges associated with establishing embryonic stem cells, limited embryo sources and immune rejection following embryo transfer. To provide more opportunities and room for growth in our work on animal welfare, in this paper we will focus on the progress made with iPSC lines derived from endangered and extinct species, exploring their potential applications and limitations in animal welfare research.
Collapse
Affiliation(s)
- Yurou Wu
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Chengwei Wang
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Xinyun Fan
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Yuxiao Ma
- Department of Biology, New York University, New York, NY, USA
| | - Zibo Liu
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Xun Ye
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Chongyang Shen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China.
| | - Chunjie Wu
- Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu Univesity of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China.
- Sichuan Engineering Research Center for Endangered Medicinal Animals, Chengdu, China.
| |
Collapse
|
3
|
Aponte PM, Gutierrez-Reinoso MA, Garcia-Herreros M. Bridging the Gap: Animal Models in Next-Generation Reproductive Technologies for Male Fertility Preservation. Life (Basel) 2023; 14:17. [PMID: 38276265 PMCID: PMC10820126 DOI: 10.3390/life14010017] [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: 09/05/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
This review aims to explore advanced reproductive technologies for male fertility preservation, underscoring the essential role that animal models have played in shaping these techniques through historical contexts and into modern applications. Rising infertility concerns have become more prevalent in human populations recently. The surge in male fertility issues has prompted advanced reproductive technologies, with animal models playing a pivotal role in their evolution. Historically, animal models have aided our understanding in the field, from early reproductive basic research to developing techniques like artificial insemination, multiple ovulation, and in vitro fertilization. The contemporary landscape of male fertility preservation encompasses techniques such as sperm cryopreservation, testicular sperm extraction, and intracytoplasmic sperm injection, among others. The relevance of animal models will undoubtedly bridge the gap between traditional methods and revolutionary next-generation reproductive techniques, fortifying our collective efforts in enhancing male fertility preservation strategies. While we possess extensive knowledge about spermatogenesis and its regulation, largely thanks to insights from animal models that paved the way for human infertility treatments, a pressing need remains to further understand specific infertility issues unique to humans. The primary aim of this review is to provide a comprehensive analysis of how animal models have influenced the development and refinement of advanced reproductive technologies for male fertility preservation, and to assess their future potential in bridging the gap between current practices and cutting-edge fertility techniques, particularly in addressing unique human male factor infertility.
Collapse
Affiliation(s)
- Pedro M. Aponte
- Colegio de Ciencias Biológicas y Ambientales (COCIBA), Universidad San Francisco de Quito (USFQ), Quito 170901, Ecuador
- Instituto de Investigaciones en Biomedicina “One-Health”, Universidad San Francisco de Quito (USFQ), Campus Cumbayá, Quito 170901, Ecuador
| | - Miguel A. Gutierrez-Reinoso
- Facultad de Ciencias Agropecuarias y Recursos Naturales, Carrera de Medicina Veterinaria, Universidad Técnica de Cotopaxi (UTC), Latacunga 050150, Ecuador;
- Laboratorio de Biotecnología Animal, Departamento de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad de Concepción (UdeC), Chillán 3780000, Chile
| | - Manuel Garcia-Herreros
- Instituto Nacional de Investigação Agrária e Veterinária (INIAV), 2005-048 Santarém, Portugal
| |
Collapse
|
4
|
Adriansyah RF, Margiana R, Supardi S, Narulita P. Current Progress in Stem Cell Therapy for Male Infertility. Stem Cell Rev Rep 2023; 19:2073-2093. [PMID: 37440145 DOI: 10.1007/s12015-023-10577-3] [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] [Accepted: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Infertility has become one of the most common issues worldwide, which has negatively affected society and infertile couples. Meanwhile, male infertility is responsible for about 50% of infertility. Accordingly, a great number of researchers have focused on its treatment during the last few years; however, current therapies such as assisted reproductive technology (ART) are not effective enough in treating male infertility. Because of their self-renewal and differentiation capabilities and unlimited sources, stem cells have recently raised great hope in the treatment of reproductive system disorders. Stem cells are undifferentiated cells that can induce different numbers of specific cells, such as male and female gametes, demonstrating their potential application in the treatment of infertility. The present review aimed at identifying the causes and potential factors that influence male fertility. Besides, we highlighted the recent studies that investigated the efficiency of stem cells such as spermatogonial stem cells (SSCs), embryonic stem cells (ESCs), very small embryonic-like stem cells (VSELs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs) in the treatment of various types of male infertility.
Collapse
Affiliation(s)
| | - Ria Margiana
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Indonesia General Academic Hospital, Depok, Indonesia.
- Ciptomangunkusumo General Academic Hospital, Jakarta, Indonesia.
| | - Supardi Supardi
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Pety Narulita
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
5
|
Fath-Bayati L, Naserpour L, Khoshandam M, Jannatifar R, Fazaeli H. Recent advances in developing 3D culture systems of spermatogonial stem cell preservation and differentiation: A narrative review. Int J Reprod Biomed 2023; 21:681-696. [PMID: 37969562 PMCID: PMC10643686 DOI: 10.18502/ijrm.v21i9.14397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/02/2023] [Accepted: 08/12/2023] [Indexed: 11/17/2023] Open
Abstract
Male infertility has received vast attention in recent years and has no clear etiology in almost 40% of cases. Several methods have been suggested for preserving sperm and spermatogonial stem cells (SSCs) in both in vivo and in vitro conditions. The efficacy of these methods is related to their abilities, including providing an optimal environment for sperm preservation and long-term SSC culture for in vivo and in vitro differentiation of these cells. In this review article, a full MEDLINE/PubMed search was performed using the following search terms: "Spermatogonial Progenitor Cells, Stem Cells, Fertility Preservations, Sperm Freezing, Cell Differentiations, Tissue Scaffold, 3-Dimensional Cell Culture", which retrieved results from 1973-2022. Related articles were added to the bibliography of selected articles. Exclusion criteria included non-English language, abstract only, and unrelated articles. The production of functioning male germ cells is suggested by introducing modern bioengineered systems as a new hope for the maintenance of male fertility. Till now, few in vitro spermatogenesis investigations have provided appreciable amounts of mature gametes. Each method had benefits and disadvantages, but the 3-dimensional culture method had the greatest impact on the differentiation and preservation of SSCs. One of the critical elements of research is the preservation of sperm and the differentiation of SSCs. Several methods have been employed in this area. Various scaffolds providing an environment similar to an extracellular matrix and conditions for germ cell development and survival have been employed in recent research.
Collapse
Affiliation(s)
- Leyla Fath-Bayati
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Leila Naserpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Department of Reproductive Biology, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| | - Mohadeseh Khoshandam
- Department of Reproductive Biology, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
- National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Rahil Jannatifar
- Department of Reproductive Biology, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| | - Hoda Fazaeli
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| |
Collapse
|
6
|
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: 3.0] [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
|
7
|
In vitro differentiation of primed human induced pluripotent stem cells into primordial germ cell-like cells. Mol Biol Rep 2023; 50:1971-1979. [PMID: 36534237 DOI: 10.1007/s11033-022-08012-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/06/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Previous studies have shown significant results in the differentiation of mouse-induced pluripotent stem cells (miPSCs) into primordial germ cell-like cells (PGCLCs) and that human iPSCs (hiPSCs) can also differentiate into PGCLCs; however, the efficiency of PGCLC induction from hiPSCs is < 5%. In this study, we examined a new protocol to differentiate hiPSCs into PGCLCs. METHODS AND RESULTS hiPSCs-derived embryoid bodies (EBs) were exposed to differentiate inducing factors, bone morphogenetic protein 4 (BMP4), and retinoic acid (RA) for 6 days. Cell differentiation was assessed by reverse transcriptase-polymerase chain reaction (RT-PCR) and immunofluorescence (IF) studies. Our results showed increased expression of the PRDM1 gene on the first day of differentiation. On other days, DAZL, VASA, and STRA8 genes increased, and the expression of PRDM1, NANOG, and OCT4 genes decreased. The expression of VASA, C-KIT, and STRA8 proteins was confirmed by IF. A flow cytometry analysis revealed that ~ 60% of differentiated cells were VASA- and STRA8-positive. CONCLUSION EB formation and constant exposure of EBs to BMP4 and RA lead to the differentiation of hiPSCs into PGCLCs.
Collapse
|
8
|
Babaei K, Aziminezhad M, Norollahi SE, Vahidi S, Samadani AA. Cell therapy for the treatment of reproductive diseases and infertility: an overview from the mechanism to the clinic alongside diagnostic methods. Front Med 2022; 16:827-858. [PMID: 36562947 DOI: 10.1007/s11684-022-0948-8] [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: 03/12/2022] [Accepted: 06/28/2022] [Indexed: 12/24/2022]
Abstract
Infertility is experienced by 8%-12% of adults in their reproductive period globally and has become a prevalent concern. Besides routine therapeutic methods, stem cells are rapidly being examined as viable alternative therapies in regenerative medicine and translational investigation. Remarkable progress has been made in understanding the biology and purpose of stem cells. The affected pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs) are further studied for their possible use in reproductive medicine, particularly for infertility induced by premature ovarian insufficiency and azoospermia. Accordingly, this study discusses current developments in the use of some kinds of MSCs such as adipose-derived stem cells, bone marrow stromal cells, umbilical cord MSCs, and menstrual blood MSCs. These methods have been used to manage ovarian and uterine disorders, and each technique presents a novel method for the therapy of infertility.
Collapse
Affiliation(s)
- Kosar Babaei
- Non-Communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Mohsen Aziminezhad
- Non-Communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.,UMR INSERM U 1122, IGE-PCV, Interactions Gène-Environment En Physiopathologie Cardiovascular Université De Lorraine, Nancy, France
| | - Seyedeh Elham Norollahi
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Sogand Vahidi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
9
|
Roshandel E, Mehravar M, Nikoonezhad M, Alizadeh AM, Majidi M, Salimi M, Hajifathali A. Cell-Based Therapy Approaches in Treatment of Non-obstructive Azoospermia. Reprod Sci 2022; 30:1482-1494. [PMID: 36380137 PMCID: PMC9666961 DOI: 10.1007/s43032-022-01115-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022]
Abstract
The rate of infertility has globally increased in recent years for a variety of reasons. One of the main causes of infertility in men is azoospermia that is defined by the absence of sperm in the ejaculate and classified into two categories: obstructive azoospermia and non-obstructive azoospermia. In non-obstructive azoospermia, genital ducts are not obstructed, but the testicles do not produce sperm at all, due to various reasons. Non-obstructive azoospermia in most cases has no therapeutic options other than assisted reproductive techniques, which in most cases require sperm donors. Here we discuss cell-based therapy approaches to restore fertility in men with non-obstructive azoospermia including cell-based therapies of non-obstructive azoospermia using regenerative medicine and cell-based therapies of non-obstructive azoospermia by paracrine and anti-inflammatory pathway, technical and ethical challenges for using different cell sources and alternative options will be described, and then the more effectual approaches will be mentioned as future trends.
Collapse
Affiliation(s)
- Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| | - Maryam Mehravar
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| | - Maryam Nikoonezhad
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| | - Afshin Mohammad Alizadeh
- Department of Internal Medicine, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Majidi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Salimi
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| |
Collapse
|
10
|
Tahmasbpour Marzouni E, Stern C, Henrik Sinclair A, Tucker EJ. Stem Cells and Organs-on-chips: New Promising Technologies for Human Infertility Treatment. Endocr Rev 2022; 43:878-906. [PMID: 34967858 DOI: 10.1210/endrev/bnab047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Indexed: 11/19/2022]
Abstract
Having biological children remains an unattainable dream for most couples with reproductive failure or gonadal dysgenesis. The combination of stem cells with gene editing technology and organ-on-a-chip models provides a unique opportunity for infertile patients with impaired gametogenesis caused by congenital disorders in sex development or cancer survivors. But how will these technologies overcome human infertility? This review discusses the regenerative mechanisms, applications, and advantages of different types of stem cells for restoring gametogenesis in infertile patients, as well as major challenges that must be overcome before clinical application. The importance and limitations of in vitro generation of gametes from patient-specific human-induced pluripotent stem cells (hiPSCs) will be discussed in the context of human reproduction. The potential role of organ-on-a-chip models that can direct differentiation of hiPSC-derived primordial germ cell-like cells to gametes and other reproductive organoids is also explored. These rapidly evolving technologies provide prospects for improving fertility to individuals and couples who experience reproductive failure.
Collapse
Affiliation(s)
- Eisa Tahmasbpour Marzouni
- Laboratory of Regenerative Medicine & Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Catharyn Stern
- Royal Women's Hospital, Parkville and Melbourne IVF, Melbourne, Australia
| | - Andrew Henrik Sinclair
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Elena Jane Tucker
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
11
|
Bashiri Z, Gholipourmalekabadi M, Falak R, Amiri I, Asgari H, Chauhan NPS, Koruji M. In vitro production of mouse morphological sperm in artificial testis bioengineered by 3D printing of extracellular matrix. Int J Biol Macromol 2022; 217:824-841. [PMID: 35905760 DOI: 10.1016/j.ijbiomac.2022.07.127] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 11/30/2022]
Abstract
Since autologous stem cell transplantation is prone to cancer recurrence, in vitro sperm production is regarded a safer approach to fertility preservation. In this study, the spermatogenesis process on testicular tissue extracellular matrix (T-ECM)-derived printing structure was evaluated. Ram testicular tissue was decellularized using a hypertonic solution containing triton and the extracted ECM was used as a bio-ink to print an artificial testis. Following cell adhesion and viability examination, pre-meiotic and post-meiotic cells in the study groups (as testicular suspension and co-culture with Sertoli cells) were confirmed by real-time PCR, flow-cytometry and immunocytochemistry methods. Morphology of differentiated cells was evaluated using transmission electron microscopy (TEM), toluidine blue, Giemsa, and hematoxylin and eosin (H&E) staining. The functionality of Leydig and Sertoli cells was determined by their ability for hormone secretion. The decellularization of testicular tissue fragments was successful and had efficiently removed the cellular debris and preserved the ECM compounds. High cell viability, colonization, and increased expression of pre-meiotic markers in cultured testicular cells on T-ECM-enriched scaffolds confirmed their proliferation. Furthermore, the inoculation of neonatal mouse testicular cells onto T-ECM-enriched scaffolds resulted in the generation of sperm. Morphology evaluation showed that the structure of these cells was quite similar to mature sperm with a specialized tail structure. The hormonal analysis also confirmed production and secretion of testosterone and inhibin B by Leydig and Sertoli cells. T-ECM printed artificial testis is a future milestone that promises for enhancing germ cell maintenance and differentiation, toxicology studies, and fertility restoration to pave the way for new human infertility treatments in the future.
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
| | - 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
| | - Reza Falak
- Immunology Research Center (IRC), Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Iraj Amiri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Endometrium and Research Center, Hamadan University of Medical Sciences, Hamadan, 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
|
12
|
Chakraborty S, Roychoudhury S. Pathological Roles of Reactive Oxygen Species in Male Reproduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1358:41-62. [PMID: 35641865 DOI: 10.1007/978-3-030-89340-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Reactive oxygen species (ROS) are free radicals that have at least one unpaired electron and play specific roles in the human body. An imbalance of ROS and antioxidant levels gives rise to a condition called oxidative stress. High levels of ROS in the male reproductive tract can interfere with its normal functioning and can even pose as toxic to the sperm, inhibiting sperm functioning (including motility) and metabolism. Oxidative stress resulting from ROS and lipid peroxidation is one of the major causes of male infertility including infertility in varicocele patients. These may cause DNA and peroxidative damage and apoptosis. Production of ROS in excess also leads to erectile dysfunction (ED). In recent years, studies have also linked oxidative stress with the development, progress, and therapy response of prostate cancer patients. The present study summarizes the pathological roles of ROS in male reproductive problems such as infertility, ED, and prostate cancer and also provide an insight into the probable mechanism through which ROS exert their pathological impact.
Collapse
|
13
|
Ganjibakhsh M, Mehraein F, Koruji M, Bashiri Z. The therapeutic potential of adipose tissue-derived mesenchymal stromal cells in the treatment of busulfan-induced azoospermic mice. J Assist Reprod Genet 2022; 39:153-163. [PMID: 34519944 PMCID: PMC8866597 DOI: 10.1007/s10815-021-02309-8] [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: 09/12/2020] [Accepted: 08/30/2021] [Indexed: 01/03/2023] Open
Abstract
PURPOSE The generation of germ cells from mesenchymal stromal cells (MSCs) provides a valuable in vitro platform for infertility modeling. The establishment of these cells is a new approach for assisted reproductive technology (ART) to help infertile patients who lack functional gametes. METHODS Human adipose-derived MSCs were isolated and then characterized for multipotency by flow cytometry, differentiation capacity, and cytogenetic assays. These cells were used in a male germ cell differentiation study. The expression of male germ cell markers was evaluated at day 21 of differentiation using an immunofluorescence assay, flow cytometry, and RT-qPCR. Undifferentiated MSCs were used for transplantation in busulfan-induced azoospermic mice. RESULTS In this study, MSCs were successfully isolated from human adipose tissues which were positive for cell markers such as CD90, CD105, CD73, and CD29 but negative for CD34 and CD45. The results of flow cytometry, immunocytochemistry, and RT-qPCR analysis at day 21 of differentiation showed that the undifferentiated adipose-derived MSCs are able to differentiate into male germ cells. Additionally, transplantation of undifferentiated MSCs in busulfan-induced azoospermic mice caused spermatogenesis recovery in the majority of seminiferous tubules. CONCLUSION In this study, we showed that differentiation of human adipose-derived MSCs into male germ cells is a useful tool for in vitro study of human germ cell development. Our results demonstrated that cell therapy with adipose-derived MSCs could help the repair of pathological changes in testicular seminiferous tubules. Therefore, it may have a clinical application for the treatment of azoospermia in infertile patients.
Collapse
Affiliation(s)
- Meysam Ganjibakhsh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Mehraein
- Department of Anatomy, School of Medicine, Iran 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
| | - Zahra Bashiri
- 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
|
14
|
Hajiesmailpoor A, Emami P, Kondori BJ, Ghorbani M. Stem cell therapy as a recent advanced approach in male infertility. Tissue Cell 2021; 73:101634. [PMID: 34481231 DOI: 10.1016/j.tice.2021.101634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023]
Abstract
Infertility is one of the most common problems in the world that has negative effects on society and infertile people. Among the various causes of infertility, male infertility accounts for almost half of all infertility cases. Despite advances in medicine, current male infertility treatments such as assisted reproductive technology (ART) have not been successful in treating all types of male infertility. Recently, stem cells have been considered as therapeutic targets for many diseases, including infertility, due to their self-renewing and high differentiation. The purpose of this review is to discuss different types of male infertility and the effect of various stem cells against the treatment of male infertility.
Collapse
Affiliation(s)
- Ayshe Hajiesmailpoor
- Department of Emergency Medical Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Payam Emami
- Department of Emergency Medical Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Bahman Jalali Kondori
- Department of Anatomical Sciences, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Masoud Ghorbani
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Saha S, Roy P, Corbitt C, Kakar SS. Application of Stem Cell Therapy for Infertility. Cells 2021; 10:1613. [PMID: 34203240 PMCID: PMC8303590 DOI: 10.3390/cells10071613] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Infertility creates an immense impact on the psychosocial wellbeing of affected couples, leading to poor quality of life. Infertility is now considered to be a global health issue affecting approximately 15% of couples worldwide. It may arise from factors related to the male (30%), including varicocele, undescended testes, testicular cancer, and azoospermia; the female (30%), including premature ovarian failure and uterine disorders; or both partners (30%). With the recent advancement in assisted reproduction technology (ART), many affected couples (80%) could find a solution. However, a substantial number of couples cannot conceive even after ART. Stem cells are now increasingly being investigated as promising alternative therapeutics in translational research of regenerative medicine. Tremendous headway has been made to understand the biology and function of stem cells. Considering the minimum ethical concern and easily available abundant resources, extensive research is being conducted on induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSC) for their potential application in reproductive medicine, especially in cases of infertility resulting from azoospermia and premature ovarian insufficiency. However, most of these investigations have been carried out in animal models. Evolutionary divergence observed in pluripotency among animals and humans requires caution when extrapolating the data obtained from murine models to safely apply them to clinical applications in humans. Hence, more clinical trials based on larger populations need to be carried out to investigate the relevance of stem cell therapy, including its safety and efficacy, in translational infertility medicine.
Collapse
Affiliation(s)
- Sarama Saha
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh 249203, India;
| | - Partha Roy
- Department of Biotechnology, Indian Institute of Technology, Roorkee 247667, India;
| | - Cynthia Corbitt
- Department of Biology, University of Louisville, Louisville, KY 40292, USA;
| | - Sham S. Kakar
- Department of Physiology and James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
16
|
Kumar D, Talluri TR, Selokar NL, Hyder I, Kues WA. Perspectives of pluripotent stem cells in livestock. World J Stem Cells 2021; 13:1-29. [PMID: 33584977 PMCID: PMC7859985 DOI: 10.4252/wjsc.v13.i1.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The recent progress in derivation of pluripotent stem cells (PSCs) from farm animals opens new approaches not only for reproduction, genetic engineering, treatment and conservation of these species, but also for screening novel drugs for their efficacy and toxicity, and modelling of human diseases. Initial attempts to derive PSCs from the inner cell mass of blastocyst stages in farm animals were largely unsuccessful as either the cells survived for only a few passages, or lost their cellular potency; indicating that the protocols which allowed the derivation of murine or human embryonic stem (ES) cells were not sufficient to support the maintenance of ES cells from farm animals. This scenario changed by the innovation of induced pluripotency and by the development of the 3 inhibitor culture conditions to support naïve pluripotency in ES cells from livestock species. However, the long-term culture of livestock PSCs while maintaining the full pluripotency is still challenging, and requires further refinements. Here, we review the current achievements in the derivation of PSCs from farm animals, and discuss the potential application areas.
Collapse
Affiliation(s)
- Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India.
| | - Thirumala R Talluri
- Equine Production Campus, ICAR-National Research Centre on Equines, Bikaner 334001, India
| | - Naresh L Selokar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India
| | - Iqbal Hyder
- Department of Physiology, NTR College of Veterinary Science, Gannavaram 521102, India
| | - Wilfried A Kues
- Department of Biotechnology, Friedrich-Loeffler-Institute, Federal Institute of Animal Health, Neustadt 31535, Germany
| |
Collapse
|
17
|
Xu H, Yang M, Tian R, Wang Y, Liu L, Zhu Z, Yang S, Yuan Q, Niu M, Yao C, Zhi E, Li P, Zhou C, He Z, Li Z, Gao WQ. Derivation and propagation of spermatogonial stem cells from human pluripotent cells. Stem Cell Res Ther 2020; 11:408. [PMID: 32967715 PMCID: PMC7509941 DOI: 10.1186/s13287-020-01896-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES This study is designed to generate and propagate human spermatogonial stem cells (SSCs) derived from human pluripotent stem cells (hPSCs). METHODS hPSCs were differentiated into SSC-like cells (SSCLCs) by a three-step strategy. The biological characteristics of SSCLCs were detected by immunostaining with antibodies against SSC markers. The ability of self-renewal was measured by propagating for a long time and still maintaining SSCs morphological property. The differentiation potential of SSCLCs was determined by the generation of spermatocytes and haploid cells, which were identified by immunostaining and flow cytometry. The transcriptome analysis of SSCLCs was performed by RNA sequencing. The biological function of SSCLCs was assessed by xeno-transplantation into busulfan-treated mouse testes. RESULTS SSCLCs were efficiently generated by a 3-step strategy. The SSCLCs displayed a grape-like morphology and expressed SSC markers. Moreover, SSCLCs could be propagated for approximately 4 months and still maintained their morphological properties. Furthermore, SSCLCs could differentiate into spermatocytes and haploid cells. In addition, SSCLCs displayed a similar gene expression pattern as human GPR125+ spermatogonia derived from human testicular tissues. And more, SSCLCs could survive and home at the base membrane of seminiferous tubules. CONCLUSION SSCLCs were successfully derived from hPSCs and propagated for a long time. The SSCLCs resembled their counterpart human GPR125+ spermatogonia, as evidenced by the grape-like morphology, transcriptome, homing, and functional characteristics. Therefore, hPSC-derived SSCLCs may provide a reliable cell source for studying human SSCs biological properties, disease modeling, and drug toxicity screening.
Collapse
Affiliation(s)
- Huiming Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China. .,School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| | - Mengbo Yang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Ruhui Tian
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Yonghui Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Linhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Zijue Zhu
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Shi Yang
- Department of Urology, Shanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
| | - Qingqing Yuan
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Minghui Niu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Chencheng Yao
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Erlei Zhi
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Peng Li
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Chenhao Zhou
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Zuping He
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Zheng Li
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China. .,School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
18
|
Yang M, Deng B, Geng L, Li L, Wu X. Pluripotency factor NANOG promotes germ cell maintenance in vitro without triggering dedifferentiation of spermatogonial stem cells. Theriogenology 2020; 148:68-75. [DOI: 10.1016/j.theriogenology.2020.02.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 02/18/2020] [Accepted: 02/18/2020] [Indexed: 12/25/2022]
|
19
|
Amini Mahabadi J, Karimian M, Aghighi F, Enderami SE, Seyyed Hosseini E, Talaei SA, Gheibi Hayat SM, Nikzad H. Retinoic acid and 17β-estradiol improve male germ cell differentiation from mouse-induced pluripotent stem cells. Andrologia 2019; 52:e13466. [PMID: 31736115 DOI: 10.1111/and.13466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/04/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022] Open
Abstract
This research aimed to explore the impacts of retinoic acid (RA)/17β-estradiol (E) induction and embryoid body formation to enhance differentiation of mouse-induced pluripotent stem cells (miPSCs) into male germ cells in vitro. Flow cytometry and qPCR were conducted to describe miPSCs differentiation process. Various temporal expression profiles of germ cell-related genes were traced. Stra8 gene expression increased in the RA group on the 4th day compared to other groups. The RA group experienced a more significant increase than E group. The expression of Sycp3 increased in RA + E group on 4th day compared with other groups. Expression of AKAP3 enhanced in the RA + E group than other groups on day 4. Moreover, miPSCs showed that this gene expression in the RA + E group was increased in comparison to RA and E groups on day 7. AKAP3 gene expression on day 7 of miPSCs decreased in RA and E groups. Flow cytometry data indicated that 3%-8% of the cells in sub-G1 stage were haploid after RA and E induction compared to other groups on day 4. This study showed that miPSCs possess the power for differentiating into male germ cells in vitro via formation of embryoid body by RA with/or E induction.
Collapse
Affiliation(s)
- Javad Amini Mahabadi
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Karimian
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran.,Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Fatemeh Aghighi
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Ehsan Enderami
- Department of Medical Biotechnology, Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Sayyed Alireza Talaei
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Gheibi Hayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Nikzad
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
20
|
Three-dimensional decellularized amnion membrane scaffold promotes the efficiency of male germ cells generation from human induced pluripotent stem cells. Exp Cell Res 2019; 384:111544. [DOI: 10.1016/j.yexcr.2019.111544] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/21/2019] [Accepted: 08/01/2019] [Indexed: 12/30/2022]
|
21
|
Mahabadi JA, Tameh AA, Talaei SA, Karimian M, Rahiminia T, Enderami SE, Gheibi Hayat SM, Nikzad H. Retinoic acid and/or progesterone differentiate mouse induced pluripotent stem cells into male germ cells in vitro. J Cell Biochem 2019; 121:2159-2169. [PMID: 31646671 DOI: 10.1002/jcb.29439] [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: 05/16/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022]
Abstract
Numerous reagents were employed for differentiating induced pluripotent stem cells (iPSCs) into male germ cells; however, the induction procedure was ineffective. The aim of this study was to improve the in vitro differentiation of mice iPSCs (miPSCs) into male germ cells with retinoic acid (RA) and progesterone (P). miPSCs were differentiated to embryoid bodies (EBs) in suspension with RA with or without progesterone for 0, 4, and 7 days. Then, the expression of certain genes at different stages of male germ cell development including Ddx4 (pre meiosis), Stra8 (meiosis), AKAP3 (post meiosis), and Mvh protein was examined in RNA and/or protein levels by real-time polymerase chain reaction or flow cytometry, respectively. The Stra8 gene expression increased in the RA groups on all days. But, expression of this gene declined in RA + P groups. In addition, an increased expression of Ddx4 gene was observed on day 0 in the P group. Also, a significant upregulation was observed in the expression of AKAP3 gene in the RA + P group on days 0 and 4. However, gene expression decreased in P and RA groups on day 7. The expression of Mvh protein significantly increased in the RA group on day 7. The Mvh expression was also enhanced in the P group on day 4, but it decreased on day 7, while this protein upregulated on day 0 and 7 in the RA + P group. The miPSCs have the capacity for in vitro differentiation into male germ cells by RA and/or progesterone. However, the effects of these inducers depend on the type of combination and an effective time.
Collapse
Affiliation(s)
- Javad Amini Mahabadi
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Abolfazl Aazami Tameh
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Mohammad Karimian
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Tahereh Rahiminia
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Ehsan Enderami
- Department of Medical Biotechnology, Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Gheibi Hayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Nikzad
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
22
|
Wang J, Liu C, Fujino M, Tong G, Zhang Q, Li XK, Yan H. Stem Cells as a Resource for Treatment of Infertility-related Diseases. Curr Mol Med 2019; 19:539-546. [PMID: 31288721 PMCID: PMC6806537 DOI: 10.2174/1566524019666190709172636] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 06/11/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022]
Abstract
Worldwide, infertility affects 8-12% of couples of reproductive age and has become a common problem. There are many ways to treat infertility, including medication, intrauterine insemination, and in vitro fertilization. In recent years, stem-cell therapy has raised new hope in the field of reproductive disability management. Stem cells are self-renewing, self-replicating undifferentiated cells that are capable of producing specialized cells under appropriate conditions. They exist throughout a human’s embryo, fetal, and adult stages and can proliferate into different cells. While many issues remain to be addressed concerning stem cells, stem cells have undeniably opened up new ways to treat infertility. In this review, we describe past, present, and future strategies for the use of stem cells in reproductive medicine
Collapse
Affiliation(s)
- Jing Wang
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Chi Liu
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Guoqing Tong
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qinxiu Zhang
- Department of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hua Yan
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
23
|
Determining Influence of Culture Media and Dose-Dependent Supplementation with Basic Fibroblast Growth Factor on the Ex Vivo Proliferative Activity of Domestic Cat Dermal Fibroblasts in Terms of Their Suitability for Cell Banking and Somatic Cell Cloning of Felids. ANNALS OF ANIMAL SCIENCE 2019. [DOI: 10.2478/aoas-2018-0057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Abstract
Dermal fibroblasts are commonly used as donors of genetic material for somatic cell nuclear transfer in mammals. Basic fibroblast growth factor (bFGF) is a cytokine that regulates proliferation and differentiation of different cell types. The study was aimed at optimizing the cell culture protocol for cat dermal fibroblasts by assessing the influence of culture media and different doses of bFGF on proliferation of fibroblasts and their viability in terms of cell banking and somatic cloning of felids. In Experiment I, skin biopsies of domestic cats were cultured in DMEM (D) and/or DMEM/F12 (F), both supplemented with 5 ng bFGF/ml (D-5, F-5, respectively). After the primary culture reached ~80% of confluency, the cells were passaged (3–4 times) and cultured in media with (D-5, F-5) or without (D-0, F-0) bFGF. To determine the optimal doses of bFGF, in Experiment II, secondary fibroblasts were cultured in DMEM with 0 (D-0), 2.5 (D-2.5), 5 (D-5) or 10 (D-10) ng bFGF/ml. The results showed that in D-5 the cells proliferated faster than in D-0, F-5 and F-0. Due to their poor proliferation, passages IV were not performed for cells cultured in F-0. In experiment II, a dose-dependent effect of bFGF on proliferation of cat dermal fibroblasts was found. In D-5 and D-10, the cells exhibited higher (P<0.05) proliferation compared with D-0. In D-2.5 the cells showed a tendency to proliferate slower than in D-5 and D-10 and at the same faster than in D-0. In conclusion. DMEM supplemented with bFGF provides better proliferation of domestic cat dermal fibroblasts culture than DMEM/F12. Supplementation of culture medium with bFGF has a beneficial effect on cat dermal fibroblast proliferation and could be recommended for addition to culture media.
Collapse
|
24
|
Fang F, Li Z, Zhao Q, Li H, Xiong C. Human induced pluripotent stem cells and male infertility: an overview of current progress and perspectives. Hum Reprod 2019; 33:188-195. [PMID: 29315416 PMCID: PMC5850345 DOI: 10.1093/humrep/dex369] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Indexed: 01/30/2023] Open
Abstract
Recently, significant progress has been made in ART for the treatment of male infertility. However, current ART has failed to help infertile patients with non-obstructive azoospermia, unless donor sperm is used. In fact, most couples wish to have their own genetically related child. Human induced pluripotent stem cells (hiPSCs) can be generated from patients’ somatic cells and in vitro derivation of functional germ cells from patient-specific iPSCs may provide new therapeutic strategies for infertile couples. The overall developmental dynamics of human primordial germ cells are similar to that in mice, but accumulating evidence suggests that there are crucial differences between human and mouse PGC specification. Unlike mouse iPSCs (miPSCs) in naive state, hiPSCs exhibit a primed pluripotency which possess less potential for the germ cell fate. Based on research in mice, male germ cells at different stages have been derived from hiPSCs with different protocols, including spontaneous differentiation, overexpression of germ cell regulators, addition of cytokines, co-culture with gonadal cells in vitro and xeno-transplantation. The aim of this review is to summarize the current advances in derivation of male germ cells from hiPSCs and raise the perspectives of hiPSCs in medical application for male infertility, as well as in basic research for male germ cell development.
Collapse
Affiliation(s)
- Fang Fang
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Zili Li
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.,Center for Reproductive Medicine, Wuhan Tongji Reproductive Medicine Hospital, 128 Sanyang Road, Wuhan 430013, China
| | - Qian Zhao
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Honggang Li
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Chengliang Xiong
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.,Center for Reproductive Medicine, Wuhan Tongji Reproductive Medicine Hospital, 128 Sanyang Road, Wuhan 430013, China
| |
Collapse
|
25
|
Ghorbanlou M, Abdanipour A, Shirazi R, Malekmohammadi N, Shokri S, Nejatbakhsh R. Indirect Co-Culture of Testicular Cells with Bone Marrow Mesenchymal Stem Cells Leads to Male Germ Cell-Specific Gene Expressions. CELL JOURNAL 2018; 20:505-512. [PMID: 30123996 PMCID: PMC6099144 DOI: 10.22074/cellj.2019.5654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 02/07/2017] [Indexed: 01/01/2023]
Abstract
Objective Non-obstructive azoospermia is mostly irreversible. Efforts to cure this type of infertility have led to the application
of stem cells in the reproduction field. In the present study, testicular cell-mediated differentiation of male germ-like cells from
bone marrow-derived mesenchymal stem cells (BM-MSCs) in an in vitro indirect co-culture system is investigated.
Materials and Methods In this experimental study, mouse BM-MSCs were isolated and cultured up to passage three.
Identification of the cells was evaluated using specific surface markers by flow-cytometry technique. Four experimental groups
were investigated: control, treatment with retinoic acid (RA), indirect co-culture with testicular cells, and combination of RA
and indirect co-culture with testicular cells. Finally, following differentiation, the quantitative expression of germ cell-specific
markers including Dazl, Piwil2 and Stra8 were evaluated by real-time polymerase chain reaction (PCR).
Results Molecular analysis revealed a significant increase in Dazl expression in the indirect co-culture with testicular
cells group in comparison to the control group. Quantitative expression level of Piwil2 was not significantly changed in
comparison to the control group. Stra8 expression was significantly higher in RA group in comparison to other groups.
Conclusion Indirect co-culture of BM-MSCs in the presence of testicular cells leads to expression of male germ cell-specific
gene, Dazl, in the induced cells. Combination of co-culture with testicular cells and RA did not show any positive effect on the
specific gene expressions.
Collapse
Affiliation(s)
- Mehrdad Ghorbanlou
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Alireza Abdanipour
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran. Electronic Address:
| | - Reza Shirazi
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nasim Malekmohammadi
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saeed Shokri
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Nejatbakhsh
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran. Electronic Address:
| |
Collapse
|
26
|
Mahabadi JA, Sabzalipour H, Bafrani HH, Gheibi Hayat SM, Nikzad H. Application of induced pluripotent stem cell and embryonic stem cell technology to the study of male infertility. J Cell Physiol 2018; 233:8441-8449. [PMID: 29870061 DOI: 10.1002/jcp.26757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/23/2018] [Indexed: 01/18/2023]
Abstract
Stem cells (SCs) are classes of undifferentiated biological cells existing only at the embryonic, fetal, and adult stages that can divide to produce specialized cell types during fetal development and remain in our bodies throughout life. The progression of regenerative and reproductive medicine owes the advancement of respective in vitro and in vivo biological science on the stem cell nature under appropriate conditions. The SCs are promising therapeutic tools to treat currently of infertility because of wide sources and high potency to differentiate. Nevertheless, no effective remedies are available to deal with severe infertility due to congenital or gonadotoxic stem cell deficiency in prepubertal childhood. Some recent solutions have been developed to address the severe fertility problems, including in vitro formation of germ cells from stem cells, induction of pluripotency from somatic cells, and production of patient-specific pluripotent stem cells. There is a possibility of fertility restoration using the in vitro formation of germ cells from somatic cells. Accordingly, the present review aimed at studying the literature published on the medical application of stem cells in reproductive concerns.
Collapse
Affiliation(s)
- Javad Amini Mahabadi
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Sabzalipour
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Seyed Mohammad Gheibi Hayat
- Student Research Committee, Department of Medical Biotechnology, Faculty Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Nikzad
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
27
|
Tunstall T, Kock R, Vahala J, Diekhans M, Fiddes I, Armstrong J, Paten B, Ryder OA, Steiner CC. Evaluating recovery potential of the northern white rhinoceros from cryopreserved somatic cells. Genome Res 2018; 28:780-788. [PMID: 29798851 PMCID: PMC5991516 DOI: 10.1101/gr.227603.117] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 04/26/2018] [Indexed: 01/04/2023]
Abstract
The critically endangered northern white rhinoceros is believed to be extinct in the wild, with the recent death of the last male leaving only two remaining individuals in captivity. Its extinction would appear inevitable, but the development of advanced cell and reproductive technologies such as cloning by nuclear transfer and the artificial production of gametes via stem cells differentiation offer a second chance for its survival. In this work, we analyzed genome-wide levels of genetic diversity, inbreeding, population history, and demography of the white rhinoceros sequenced from cryopreserved somatic cells, with the goal of informing how genetically valuable individuals could be used in future efforts toward the genetic rescue of the northern white rhinoceros. We present the first sequenced genomes of the northern white rhinoceros, which show relatively high levels of heterozygosity and an average genetic divergence of 0.1% compared with the southern subspecies. The two white rhinoceros subspecies appear to be closely related, with low genetic admixture and a divergent time <80,000 yr ago. Inbreeding, as measured by runs of homozygosity, appears slightly higher in the southern than the northern white rhinoceros. This work demonstrates the value of the northern white rhinoceros cryopreserved genetic material as a potential gene pool for saving this subspecies from extinction.
Collapse
Affiliation(s)
- Tate Tunstall
- San Diego Zoo Institute for Conservation Research, Escondido, California 92027, USA
| | - Richard Kock
- Royal Veterinary College, University of London, London NW1 0TU, United Kingdom
| | - Jiri Vahala
- Dvur Krlov Zoo, Dvr Krlov nad Labem 544 01, Czech Republic
| | - Mark Diekhans
- Jack Baskin School of Engineering, University California Santa Cruz, Santa Cruz, California 95064, USA
| | - Ian Fiddes
- Jack Baskin School of Engineering, University California Santa Cruz, Santa Cruz, California 95064, USA
| | - Joel Armstrong
- Jack Baskin School of Engineering, University California Santa Cruz, Santa Cruz, California 95064, USA
| | - Benedict Paten
- Jack Baskin School of Engineering, University California Santa Cruz, Santa Cruz, California 95064, USA
| | - Oliver A Ryder
- San Diego Zoo Institute for Conservation Research, Escondido, California 92027, USA
| | - Cynthia C Steiner
- San Diego Zoo Institute for Conservation Research, Escondido, California 92027, USA
| |
Collapse
|
28
|
Pourmoghadam Z, Aghebati‐Maleki L, Motalebnezhad M, Yousefi B, Yousefi M. Current approaches for the treatment of male infertility with stem cell therapy. J Cell Physiol 2018; 233:6455-6469. [DOI: 10.1002/jcp.26577] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 02/27/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Zahra Pourmoghadam
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
- Immunology Research Center Tabriz University of Medical Sciences Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Leili Aghebati‐Maleki
- Immunology Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| | | | - Bahman Yousefi
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Mehdi Yousefi
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
29
|
Amini Mahabadi J, Sabzalipoor H, Kehtari M, Enderami SE, Soleimani M, Nikzad H. Derivation of male germ cells from induced pluripotent stem cells by inducers: A review. Cytotherapy 2018; 20:279-290. [PMID: 29397308 DOI: 10.1016/j.jcyt.2018.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 11/15/2017] [Accepted: 01/01/2018] [Indexed: 12/29/2022]
Abstract
Induced pluripotent stem cells (iPSCs) refer to stem cells that are artificially produced using a new technology known as cellular reprogramming, which can use gene transduction in somatic cells. There are numerous potential applications for iPSCs in the field of stem cell biology becauase they are able to give rise to several different cell features of lineages such as three-germ layers. Primordial germ cells, generated via in vitro differentiation of iPSCs, have been demonstrated to produce functional gametes. Therefore, in this review we discussed past and recent advances in the in vitro differentiation of germ cells using pluripotent stem cells with an emphasis on iPSCs. Although this domain of research is still in its infancy, exploring development mechanisms of germ cells is promising, especially in humans, to promote future reproductive and developmental engineering technologies. While few studies have evaluated the ability and efficiency of iPSCs to differentiate toward male germ cells in vitro by different inducers, the given effect was investigated in this review.
Collapse
Affiliation(s)
- Javad Amini Mahabadi
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Sabzalipoor
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mousa Kehtari
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Seyed Ehsan Enderami
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Masoud Soleimani
- Hematology Department, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Nikzad
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
30
|
Vij SC, Agarwal A. Editorial on "An automated smartphone-based diagnostic assay for point-of-care semen analysis". ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:507. [PMID: 29299468 DOI: 10.21037/atm.2017.09.12] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Sarah Coleman Vij
- Department of Urology, Glickman Urologic and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Ashok Agarwal
- Department of Urology, Glickman Urologic and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
31
|
Retinoic acid induces differentiation of buffalo (Bubalus bubalis) embryonic stem cells into germ cells. Gene 2017; 631:54-67. [DOI: 10.1016/j.gene.2017.07.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
32
|
Vij SC, Sabanegh E, Agarwal A. Biological therapy for non-obstructive azoospermia. Expert Opin Biol Ther 2017; 18:19-23. [DOI: 10.1080/14712598.2018.1380622] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sarah C. Vij
- Glickman Urologic and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Edmund Sabanegh
- Department of Urology, Glickman Urologic and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Ashok Agarwal
- Andrology Laboratory, Glickman Urologic and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
33
|
Rawat N, Singh MK. Induced pluripotent stem cell: A headway in reprogramming with promising approach in regenerative biology. Vet World 2017; 10:640-649. [PMID: 28717316 PMCID: PMC5499081 DOI: 10.14202/vetworld.2017.640-649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 04/26/2017] [Indexed: 12/17/2022] Open
Abstract
Since the embryonic stem cells have knocked the doorsteps, they have proved themselves in the field of science, research, and medicines, but the hovered restrictions confine their application in human welfare. Alternate approaches used to reprogram the cells to the pluripotent state were not up to par, but the innovation of induced pluripotent stem cells (iPSCs) paved a new hope for the researchers. Soon after the discovery, iPSCs technology is undergoing renaissance day by day, i.e., from the use of genetic material to recombinant proteins and now only chemicals are employed to convert somatic cells to iPSCs. Thus, this technique is moving straightforward and productive at an astonishing pace. Here, we provide a brief introduction to iPSCs, the mechanism and methods for their generation, their prevailing and prospective applications and the future opportunities that can be expected from them.
Collapse
Affiliation(s)
- N Rawat
- Embryo Biotechnology Lab, Animal Biotechnology Centre, ICAR - National Dairy Research Institute, Karnal - 132 001, Haryana, India
| | - M K Singh
- Embryo Biotechnology Lab, Animal Biotechnology Centre, ICAR - National Dairy Research Institute, Karnal - 132 001, Haryana, India
| |
Collapse
|
34
|
Retinoic acid induces differentiation of buffalo (Bubalus bubalis) embryonic stem cells into germ cells. Gene 2017; 626:358-366. [PMID: 28526652 DOI: 10.1016/j.gene.2017.05.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/27/2017] [Accepted: 05/15/2017] [Indexed: 01/12/2023]
Abstract
Development of precise and reproducible culture system for in vitro differentiation of embryonic stem (ES) cells into germ cells counts as a major leap forward for understanding not only the remarkable process of gametogenesis, otherwise obscured by limited availability of precursor primordial germ cells (PGCs), but in finally treating the catastrophic infertility. Taking into account the significant role of retinoic acid (RA) during in vivo gametogenesis, we designed the present study to investigate the effects of its stimulation on directing the differentiation of ES cells into germ cells. The effects of RA were analyzed across dose-and-time upon various stages of gametogenesis like PGC induction, meiosis initiation and completion, haploid cell formation and development of the final gamete (oocyte and spermatozoa). Out of the series of RA doses (2, 4, 8, 16, 20 and 30μM), 16μM RA for 8day culture interval was found to induce highest expression of PGC- and meiosis-associated genes like DAZL, VASA, SYCP3, MLH1, TNP1/2 and PRM2, while mature germ cell genes like BOULE and TEKT1 (Spermatocyte markers), GDF9 and ZP2 (Oocyte markers) showed higher expression at 2μM RA dose, suggesting functional concentration-gradient of RA activity. Immunocytochemistry revealed expression of germ lineage-specific markers like: c-KIT, DAZL and VASA (PGC-markers); SYCP3, MLH1 and PROTAMINE1 (Meiotic-markers); ACROSIN and HAPRIN (Spermatocyte-markers); and GDF9 and ZP4 (Oocyte-markers) in optimally differentiated embryoid bodies (EBs) and adherent cultures. We observed significantly reduced (p<0.05) concentration of 5-methyl-2-deoxycytidine in RA-differentiated EBs which is suggestive of the occurrence of methylation erasure. FACS analysis of optimally differentiated cultures detected 3.07% haploid cell population, indicating completion of meiosis. Oocyte-like structures (OLS) were obtained in adherent differentiated cultures. They had a big nucleus and a zona pellucida (ZP4) coat. They showed progression through 2-cell, 4-cell, 8-cell, morula and blastocyst-like structures upon extended culture beyond 14days.
Collapse
|
35
|
Mouka A, Tachdjian G, Dupont J, Drévillon L, Tosca L. In Vitro Gamete Differentiation from Pluripotent Stem Cells as a Promising Therapy for Infertility. Stem Cells Dev 2016; 25:509-21. [PMID: 26873432 DOI: 10.1089/scd.2015.0230] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Generation of gametes derived in vitro from pluripotent stem cells holds promising prospects for future reproductive applications. Indeed, it provides information on molecular and cellular mechanisms underlying germ cell (GC) development and could offer a new potential treatment for infertility. Great progress has been made in derivation of gametes from embryonic stem cells, despite ethical issues. Induced pluripotent stem cells (iPSCs) technology allows the reprogramming of a differentiated somatic cell, possibly emanating from the patient, into a pluripotent state. With the emergence of iPSCs, several studies created primordial GC stage to mature gamete-like cells in vitro in mice and humans. Recent findings in GC derivation suggest that in mice, functional gametes can be generated in vitro. This strengthens the idea that it might be possible in the future to generate functional human sperm and oocytes from pluripotent stem cells in culture.
Collapse
Affiliation(s)
- Aurélie Mouka
- 1 AP-HP, Service d'Histologie, Embryologie et Cytogénétique, Hôpitaux Universitaires Paris-Sud , Clamart, France .,2 Université Paris-Sud , Le Kremlin-Bicêtre Cedex, France
| | - Gérard Tachdjian
- 1 AP-HP, Service d'Histologie, Embryologie et Cytogénétique, Hôpitaux Universitaires Paris-Sud , Clamart, France .,2 Université Paris-Sud , Le Kremlin-Bicêtre Cedex, France
| | - Joëlle Dupont
- 3 Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique , Nouzilly, France
| | - Loïc Drévillon
- 1 AP-HP, Service d'Histologie, Embryologie et Cytogénétique, Hôpitaux Universitaires Paris-Sud , Clamart, France
| | - Lucie Tosca
- 1 AP-HP, Service d'Histologie, Embryologie et Cytogénétique, Hôpitaux Universitaires Paris-Sud , Clamart, France .,2 Université Paris-Sud , Le Kremlin-Bicêtre Cedex, France
| |
Collapse
|
36
|
Rahmanifar F, Tamadon A, Mehrabani D, Zare S, Abasi S, Keshavarz S, Dianatpour M, Khodabandeh Z, Jahromi IRG, Koohi-Hoseinabadi O. Histomorphometric evaluation of treatment of rat azoosper-mic seminiferous tubules by allotransplantation of bone marrow-derived mesenchymal stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:653-61. [PMID: 27482347 PMCID: PMC4951605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVES Bone marrow-derived mesenchymal stem cells (BM-MSCs) potentials make them appropriate for cell therapy including ability of differentiation and release of anti-inflammatory cytokines and growth factors secreta. For treatment of azoospermia to induce proliferation and differentiation of germ cells, MSCs transplantation has been introduced. The aim of the present experimental case-control study was to histomorphometric evaluation of the germinal cells in seminiferous tubules of azoospermic rats before and after BM-MSCs allotransplantation. MATERIALS AND METHODS In the present study, BM-MSCs were isolated from six male rats and confirmed. Their testes also served as intact negative controls. The recipient rats (n=6) were received two doses of 10 mg/kg of busulfan with 21 days interval to induce azoospermia. After cessation of spermatogenesis, the rats were allotransplanted with the BM-MSCs into efferent duct of right testes. Thirty-five days later, the right cell-treated testes were compared to left azoospermic ones. RESULTS Histomorphometric analyses showed that the seminiferous tubules treated with BM-MSCs had normal morphology in comparison with azoospermic testes, which were without germinal layer. In most BM-MSCs-treated seminiferous tubules, spermatogenesis was observed. CONCLUSION The allotransplanted BM-MSCs could induce spermatogenesis in seminiferous tubules of azoospermic rats.
Collapse
Affiliation(s)
- Farhad Rahmanifar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amin Tamadon
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Davood Mehrabani
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,Corresponding author: Davood Mehrabani. Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. Tel/Fax: +98-71 32341025; emails:
| | - Shahrokh Zare
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sorush Abasi
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeideh Keshavarz
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khodabandeh
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | |
Collapse
|
37
|
Saragusty J, Diecke S, Drukker M, Durrant B, Friedrich Ben-Nun I, Galli C, Göritz F, Hayashi K, Hermes R, Holtze S, Johnson S, Lazzari G, Loi P, Loring JF, Okita K, Renfree MB, Seet S, Voracek T, Stejskal J, Ryder OA, Hildebrandt TB. Rewinding the process of mammalian extinction. Zoo Biol 2016; 35:280-92. [PMID: 27142508 DOI: 10.1002/zoo.21284] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/11/2016] [Indexed: 12/15/2022]
Abstract
With only three living individuals left on this planet, the northern white rhinoceros (Ceratotherium simum cottoni) could be considered doomed for extinction. It might still be possible, however, to rescue the (sub)species by combining novel stem cell and assisted reproductive technologies. To discuss the various practical options available to us, we convened a multidisciplinary meeting under the name "Conservation by Cellular Technologies." The outcome of this meeting and the proposed road map that, if successfully implemented, would ultimately lead to a self-sustaining population of an extremely endangered species are outlined here. The ideas discussed here, while centered on the northern white rhinoceros, are equally applicable, after proper adjustments, to other mammals on the brink of extinction. Through implementation of these ideas we hope to establish the foundation for reversal of some of the effects of what has been termed the sixth mass extinction event in the history of Earth, and the first anthropogenic one. Zoo Biol. 35:280-292, 2016. © 2016 The Authors. Zoo Biology published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joseph Saragusty
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | | | - Micha Drukker
- Institute of Stem Cell Research, German Research Center for Environmental Health, Helmholtz Center Munich, Neuherberg, Germany
| | - Barbara Durrant
- San Diego Zoo Institute for Conservation Research, Escondido, California
| | - Inbar Friedrich Ben-Nun
- Department of Chemical Physiology, Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California
| | - Cesare Galli
- Avantea srl, Laboratorio di Tecnologie della Riproduzione, Cremona, Italy.,Dipartimento Scienze Mediche Veterinarie, Università di Bologna, Ozzano dell'Emilia, Italy.,Fondazione Avantea, Cremona, Italy
| | - Frank Göritz
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Katsuhiko Hayashi
- Faculty of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Robert Hermes
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Susanne Holtze
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | | | - Giovanna Lazzari
- Avantea srl, Laboratorio di Tecnologie della Riproduzione, Cremona, Italy.,Fondazione Avantea, Cremona, Italy
| | - Pasqualino Loi
- Faculty of Veterinary Medicine, Univeristy of Teramo, Campus Coste San Agostino, Teramo, Italy
| | - Jeanne F Loring
- Department of Chemical Physiology, Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California
| | - Keisuke Okita
- Center for iPS Cell Research and Application, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Steven Seet
- The Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | | | - Jan Stejskal
- ZOO Dvůr Králové, Dvůr Králové nad Labem, Czech Republic
| | - Oliver A Ryder
- San Diego Zoo Institute for Conservation Research, Escondido, California
| | | |
Collapse
|
38
|
Chen Z, Li Z, He Z. Plasticity of male germline stem cells and their applications in reproductive and regenerative medicine. Asian J Androl 2016; 17:367-72. [PMID: 25532577 PMCID: PMC4430934 DOI: 10.4103/1008-682x.143739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Spermatogonial stem cells (SSCs), also known as male germline stem cells, are a small subpopulation of type A spermatogonia with the potential of self-renewal to maintain stem cell pool and differentiation into spermatids in mammalian testis. SSCs are previously regarded as the unipotent stem cells since they can only give rise to sperm within the seminiferous tubules. However, this concept has recently been challenged because numerous studies have demonstrated that SSCs cultured with growth factors can acquire pluripotency to become embryonic stem-like cells. The in vivo and in vitro studies from peers and us have clearly revealed that SSCs can directly transdifferentiate into morphologic, phenotypic, and functional cells of other lineages. Direct conversion to the cells of other tissues has important significance for regenerative medicine. SSCs from azoospermia patients could be induced to differentiate into spermatids with fertilization and developmental potentials. As such, SSCs could have significant applications in both reproductive and regenerative medicine due to their unique and great potentials. In this review, we address the important plasticity of SSCs, with focuses on their self-renewal, differentiation, dedifferentiation, transdifferentiation, and translational medicine studies.
Collapse
Affiliation(s)
| | | | - Zuping He
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Cancer, Shanghai 200127; Department of Urology, Shanghai Human Sperm Bank, Shanghai Institute of Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001; Shanghai Key Laboratory of Assisted Reproduction and Reproductive Genetics, Shanghai 200127; Shanghai Key Laboratory of Reproductive Medicine, Shanghai 200025, China
| |
Collapse
|
39
|
Abstract
Germ cells are the only cell type in the body that can transfer genetic information to the next generation. Germline-competent stem cells can self-renew and contribute to the germ cell lineage giving rise to pluripotent stem cells under specific conditions. Hence far, studies on germline-competent stem cells have contributed to the generation of avian model systems and the conservation of avian genetic resources. In this review, we focus on previous studies on germline-competent stem cells from avian species, mainly chicken germline-competent stem cells, which have been well established and characterized. We discuss different sources of germline-competent stem cells and recent advances for the future applications in birds.
Collapse
Affiliation(s)
- Jae Yong Han
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul-151 921, Korea
| | | | | |
Collapse
|
40
|
Tamadon A, Mehrabani D, Rahmanifar F, Jahromi AR, Panahi M, Zare S, Khodabandeh Z, Jahromi IR, Tanideh N, Dianatpour M, Ramzi M, Koohi-Hoseinabadi O. Induction of Spermatogenesis by Bone Marrow-derived Mesenchymal Stem Cells in Busulfan-induced Azoospermia in Hamster. Int J Stem Cells 2015; 8:134-45. [PMID: 26634062 PMCID: PMC4651278 DOI: 10.15283/ijsc.2015.8.2.134] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Bone marrow-derived mesenchymal stem cells (BM-MSCs) have potential of differentiation and they secrete anti-inflammatory cytokines and growth factors which make them appropriate for cell therapy. Aim of the Work Were to evaluate the healing effect of BM-MSCs transplantation on germinal cells of busulfan-induced azoospermic hamsters. Material and Methods In the present experimental case control study, BM-MSCs were isolated from bone marrow of donor albino hamsters. Five mature male recipient hamsters received two doses of 10 mg/kg of busulfan with 21 days interval to stop endogenous spermatogenesis. After induction of azoospermia, right testis of hamsters was injected with 106 BM-MSCs via efferent duct and the left one remained as azoospermia control testis. Five normal mature hamsters were selected as normal intact control. After 35 days, testes and epididymis of three groups were removed for histological evaluation. Results Histomorphological analyses of BM-MSCs treated testes and epididymis showed the epithelial tissue of seminiferous tubules had normal morphology and spermatozoa were present in epididymis tubes. Spermatogenesis was observed in most cell-treated seminiferous tubules. The untreated seminiferous tubules were empty. Conclusion Transplanted BM-MSCs could successfully induce spermatogenesis in seminiferous tubules of azoospermic hamster. Therefore, BM-MSCs can be an attractive candidate in cell transplantation of azoospermia.
Collapse
Affiliation(s)
- Amin Tamadon
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Davood Mehrabani
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Rahmanifar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Alireza Raayat Jahromi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mohadeseh Panahi
- DVM graduated, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Shahrokh Zare
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khodabandeh
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Razeghian Jahromi
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Tanideh
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ; Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ; Department of Human Genetic, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mani Ramzi
- Hematology and Bone Marrow Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
41
|
Ge W, Chen C, De Felici M, Shen W. In vitro differentiation of germ cells from stem cells: a comparison between primordial germ cells and in vitro derived primordial germ cell-like cells. Cell Death Dis 2015; 6:e1906. [PMID: 26469955 PMCID: PMC4632295 DOI: 10.1038/cddis.2015.265] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 01/15/2023]
Abstract
Stem cells are unique cell types capable to proliferate, some of them indefinitely, while maintaining the ability to differentiate into a few or any cell lineages. In 2003, a group headed by Hans R. Schöler reported that oocyte-like cells could be produced from mouse embryonic stem (ES) cells in vitro. After more than 10 years, where have these researches reached? Which are the major successes achieved and the problems still remaining to be solved? Although during the last years, many reviews have been published about these topics, in the present work, we will focus on an aspect that has been little considered so far, namely a strict comparison between the in vitro and in vivo developmental capabilities of the primordial germ cells (PGCs) isolated from the embryo and the PGC-like cells (PGC-LCs) produced in vitro from different types of stem cells in the mouse, the species in which most investigation has been carried out. Actually, the formation and differentiation of PGCs are crucial for both male and female gametogenesis, and the faithful production of PGCs in vitro represents the basis for obtaining functional germ cells.
Collapse
Affiliation(s)
- W Ge
- Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - C Chen
- Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - M De Felici
- Department of Biomedicine and Prevention, University of Rome ‘Tor Vergata', Rome 00133, Italy
| | - W Shen
- Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
42
|
Mehrabani D, Hassanshahi MA, Tamadon A, Zare S, Keshavarz S, Rahmanifar F, Dianatpour M, Khodabandeh Z, Jahromi I, Tanideh N, Ramzi M, Aqababa H, Kuhi-Hoseinabadi O. Adipose tissue-derived mesenchymal stem cells repair germinal cells of seminiferous tubules of busulfan-induced azoospermic rats. J Hum Reprod Sci 2015; 8:103-10. [PMID: 26157302 PMCID: PMC4477447 DOI: 10.4103/0974-1208.158618] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/15/2015] [Accepted: 05/19/2015] [Indexed: 12/24/2022] Open
Abstract
CONTEXT: Adipose tissue-derived mesenchymal stem cells (AT-MSCs) are less invasive than bone marrow mesenchymal stem cells to obtain for cell therapy. AIMS: The aims of this study were to evaluate the germinal cells characteristics and repairs in seminiferous tubules of busulfan-induced azoospermic rats after AT-MSCs transplantation. SETTINGS AND DESIGN: Experimental case-control study. MATERIALS AND METHODS: In the present experimental study, donors AT-MSCs were isolated from subcutaneous adipose tissue of two Sprague-Dawley rats. The recipients (n = 5) were received two doses of 10 mg/kg of busulfan with 21 days interval to stop endogenous spermatogenesis. After induction of azoospermia by busulfan, rats were injected with the AT-MSCs into the efferent duct of right testes. After 60 days, the right testes were injected AT-MSCs were compared to left azoospermic testes. Five untreated male rats served as negative control. STATISTICAL ANALYSIS USED: Stereological indices were analyzed by one-way ANOVA and LSD post-hoc test. The spermatogenesis index was compared using Mann–Whitney U test. RESULTS: After stereological analyses, the seminiferous tubules treated with AT-MSCs had normal morphology. The untreated seminiferous tubules were empty. Spermatogenesis was observed in most cell-treated seminiferous tubules. CONCLUSIONS: The testis of busulfan-induced azoospermic rats accepted transplanted AT-MSCs. The transplanted AT-MSCs could induce spermatogenesis in seminiferous tubules of the rat.
Collapse
Affiliation(s)
- Davood Mehrabani
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Amin Hassanshahi
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Tamadon
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrokh Zare
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeideh Keshavarz
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Rahmanifar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ; Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khodabandeh
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - ImanRazeghian Jahromi
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Tanideh
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mani Ramzi
- Hematology and Bone Marrow Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Heydar Aqababa
- Department of Biology, Islamic Azad University, Arsanjan Branch, Arsanjan, Iran
| | | |
Collapse
|
43
|
Kumar D, Talluri TR, Anand T, Kues WA. Induced pluripotent stem cells: Mechanisms, achievements and perspectives in farm animals. World J Stem Cells 2015; 7:315-328. [PMID: 25815117 PMCID: PMC4369489 DOI: 10.4252/wjsc.v7.i2.315] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/19/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Pluripotent stem cells are unspecialized cells with unlimited self-renewal, and they can be triggered to differentiate into desired specialized cell types. These features provide the basis for an unlimited cell source for innovative cell therapies. Pluripotent cells also allow to study developmental pathways, and to employ them or their differentiated cell derivatives in pharmaceutical testing and biotechnological applications. Via blastocyst complementation, pluripotent cells are a favoured tool for the generation of genetically modified mice. The recently established technology to generate an induced pluripotency status by ectopic co-expression of the transcription factors Oct4, Sox2, Klf4 and c-Myc allows to extending these applications to farm animal species, for which the derivation of genuine embryonic stem cells was not successful so far. Most induced pluripotent stem (iPS) cells are generated by retroviral or lentiviral transduction of reprogramming factors. Multiple viral integrations into the genome may cause insertional mutagenesis and may increase the risk of tumour formation. Non-integration methods have been reported to overcome the safety concerns associated with retro and lentiviral-derived iPS cells, such as transient expression of the reprogramming factors using episomal plasmids, and direct delivery of reprogramming mRNAs or proteins. In this review, we focus on the mechanisms of cellular reprogramming and current methods used to induce pluripotency. We also highlight problems associated with the generation of iPS cells. An increased understanding of the fundamental mechanisms underlying pluripotency and refining the methodology of iPS cell generation will have a profound impact on future development and application in regenerative medicine and reproductive biotechnology of farm animals.
Collapse
|
44
|
Zeng F, Huang F, Guo J, Hu X, Liu C, Wang H. Emerging methods to generate artificial germ cells from stem cells. Biol Reprod 2015; 92:89. [PMID: 25715792 DOI: 10.1095/biolreprod.114.124800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/16/2015] [Indexed: 12/29/2022] Open
Abstract
Germ cells are responsible for the transmission of genetic and epigenetic information across generations. At present, the number of infertile couples is increasing worldwide; these infertility problems can be traced to environmental pollutions, infectious diseases, cancer, psychological or work-related stress, and other factors, such as lifestyle and genetics. Notably, lack of germ cells and germ cell loss present real obstacles in infertility treatment. Recent research aimed at producing gametes through artificial germ cell generation from stem cells may offer great hope for affected couples to treat infertility in the future. Therefore, this rapidly emerging area of artificial germ cell generation from nongermline cells has gained considerable attention from basic and clinical research in the fields of stem cell biology, developmental biology, and reproductive biology. Here, we review the state of the art in artificial germ cell generation.
Collapse
Affiliation(s)
- Fanhui Zeng
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Fajun Huang
- School of Medical Science, Hubei University for Nationalities, Enshi, China
| | - Jingjing Guo
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, China
| | - Xingchang Hu
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, China
| | - Changbai Liu
- The Institute of Molecular Biology, China Three Gorges University, Yichang, China
| | - Hu Wang
- Medical School, China Three Gorges University, Yichang, China
| |
Collapse
|
45
|
Hendriks S, Dancet EA, van Pelt AM, Hamer G, Repping S. Artificial gametes: a systematic review of biological progress towards clinical application. Hum Reprod Update 2015; 21:285-96. [DOI: 10.1093/humupd/dmv001] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/29/2014] [Indexed: 01/15/2023] Open
|
46
|
Zhao J, Jiang WJ, Sun C, Hou CZ, Yang XM, Gao JG. Induced pluripotent stem cells: origins, applications, and future perspectives. J Zhejiang Univ Sci B 2014; 14:1059-69. [PMID: 24302707 DOI: 10.1631/jzus.b1300215] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Embryonic stem (ES) cells are widely used for different purposes, including gene targeting, cell therapy, tissue repair, organ regeneration, and so on. However, studies and applications of ES cells are hindered by ethical issues regarding cell sources. To circumvent ethical disputes, great efforts have been taken to generate ES cell-like cells, which are not derived from the inner cell mass of blastocyst-stage embryos. In 2006, Yamanaka et al. first reprogrammed mouse embryonic fibroblasts into ES cell-like cells called induced pluripotent stem (iPS) cells. About one year later, Yamanaka et al. and Thomson et al. independently reprogrammed human somatic cells into iPS cells. Since the first generation of iPS cells, they have now been derived from quite a few different kinds of cell types. In particular, the use of peripheral blood facilitates research on iPS cells because of safety, easy availability, and plenty of cell sources. Now iPS cells have been used for cell therapy, disease modeling, and drug discovery. In this review, we describe the generations, applications, potential issues, and future perspectives of iPS cells.
Collapse
Affiliation(s)
- Jing Zhao
- School of Life Sciences, Shandong University, Jinan 250100, China
| | | | | | | | | | | |
Collapse
|
47
|
Stem cells as new agents for the treatment of infertility: current and future perspectives and challenges. BIOMED RESEARCH INTERNATIONAL 2014; 2014:507234. [PMID: 24826378 PMCID: PMC4009115 DOI: 10.1155/2014/507234] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/12/2014] [Accepted: 03/17/2014] [Indexed: 12/21/2022]
Abstract
Stem cells are undifferentiated cells that are present in the embryonic, fetal, and adult stages of life and give rise to differentiated cells that make up the building blocks of tissue and organs. Due to their unlimited source and high differentiation potential, stem cells are considered as potentially new therapeutic agents for the treatment of infertility. Stem cells could be stimulated in vitro to develop various numbers of specialized cells including male and female gametes suggesting their potential use in reproductive medicine. During past few years a considerable progress in the derivation of male germ cells from pluripotent stem cells has been made. In addition, stem cell-based strategies for ovarian regeneration and oocyte production have been proposed as future clinical therapies for treating infertility in women. In this review, we summarized current knowledge and present future perspectives and challenges regarding the use of stem cells in reproductive medicine.
Collapse
|
48
|
Botman O, Wyns C. Induced pluripotent stem cell potential in medicine, specifically focused on reproductive medicine. Front Surg 2014; 1:5. [PMID: 25671222 PMCID: PMC4313692 DOI: 10.3389/fsurg.2014.00005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/05/2014] [Indexed: 01/15/2023] Open
Abstract
Since 2006, several laboratories have proved that somatic cells can be reprogramed into induced pluripotent stem cells (iPSCs). iPSCs have enormous potential in stem cell biology as they can give rise to numerous cell lineages, including the three germ layers. In this review, we discuss past and recent advances in human iPSCs used for modeling diseases in vitro, screening drugs to test new treatments, and autologous cell and tissue regenerative therapies, with a special focus on reproductive medicine applications. While this latter field of research is still in its infancy, it holds great promise for investigating germ cell development and studying the genetic and physiopathological mechanisms of infertility. A major cause of infertility is the absence of germ cells in the testes, mainly due to genetic background or as a consequence of gonadotoxic treatments. For these patients, no effective fertility restoration strategy has so far been identified. The derivation of germ cells from iPSCs represents an alternative source of stem cells able to differentiate into spermatozoa. Lessons learned from animal models as well as studies on human iPSCs for reproductive purposes are reviewed.
Collapse
Affiliation(s)
- Olivier Botman
- Gynecology Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain , Brussels , Belgium
| | - Christine Wyns
- Gynecology Unit, Medical School, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain , Brussels , Belgium ; Cliniques Universitaires Saint-Luc, Université Catholique de Louvain , Brussels , Belgium
| |
Collapse
|
49
|
Kanto S, Yamasaki K, Iwamoto T. Progressing management of non-obstructive azoospermia in the era of microdissection testicular sperm extraction. Reprod Med Biol 2014; 13:119-125. [PMID: 29699155 DOI: 10.1007/s12522-014-0178-0] [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: 01/15/2014] [Accepted: 02/13/2014] [Indexed: 10/25/2022] Open
Abstract
Previously, it was absolutely impossible for azoospermic men to reproduce except in some obstructive azoospermic cases for whom reconstruction of the seminal pathway was successful. However, nowadays, intracytoplasmic sperm injection and microdissection testicular sperm extraction have brought about chances of biological paternity in some non-obstructive azoospermic men. It is almost 15 years since the first trials of testicular sperm retrieval using surgical microscopy for non-obstructive azoospermia were reported. In this manuscript, the progress and outcomes of these novel techniques since then are reviewed, the controversial points are discussed and the latest research to achieve pregnancies in tough non-obstructive azoospermic couples are introduced. Not only the bright side of the renovations, but the underlying concerns are also discussed.
Collapse
Affiliation(s)
- Satoru Kanto
- Department of Urology International University of Health and Welfare Shioya Hospital 77 Tomita 329-2145 Yaita Tochigi Japan
| | - Kazumitsu Yamasaki
- Department of Urology International University of Health and Welfare Hospital 537-3 Iguchi Nasushiobara Japan
| | - Teruaki Iwamoto
- International University of Health and Welfare Hospital, Reproduction Center 537-3 Iguchi Nasushiobara Japan
| |
Collapse
|
50
|
Sun YC, Cheng SF, Sun R, Zhao Y, Shen W. Reconstitution of Gametogenesis In Vitro: Meiosis Is the Biggest Obstacle. J Genet Genomics 2014; 41:87-95. [DOI: 10.1016/j.jgg.2013.12.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 12/08/2013] [Accepted: 12/20/2013] [Indexed: 01/08/2023]
|