1
|
Liao C, Walters BW, DiStasio M, Lesch BJ. Human-specific epigenomic states in spermatogenesis. Comput Struct Biotechnol J 2024; 23:577-588. [PMID: 38274996 PMCID: PMC10809009 DOI: 10.1016/j.csbj.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/23/2023] [Accepted: 12/23/2023] [Indexed: 01/27/2024] Open
Abstract
Infertility is becoming increasingly common, affecting one in six people globally. Half of these cases can be attributed to male factors, many driven by abnormalities in the process of sperm development. Emerging evidence from genome-wide association studies, genetic screening of patient cohorts, and animal models highlights an important genetic contribution to spermatogenic defects, but comprehensive identification and characterization of the genes critical for male fertility remain lacking. High divergence of gene regulation in spermatogenic cells across species poses challenges for delineating the genetic pathways required for human spermatogenesis using common model organisms. In this study, we leveraged post-translational histone modification and gene transcription data for 15,491 genes in four mammalian species (human, rhesus macaque, mouse, and opossum), to identify human-specific patterns of gene regulation during spermatogenesis. We combined H3K27me3 ChIP-seq, H3K4me3 ChIP-seq, and RNA-seq data to define epigenetic states for each gene at two stages of spermatogenesis, pachytene spermatocytes and round spermatids, in each species. We identified 239 genes that are uniquely active, poised, or dynamically regulated in human spermatogenic cells distinct from the other three species. While some of these genes have been implicated in reproductive functions, many more have not yet been associated with human infertility and may be candidates for further molecular and epidemiologic studies. Our analysis offers an example of the opportunities provided by evolutionary and epigenomic data for broadly screening candidate genes implicated in reproduction, which might lead to discoveries of novel genetic targets for diagnosis and management of male infertility and male contraception.
Collapse
Affiliation(s)
- Caiyun Liao
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | | | - Marcello DiStasio
- Department of Pathology, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Department of Opthamology & Visual Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Bluma J. Lesch
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Department of Genetics, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Yale Cancer Center, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
2
|
Zhang D, Jin W, Cui Y, He Z. Establishment and Characterization of Testis Organoids with Proliferation and Differentiation of Spermatogonial Stem Cells into Spermatocytes and Spermatids. Cells 2024; 13:1642. [PMID: 39404405 PMCID: PMC11476282 DOI: 10.3390/cells13191642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Organoids play pivotal roles in uncovering the molecular mechanisms underlying organogenesis, intercellular communication, and high-throughput drug screening. Testicular organoids are essential for exploring the genetic and epigenetic regulation of spermatogenesis in vivo and the treatment of male infertility. However, the formation of testicular organoids with full spermatogenesis has not yet been achieved. In this study, neonatal mouse testicular cells were isolated by two-step enzymatic digestion, and they were combined with Matrigel and transplanted subcutaneously into nude mice. Histological examination (H&E) staining and immunohistochemistry revealed that cell grafts assembled to form seminiferous tubules that contained spermatogonial stem cells (SSCs) and Sertoli cells, as illustrated by the co-expression of PLZF (a hallmark for SSCs) and SOX9 (a marker for Sertoli cells) as well as the co-expression of UCHL1 (a hallmark for SSCs) and SOX9, after 8 weeks of transplantation. At 10 weeks of transplantation, SSCs could proliferate and differentiate into spermatocytes as evidenced by the expression of PCNA, Ki67, c-Kit, SYCP3, γ-HA2X, and MLH1. Notably, testicular organoids were seen, and spermatids were observed within the lumen of testicular organoids after 16 weeks of transplantation, as shown by the presence of TNP1 and ACROSIN (hallmarks for spermatids). Collectively, these results implicate that we successfully established testicular organoids with spermatogenesis in vivo. This study thus provides an excellent platform for unveiling the mechanisms underlying mammalian spermatogenesis, and it might offer valuable male gametes for treating male infertility.
Collapse
Affiliation(s)
- Dong Zhang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China; (D.Z.); (W.J.); (Y.C.)
- Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410008, China
- Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Wencong Jin
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China; (D.Z.); (W.J.); (Y.C.)
- Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410008, China
- Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Yinghong Cui
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China; (D.Z.); (W.J.); (Y.C.)
- Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410008, China
- Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China; (D.Z.); (W.J.); (Y.C.)
- Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410008, China
- Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| |
Collapse
|
3
|
Nguyen ALV, Julian S, Weng N, Flannigan R. Advances in human In vitro spermatogenesis: A review. Mol Aspects Med 2024; 100:101320. [PMID: 39317014 DOI: 10.1016/j.mam.2024.101320] [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: 03/27/2024] [Revised: 07/26/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Recent advances surrounding in vitro spermatogenesis (IVS) have shown potential in creating a new paradigm of regenerative medicine in the future of fertility treatments for males experiencing non-obstructive azoospermia (NOA). Male infertility is a common condition affecting approximately 15% of couples, with azoospermia being present in 15% of infertile males (Cocuzza et al., 2013; Esteves et al., 2011a). Treatment for patients with NOA has primarily been limited to surgical sperm retrieval combined with in vitro fertilization intracytoplasmic sperm injection (IVF-ICSI); however, sperm retrieval is successful in only half of these patients, and live birth rates typically range between 10 and 25% (Aljubran et al., 2022). Therefore, a significant need exists for regenerative therapies in this patient population. IVS has been considered as a model for further understanding the molecular and cellular processes of spermatogenesis and as a potential regenerative therapeutic approach. While 2D cell cultures using human testicular cells have been attempted in previous research, lack of proper spatial arrangement limits germ cell differentiation and maturation, posing challenges for clinical application. Recent research suggests that 3D technology may have advantages for IVS due to mimicry of the native cytoarchitecture of human testicular tissue along with cell-cell communication directly or indirectly. 3D organotypic cultures, scaffolds, organoids, microfluidics, testis-on-a-chip, and bioprinting techniques have all shown potential to contribute to the technology of regenerative treatment strategies, including in vitro fertilization (IVF). Although promising, further work is needed to develop technology for successful, replicable, and safe IVS for humans. The intersection between tissue engineering, molecular biology, and reproductive medicine in IVS development allows for multidisciplinary involvement, where challenges can be overcome to realize regenerative therapies as a viable option.
Collapse
Affiliation(s)
- Anna-Lisa V Nguyen
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, UK.
| | - Sania Julian
- Faculty of Integrated Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Ninglu Weng
- Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Ryan Flannigan
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Department of Urology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
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
|
5
|
von Rohden E, Jensen CFS, Andersen CY, Sønksen J, Fedder J, Thorup J, Ohl DA, Fode M, Hoffmann ER, Mamsen LS. Male fertility restoration: in vivo and in vitro stem cell-based strategies using cryopreserved testis tissue: a scoping review. Fertil Steril 2024:S0015-0282(24)00603-4. [PMID: 38992744 DOI: 10.1016/j.fertnstert.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
IMPORTANCE Advances in the treatment of childhood cancer have significantly improved survival rates, with more than 80% of survivors reaching adulthood. However, gonadotoxic cancer treatments endanger future fertility, and prepubertal males have no option to preserve fertility by sperm cryopreservation. In addition, boys with cryptorchidism are at risk of compromised fertility in adulthood. OBJECTIVE To investigate current evidence for male fertility restoration strategies, explore barriers to clinical implementation, and outline potential steps to overcome these barriers, a scoping review was conducted. This knowledge synthesis is particularly relevant for prepubertal male cancer survivors and boys with cryptorchidism. EVIDENCE REVIEW The review was conducted after the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews criteria and previously published guidelines and examined studies using human testis tissue of prepubertal boys or healthy male adults. A literature search in PubMed was conducted, and 72 relevant studies were identified, including in vivo and in vitro approaches. FINDINGS In vivo strategies, such as testis tissue engraftment and spermatogonial stem cell transplantation, hold promise for promoting cell survival and differentiation. Yet, complete spermatogenesis has not been achieved. In vitro approaches focus on the generation of male germ cells from direct germ cell maturation in various culture systems, alongside human induced pluripotent stem cells and embryonic stem cells. These approaches mark significant advancements in understanding and promoting spermatogenesis, but achieving fully functional spermatozoa in vitro remains a challenge. Barriers to clinical implementation include the risk of reintroducing malignant cells and introduction of epigenetic changes. CONCLUSION Male fertility restoration is an area in rapid development. On the basis of the reviewed studies, the most promising and advanced strategy for restoring male fertility using cryopreserved testis tissue is direct testis tissue transplantation. RELEVANCE This review identifies persistent barriers to the clinical implementation of male fertility restoration. However, direct transplantation of frozen-thawed testis tissue remains a promising strategy that is on the verge of clinical application.
Collapse
Affiliation(s)
- Elena von Rohden
- Department of Urology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Laboratory of Reproductive Biology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | | | - Claus Yding Andersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Sønksen
- Department of Urology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Fedder
- Department of Gynecology and Obstetrics, Centre of Andrology & Fertility Clinic, Odense University Hospital, Odense, Denmark; Research Unit of Human Reproduction, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jørgen Thorup
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Pediatric Surgery, Surgical Clinic, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Dana A Ohl
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Mikkel Fode
- Department of Urology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Eva R Hoffmann
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Molecular and Cellular Medicine, DNRF Center for Chromosome Stability, University of Copenhagen, Copenhagen, Denmark
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
6
|
Galdon G, Zarandi NP, Deebel NA, Zhang S, Cornett O, Lyalin D, Pettenati MJ, Lue Y, Wang C, Swerdloff R, Shupe TD, Bishop C, Stogner K, Kogan SJ, Howards S, Atala A, Sadri-Ardekani H. In Vitro Generation of Haploid Germ Cells from Human XY and XXY Immature Testes in a 3D Organoid System. Bioengineering (Basel) 2024; 11:677. [PMID: 39061759 PMCID: PMC11274239 DOI: 10.3390/bioengineering11070677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Increasing survival rates of children following cancer treatment have resulted in a significant population of adult survivors with the common side effect of infertility. Additionally, the availability of genetic testing has identified Klinefelter syndrome (classic 47,XXY) as the cause of future male infertility for a significant number of prepubertal patients. This study explores new spermatogonia stem cell (SSC)-based fertility therapies to meet the needs of these patients. Testicular cells were isolated from cryopreserved human testes tissue stored from XY and XXY prepubertal patients and propagated in a two-dimensional culture. Cells were then incorporated into a 3D human testicular organoid (HTO) system. During a 3-week culture period, HTOs maintained their structure, viability, and metabolic activity. Cell-specific PCR and flow cytometry markers identified undifferentiated spermatogonia, Sertoli, Leydig, and peritubular cells within the HTOs. Testosterone was produced by the HTOs both with and without hCG stimulation. Upregulation of postmeiotic germ cell markers was detected after 23 days in culture. Fluorescence in situ hybridization (FISH) of chromosomes X, Y, and 18 identified haploid cells in the in vitro differentiated HTOs. Thus, 3D HTOs were successfully generated from isolated immature human testicular cells from both euploid (XY) and Klinefelter (XXY) patients, supporting androgen production and germ cell differentiation in vitro.
Collapse
Affiliation(s)
- Guillermo Galdon
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Facultad de Medicina, Universidad de Barcelona, 08036 Barcelona, Spain
| | - Nima Pourhabibi Zarandi
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Internal Medicine, University of Pittsburgh Medical Center, Harrisburg, PA 17101, USA
| | - Nicholas A. Deebel
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Sue Zhang
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Olivia Cornett
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Dmitry Lyalin
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Pathology, Molecular Diagnostics Division, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Mark J. Pettenati
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - YanHe Lue
- Division of Endocrinology, Department of Medicine, The Lundquist Institute, Harbor-University of California Los Angeles (UCLA) Medical Center, Los Angeles, CA 90502, USA
| | - Christina Wang
- Division of Endocrinology, Department of Medicine, The Lundquist Institute, Harbor-University of California Los Angeles (UCLA) Medical Center, Los Angeles, CA 90502, USA
| | - Ronald Swerdloff
- Division of Endocrinology, Department of Medicine, The Lundquist Institute, Harbor-University of California Los Angeles (UCLA) Medical Center, Los Angeles, CA 90502, USA
| | - Thomas D. Shupe
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Colin Bishop
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Kimberly Stogner
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Stanley J. Kogan
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Stuart Howards
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Hooman Sadri-Ardekani
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
7
|
Kwaspen L, Kanbar M, Wyns C. Mapping the Development of Human Spermatogenesis Using Transcriptomics-Based Data: A Scoping Review. Int J Mol Sci 2024; 25:6925. [PMID: 39000031 PMCID: PMC11241379 DOI: 10.3390/ijms25136925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
In vitro maturation (IVM) is a promising fertility restoration strategy for patients with nonobstructive azoospermia or for prepubertal boys to obtain fertilizing-competent spermatozoa. However, in vitro spermatogenesis is still not achieved with human immature testicular tissue. Knowledge of various human testicular transcriptional profiles from different developmental periods helps us to better understand the testis development. This scoping review aims to describe the testis development and maturation from the fetal period towards adulthood and to find information to optimize IVM. Research papers related to native and in vitro cultured human testicular cells and single-cell RNA-sequencing (scRNA-seq) were identified and critically reviewed. Special focus was given to gene ontology terms to facilitate the interpretation of the biological function of related genes. The different consecutive maturation states of both the germ and somatic cell lineages were described. ScRNA-seq regularly showed major modifications around 11 years of age to eventually reach the adult state. Different spermatogonial stem cell (SSC) substates were described and scRNA-seq analyses are in favor of a paradigm shift, as the Adark and Apale spermatogonia populations could not distinctly be identified among the different SSC states. Data on the somatic cell lineage are limited, especially for Sertoli cells due technical issues related to cell size. During cell culture, scRNA-seq data showed that undifferentiated SSCs were favored in the presence of an AKT-signaling pathway inhibitor. The involvement of the oxidative phosphorylation pathway depended on the maturational state of the cells. Commonly identified cell signaling pathways during the testis development and maturation highlight factors that can be essential during specific maturation stages in IVM.
Collapse
Affiliation(s)
- Lena Kwaspen
- Laboratoire d’Andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (L.K.); (M.K.)
| | - Marc Kanbar
- Laboratoire d’Andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (L.K.); (M.K.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Christine Wyns
- Laboratoire d’Andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (L.K.); (M.K.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
8
|
Zhang Y, Wang M, Zhang T, Wang H, Chen Y, Zhou T, Yang R. Spermbots and Their Applications in Assisted Reproduction: Current Progress and Future Perspectives. Int J Nanomedicine 2024; 19:5095-5108. [PMID: 38836008 PMCID: PMC11149708 DOI: 10.2147/ijn.s465548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024] Open
Abstract
Sperm quality is declining dramatically during the past decades. Male infertility has been a serious health and social problem. The sperm cell driven biohybrid nanorobot opens a new era for automated and precise assisted reproduction. Therefore, it is urgent and necessary to conduct an updated review and perspective from the viewpoints of the researchers and clinicians in the field of reproductive medicine. In the present review, we first update the current classification, design, control and applications of various spermbots. Then, by a comprehensive summary of the functional features of sperm cells, the journey of sperms to the oocyte, and sperm-related dysfunctions, we provide a systematic guidance to further improve the design of spermbots. Focusing on the translation of spermbots into clinical practice, we point out that the main challenges are biocompatibility, effectiveness, and ethical issues. Considering the special requirements of assisted reproduction, we also propose the three laws for the clinical usage of spermbots: good genetics, gentle operation and no contamination. Finally, a three-step roadmap is proposed to achieve the goal of clinical translation. We believe that spermbot-based treatments can be validated and approved for in vitro clinical usage in the near future. However, multi-center and multi-disciplinary collaborations are needed to further promote the translation of spermbots into in vivo clinical applications.
Collapse
Affiliation(s)
- Yixuan Zhang
- Research Institute for Reproductive Medicine and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, People’s Republic of China
| | - Min Wang
- Center for Reproductive Medicine, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, People’s Republic of China
| | - Ting Zhang
- Department of Laboratory Medicine, Wuxi Maternity and Child Health Care Hospital, Jiangnan University, Wuxi, 214002, People’s Republic of China
| | - Honghua Wang
- Center for Reproductive Medicine, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, People’s Republic of China
| | - Ying Chen
- Research Institute for Reproductive Medicine and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, People’s Republic of China
| | - Tao Zhou
- Research Institute for Reproductive Medicine and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, People’s Republic of China
| | - Rui Yang
- Research Institute for Reproductive Medicine and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, People’s Republic of China
| |
Collapse
|
9
|
Piechka A, Sparanese S, Witherspoon L, Hach F, Flannigan R. Molecular mechanisms of cellular dysfunction in testes from men with non-obstructive azoospermia. Nat Rev Urol 2024; 21:67-90. [PMID: 38110528 DOI: 10.1038/s41585-023-00837-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 12/20/2023]
Abstract
Male factor infertility affects 50% of infertile couples worldwide; the most severe form, non-obstructive azoospermia (NOA), affects 10-15% of infertile males. Treatment for individuals with NOA is limited to microsurgical sperm extraction paired with in vitro fertilization intracytoplasmic sperm injection. Unfortunately, spermatozoa are only retrieved in ~50% of patients, resulting in live birth rates of 21-46%. Regenerative therapies could provide a solution; however, understanding the cell-type-specific mechanisms of cellular dysfunction is a fundamental necessity to develop precision medicine strategies that could overcome these abnormalities and promote regeneration of spermatogenesis. A number of mechanisms of cellular dysfunction have been elucidated in NOA testicular cells. These mechanisms include abnormalities in both somatic cells and germ cells in NOA testes, such as somatic cell immaturity, aberrant growth factor signalling, increased inflammation, increased apoptosis and abnormal extracellular matrix regulation. Future cell-type-specific investigations in identifying modulators of cellular transcription and translation will be key to understanding upstream dysregulation, and these studies will require development of in vitro models to functionally interrogate spermatogenic niche dysfunction in both somatic and germ cells.
Collapse
Affiliation(s)
- Arina Piechka
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Sydney Sparanese
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Luke Witherspoon
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Urology, Department of Surgery, University of Ottawa, Ontario, Canada
| | - Faraz Hach
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Ryan Flannigan
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.
- Department of Urology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Önen S, Gizer M, Korkusuz P. Flow Cytometric and Immunohistochemical Follow-Up of Spermatogonial Lineage Commitment. Methods Mol Biol 2024; 2849:239-251. [PMID: 37801256 DOI: 10.1007/7651_2023_506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Flow cytometry and immunohistochemistry techniques both determine the target protein by immunolabeling. Flow cytometric analysis quantifies total number of fluorescent labeled cells and qualify sup-populations according to size and granularity. Immunohistochemistry is able to map immune-labeled cells and extracellular matrix components under light and electron microscope by enzyme or fluorescent molecules. Real-time identification, in-time classification, and final plotting of spermatogonial lineage are of crucial importance for monitoring the fertility potential of spermatogonial stem cell microenvironment and predicting progress of spermatogenesis. Here we define the evaluation of mouse male germ cell microenvironment at single cell and whole tissue section levels by using flow cytometric and immunohistochemical approaches.
Collapse
Affiliation(s)
| | - Merve Gizer
- METU MEMS Center, Ankara, Turkey
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Petek Korkusuz
- METU MEMS Center, Ankara, Turkey.
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|