1
|
Sun Y, Zhong B, Meng Z, Zhang Y, Li Z, Yao C. Intact spermatogenesis in an azoospermic patient with AZFa (sY84 and sY86) microdeletion and a homozygous TG12-5T variant in CFTR. Basic Clin Androl 2025; 35:13. [PMID: 40169970 DOI: 10.1186/s12610-025-00260-7] [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: 01/15/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Azoospermia, the most severe form of male infertility, is categorized into two types: non-obstructive azoospermia (NOA) and obstructive azoospermia (OA), which exhibit significant genetic heterogeneity. Azoospermia factor (AZF) deletion is a common cause of NOA, whereas congenital bilateral absence of the vas deferens (CBAVD), a severe subtype of OA, is frequently linked to cystic fibrosis transmembrane conductance regulator (CFTR) gene variants. This case report is the first to document the coexistence of a partial AZFa microdeletion and a homozygous CFTR variant in a CBAVD-affected azoospermic patient with intact spermatogenesis. CASE PRESENTATION A 32-year-old man presented with primary infertility and azoospermia. Clinical evaluation revealed CBAVD (normal hormone levels, low semen volume, pH 6.0, and absence of the vas deferens). Genetic analysis accidentally revealed a 384.9 kb AZFa deletion (sY84 and sY86, but not sY1064, 1182) that removed USP9Y but retained DDX3Y in the proband, his fertile brother, and his father. A homozygous CFTR variant (TG12-5T) was also detected in the proband and his brother and was inherited from heterozygous parental carriers. Microdissection testicular sperm extraction (micro-TESE) revealed intact spermatogenesis, confirmed by histology and immunofluorescence, indicating normal germ cell development. CONCLUSION This case expands the intricate genetic spectrum of azoospermia by illustrating the critical role of DDX3Y in the AZFa region in spermatogenesis and the variable penetrance of CFTR variant (TG12-5T) in CBAVD. These insights may refine diagnostic strategies and underscore the necessity for tailored fertility management in individuals with multifactorial genetic anomalies.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Andrology, Shanghai Key Laboratory of Reproductive Medicine, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Beifen Zhong
- Department of Andrology, Shanghai Key Laboratory of Reproductive Medicine, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zizhou Meng
- Department of Andrology, Shanghai Key Laboratory of Reproductive Medicine, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yuxiang Zhang
- Department of Andrology, Shanghai Key Laboratory of Reproductive Medicine, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zheng Li
- Department of Andrology, Shanghai Key Laboratory of Reproductive Medicine, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, Taizhou, 225300, China.
| | - Chencheng Yao
- Department of Andrology, Shanghai Key Laboratory of Reproductive Medicine, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
2
|
Wanjari UR, Gopalakrishnan AV. Exploring the therapeutic effect of melatonin targeting common biomarkers in testicular germ cell tumor, prostate adenocarcinoma, and male infertility: an integrated biology approach. Mamm Genome 2025:10.1007/s00335-025-10119-x. [PMID: 40056207 DOI: 10.1007/s00335-025-10119-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025]
Abstract
Globally, male infertility (MI) is a major concern. Several other comorbidities related to MI are testicular germ cell tumor (TGCT) and prostate adenocarcinoma (PRAD). This study focuses on finding the common biomarkers among these diseases and their interaction with Melatonin (MLT). The differential expressed genes were retrieved using the GEPIA2 database for TGCT and PRAD, whereas the DISGENET database for MI-related genes. InteractiVenn was performed in response to identify the common genes. The STAG3, RNF212, DDX3Y, DPY19L2, TPCN1, KLK3, GNRH1, DMD, CCDC146, and DNAH1 are found to be involved in all these diseases. The gene ontologies and pathway enrichment analysis were done for these significant genes in response to identifying and accessing the involvement of these genes in other processes. MLT is a neuroendocrine hormone with high therapeutic properties. MLT showed the best binding energy with DDX3Y among all the proteins. Molecular dynamic simulation (MDS) of MLT with DDX3Y was performed and found to be -52.382 ± 13.110 kJ/mol binding energy. The RMSD, RMSF, SASA, RG, H-bond, FEL, PCA, and MM-PBSA analysis confirm the stability and compactness of the DDX3Y-MLT complex. The MDS results indicate that MLT is a promising therapeutic option for enhancing DDX3Y expression, which will support spermatogenesis. Additionally, the hub genes were identified based on MCC parameters from the merged interactive network of common genes in response to finding significant genes that can be a potential biomarker for the diagnosis of diseases.
Collapse
Affiliation(s)
- Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
3
|
Rengarajan S, Derks J, Bellott DW, Slavov N, Page DC. Post-transcriptional cross- and auto-regulation buffer expression of the human RNA helicases DDX3X and DDX3Y. Genome Res 2025; 35:20-30. [PMID: 39794123 PMCID: PMC11789639 DOI: 10.1101/gr.279707.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025]
Abstract
The Y-linked gene DDX3Y and its X-linked homolog DDX3X survived the evolution of the human sex chromosomes from ordinary autosomes. DDX3X encodes a multifunctional RNA helicase, with mutations causing developmental disorders and cancers. We find that, among X-linked genes with surviving Y homologs, DDX3X is extraordinarily dosage sensitive. Studying cells of individuals with sex chromosome aneuploidy, we observe that when the number of Y Chromosomes increases, DDX3X transcript levels fall; conversely, when the number of X Chromosomes increases, DDX3Y transcript levels fall. In 46,XY cells, CRISPRi knockdown of either DDX3X or DDX3Y causes transcript levels of the homologous gene to rise. In 46,XX cells, chemical inhibition of DDX3X protein activity elicits an increase in DDX3X transcript levels. Thus, perturbation of either DDX3X or DDX3Y expression is buffered: by negative cross-regulation of DDX3X and DDX3Y in 46,XY cells and by negative auto-regulation of DDX3X in 46,XX cells. DDX3X-DDX3Y cross-regulation is mediated through mRNA destabilization-as shown by metabolic labeling of newly transcribed RNA-and buffers total levels of DDX3X and DDX3Y protein in human cells. We infer that post-transcriptional auto-regulation of the ancestral (autosomal) DDX3X gene transmuted into auto- and cross-regulation of DDX3X and DDX3Y as these sex-linked genes evolved from ordinary alleles of their autosomal precursor.
Collapse
Affiliation(s)
- Shruthi Rengarajan
- Whitehead Institute, Cambridge, Massachusetts 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Jason Derks
- Departments of Bioengineering, Biology, Chemistry, and Chemical Biology, Single Cell Proteomics Center, and Barnett Institute, Northeastern University, Boston, Massachusetts 02115, USA
| | | | - Nikolai Slavov
- Departments of Bioengineering, Biology, Chemistry, and Chemical Biology, Single Cell Proteomics Center, and Barnett Institute, Northeastern University, Boston, Massachusetts 02115, USA
| | - David C Page
- Whitehead Institute, Cambridge, Massachusetts 02142, USA;
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
4
|
Mossa A, Dierdorff L, Lukin J, Park Y, Fiorenzani C, Akpinar Z, Garcia-Forn M, De Rubeis S. Sex-specific perturbations of neuronal development caused by mutations in the autism risk gene DDX3X. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624865. [PMID: 39605424 PMCID: PMC11601590 DOI: 10.1101/2024.11.22.624865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
DDX3X is an X-linked RNA helicases that escapes X chromosome inactivation and is expressed at higher levels in female brains. Mutations in DDX3X are associated with intellectual disability (ID) and autism spectrum disorder (ASD) and are predominantly identified in females. Using cellular and mouse models, we show that Ddx3x mediates sexual dimorphisms in brain development at a molecular, cellular, and behavioral level. During cortical neuronal development, Ddx3x sustains a female-biased signature of enhanced ribosomal biogenesis and mRNA translation. Female neurons display higher levels of ribosomal proteins and larger nucleoli, and these sex dimorphisms are obliterated by Ddx3x loss. Ddx3x regulates dendritic outgrowth in a sex- and dose-dependent manner in both female and male neurons, and dendritic spine development only in female neurons. Further, ablating Ddx3x conditionally in forebrain neurons is sufficient to yield sex-specific changes in developmental outcomes and motor function. Together, these findings pose Ddx3x as a mediator of sexual differentiation during neurodevelopment and open new avenues to understand sex differences in health and disease.
Collapse
|
5
|
Tsai SY, Lin CH, Jiang YT, Huang GJ, Pi H, Hung HY, Tarn WY, Lai MC. DDX3 is critical for female fertility via translational control in oogenesis. Cell Death Discov 2024; 10:472. [PMID: 39551844 PMCID: PMC11570671 DOI: 10.1038/s41420-024-02242-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
DEAD-box RNA helicase 3 (DDX3) and its homologs play a vital role in translation initiation by unwinding secondary structures of selected mRNAs. The human DDX3 gene is located on the sex chromosomes, so there are DDX3X and DDX3Y. DDX3X is ubiquitously expressed in almost all tissues and critical for embryonic development, whereas DDX3Y is only expressed in the testis and essential for male fertility. Drosophila belle (bel) is the single ortholog of DDX3, and mutations in bel cause male and female infertility. Using Drosophila bel mutants and Ddx3x conditional knockout (cKO) mice, we confirmed the pivotal role of DDX3 in female fertility and ovarian development. Drosophila bel mutants exhibited female infertility and immature egg chambers. Consistently, oocyte-specific Ddx3x knockout in mice resulted in female infertility and impaired oogenesis. We further found that immature egg chambers in Drosophila bel mutants and impaired follicular development in oocyte-specific Ddx3x cKO mice were caused by excessive apoptosis. We also identified a set of DDX3 target genes involved in oocyte meiosis and maturation and demonstrated that DDX3 is involved in their translation in human cells. Our results suggest that DDX3 is critical for female fertility via translational control in oogenesis.
Collapse
Affiliation(s)
- Shang-Yu Tsai
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Hung Lin
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Master and PhD Program of Biotechnology Industry, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Ting Jiang
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Guo-Jen Huang
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Haiwei Pi
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Yuan Hung
- Department of Colorectal Surgery, New Taipei Municipal Tucheng Hospital, New Taipei, Taiwan
| | - Woan-Yuh Tarn
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ming-Chih Lai
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.
- Master and PhD Program of Biotechnology Industry, Chang Gung University, Taoyuan, Taiwan.
- Department of Colorectal Surgery, New Taipei Municipal Tucheng Hospital, New Taipei, Taiwan.
| |
Collapse
|
6
|
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; 122:828-843. [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] [MESH Headings] [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
|
7
|
Diaz Escarcega R, Marshall P, Tsvetkov AS. G-quadruplex DNA and RNA in cellular senescence. FRONTIERS IN AGING 2024; 5:1491389. [PMID: 39444378 PMCID: PMC11496277 DOI: 10.3389/fragi.2024.1491389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Normal cells divide, are damaged, and are repaired across their lifetime. As cells age, they enter cellular senescence, characterized by a permanent state of cell-cycle arrest triggered by various stressors. The molecular mechanisms that regulate senescent phenotypes have been actively investigated over the last several decades; however, one area that has been neglected is how G-quadruplex (G4) DNA and RNA (G4-DNA and G4-RNA) mediate senescence. These non-canonical four-stranded DNA and RNA structures regulate most normative DNA and RNA-dependent processes, such as transcription, replication, and translation, as well as pathogenic mechanisms, including genomic instability and abnormal stress granule function. This review also highlights the contribution of G4s to sex differences in age-associated diseases and emphasizes potential translational approaches to target senescence and anti-aging mechanisms through G4 manipulation.
Collapse
Affiliation(s)
- Rocio Diaz Escarcega
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX, United States
| | - Paul Marshall
- College of Health and Medicine, The Australian National University, Canberra, ACT, Australia
| | - Andrey S. Tsvetkov
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX, United States
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
- UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, TX, United States
| |
Collapse
|
8
|
Rengarajan S, Derks J, Bellott DW, Slavov N, Page DC. Post-transcriptional cross- and auto-regulation buffer expression of the human RNA helicases DDX3X and DDX3Y. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602613. [PMID: 39026797 PMCID: PMC11257633 DOI: 10.1101/2024.07.08.602613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The Y-linked gene DDX3Y and its X-linked homolog DDX3X survived the evolution of the human sex chromosomes from ordinary autosomes. DDX3X encodes a multi-functional RNA helicase, with mutations causing developmental disorders and cancers. We find that, among X-linked genes with surviving Y homologs, DDX3X is extraordinarily dosage-sensitive. Studying cells of individuals with sex chromosome aneuploidy, we observe that when the number of Y chromosomes increases, DDX3X transcript levels fall; conversely, when the number of X chromosomes increases, DDX3Y transcript levels fall. In 46,XY cells, CRISPRi knockdown of either DDX3X or DDX3Y causes transcript levels of the homologous gene to rise. In 46,XX cells, chemical inhibition of DDX3X protein activity elicits an increase in DDX3X transcript levels. Thus, perturbation of either DDX3X or DDX3Y expression is buffered - by negative cross-regulation of DDX3X and DDX3Y in 46,XY cells, and by negative auto-regulation of DDX3X in 46,XX cells. DDX3X-DDX3Y cross-regulation is mediated through mRNA destabilization - as shown by metabolic labeling of newly transcribed RNA - and buffers total levels of DDX3X and DDX3Y protein in human cells. We infer that post-transcriptional auto-regulation of the ancestral (autosomal) DDX3 gene transmuted into auto- and cross-regulation of DDX3X and DDX3Y as these sex-linked genes evolved from ordinary alleles of their autosomal precursor.
Collapse
Affiliation(s)
- Shruthi Rengarajan
- Whitehead Institute, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jason Derks
- Departments of Bioengineering, Biology, Chemistry and Chemical Biology, Single Cell Proteomics Center, and Barnett Institute, Northeastern University, Boston, MA, USA
| | | | - Nikolai Slavov
- Departments of Bioengineering, Biology, Chemistry and Chemical Biology, Single Cell Proteomics Center, and Barnett Institute, Northeastern University, Boston, MA, USA
| | - David C Page
- Whitehead Institute, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| |
Collapse
|
9
|
Paylar B, Pramanik S, Bezabhe YH, Olsson PE. Temporal sex specific brain gene expression pattern during early rat embryonic development. Front Cell Dev Biol 2024; 12:1343800. [PMID: 38961864 PMCID: PMC11219815 DOI: 10.3389/fcell.2024.1343800] [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: 11/24/2023] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
Background: The classical concept of brain sex differentiation suggests that steroid hormones released from the gonads program male and female brains differently. However, several studies indicate that steroid hormones are not the only determinant of brain sex differentiation and that genetic differences could also be involved. Methods: In this study, we have performed RNA sequencing of rat brains at embryonic days 12 (E12), E13, and E14. The aim was to identify differentially expressed genes between male and female rat brains during early development. Results: Analysis of genes expressed with the highest sex differences showed that Xist was highly expressed in females having XX genotype with an increasing expression over time. Analysis of genes expressed with the highest male expression identified three early genes, Sry2, Eif2s3y, and Ddx3y. Discussion: The observed sex-specific expression of genes at early development confirms that the rat brain is sexually dimorphic prior to gonadal action on the brain and identifies Sry2 and Eif2s3y as early genes contributing to male brain development.
Collapse
Affiliation(s)
| | | | | | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| |
Collapse
|
10
|
Jebur MII, Dastmalchi N, Banamolaei P, Safaralizadeh R. Polymorphisms and expression levels of TNP2, SYCP3, and AZFa genes in patients with azoospermia. Clin Exp Reprod Med 2023; 50:253-261. [PMID: 37995753 PMCID: PMC10711250 DOI: 10.5653/cerm.2023.06219] [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: 05/27/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 11/25/2023] Open
Abstract
OBJECTIVE Azoospermia (the total absence of sperm in the ejaculate) affects approximately 10% of infertile males. Despite diagnostic advances, azoospermia remains the most challenging issue associated with infertility treatment. Our study evaluated transition nuclear protein 2 (TNP2) and synaptonemal complex protein 3 (SYCP3) polymorphisms, azoospermia factor a (AZFa) microdeletion, and gene expression levels in 100 patients with azoospermia. METHODS We investigated a TNP2 single-nucleotide polymorphism through polymerase chain reaction (PCR) restriction fragment length polymorphism analysis using a particular endonuclease. An allele-specific PCR assay for SYCP3 was performed utilizing two forward primers and a common reverse primer in two PCR reactions. Based on the European Academy of Andrology guidelines, AZFa microdeletions were evaluated by multiplex PCR. TNP2, SYCP3, and the AZFa region main gene (DEAD-box helicase 3 and Y-linked [DDX3Y]) expression levels were assessed via quantitative PCR, and receiver operating characteristic curve analysis was used to determine the diagnostic capability of these genes. RESULTS The TNP2 genotyping and allelic frequency in infertile males did not differ significantly from fertile volunteers. In participants with azoospermia, the allelic frequency of the SYCP3 mutant allele (C allele) was significantly altered. Deletion of sY84 and sY86 was discovered in patients with azoospermia and oligozoospermia. Moreover, SYCP3 and DDX3Y showed decreased expression levels in the azoospermia group, and they exhibited potential as biomarkers for diagnosing azoospermia (area under the curve, 0.722 and 0.720, respectively). CONCLUSION These results suggest that reduced SYCP3 and DDX3Y mRNA expression profiles in testicular tissue are associated with a higher likelihood of retrieving spermatozoa in individuals with azoospermia. The homozygous genotype TT of the SYCP3 polymorphism was significantly associated with azoospermia.
Collapse
Affiliation(s)
| | - Narges Dastmalchi
- Department of Biology, University College of Nabi Akram, Tabriz, Iran
| | - Parisa Banamolaei
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
11
|
Anisimova AS, Kolyupanova NM, Makarova NE, Egorov AA, Kulakovskiy IV, Dmitriev SE. Human Tissues Exhibit Diverse Composition of Translation Machinery. Int J Mol Sci 2023; 24:8361. [PMID: 37176068 PMCID: PMC10179197 DOI: 10.3390/ijms24098361] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
While protein synthesis is vital for the majority of cell types of the human body, diversely differentiated cells require specific translation regulation. This suggests the specialization of translation machinery across tissues and organs. Using transcriptomic data from GTEx, FANTOM, and Gene Atlas, we systematically explored the abundance of transcripts encoding translation factors and aminoacyl-tRNA synthetases (ARSases) in human tissues. We revised a few known and identified several novel translation-related genes exhibiting strict tissue-specific expression. The proteins they encode include eEF1A1, eEF1A2, PABPC1L, PABPC3, eIF1B, eIF4E1B, eIF4ENIF1, and eIF5AL1. Furthermore, our analysis revealed a pervasive tissue-specific relative abundance of translation machinery components (e.g., PABP and eRF3 paralogs, eIF2B and eIF3 subunits, eIF5MPs, and some ARSases), suggesting presumptive variance in the composition of translation initiation, elongation, and termination complexes. These conclusions were largely confirmed by the analysis of proteomic data. Finally, we paid attention to sexual dimorphism in the repertoire of translation factors encoded in sex chromosomes (eIF1A, eIF2γ, and DDX3), and identified the testis and brain as organs with the most diverged expression of translation-associated genes.
Collapse
Affiliation(s)
- Aleksandra S. Anisimova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Natalia M. Kolyupanova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nadezhda E. Makarova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Artyom A. Egorov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ivan V. Kulakovskiy
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 117971 Moscow, Russia;
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
- Laboratory of Regulatory Genomics, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Sergey E. Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
12
|
Dicke AK, Pilatz A, Wyrwoll MJ, Punab M, Ruckert C, Nagirnaja L, Aston KI, Conrad DF, Di Persio S, Neuhaus N, Fietz D, Laan M, Stallmeyer B, Tüttelmann F. DDX3Y is likely the key spermatogenic factor in the AZFa region that contributes to human non-obstructive azoospermia. Commun Biol 2023; 6:350. [PMID: 36997603 PMCID: PMC10063662 DOI: 10.1038/s42003-023-04714-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/15/2023] [Indexed: 04/01/2023] Open
Abstract
Non-obstructive azoospermia, the absence of sperm in the ejaculate due to disturbed spermatogenesis, represents the most severe form of male infertility. De novo microdeletions of the Y-chromosomal AZFa region are one of few well-established genetic causes for NOA and are routinely analysed in the diagnostic workup of affected men. So far, it is unclear which of the three genes located in the AZFa chromosomal region is indispensible for germ cell maturation. Here we present four different likely pathogenic loss-of-function variants in the AZFa gene DDX3Y identified by analysing exome sequencing data of more than 1,600 infertile men. Three of the patients underwent testicular sperm extraction and revealed the typical AZFa testicular Sertoli cell-only phenotype. One of the variants was proven to be de novo. Consequently, DDX3Y represents the AZFa key spermatogenic factor and screening for variants in DDX3Y should be included in the diagnostic workflow.
Collapse
Affiliation(s)
- Ann-Kristin Dicke
- Institute of Reproductive Genetics, University of Münster, 48149, Münster, Germany
| | - Adrian Pilatz
- Clinic for Urology, Paediatric Urology and Andrology, Justus Liebig University Gießen, 35390, Gießen, Germany
| | - Margot J Wyrwoll
- Institute of Reproductive Genetics, University of Münster, 48149, Münster, Germany
| | - Margus Punab
- Andrology Centre, Tartu University Hospital, 50406, Tartu, Estonia
- Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
| | - Christian Ruckert
- Institute of Human Genetics, University of Münster, 48149, Münster, Germany
| | - Liina Nagirnaja
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Kenneth I Aston
- Andrology and IVF Laboratory, Department of Surgery (Urology), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Donald F Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, 48149, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, 48149, Münster, Germany
| | - Daniela Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Gießen, 35392, Gießen, Germany
| | - Maris Laan
- Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
| | - Birgit Stallmeyer
- Institute of Reproductive Genetics, University of Münster, 48149, Münster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, 48149, Münster, Germany.
| |
Collapse
|
13
|
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
|
14
|
Differences in Adipose Gene Expression Profiles between Male and Female Even Reindeer (Rangifer tarandus) in Sakha (Yakutia). Genes (Basel) 2022; 13:genes13091645. [PMID: 36140812 PMCID: PMC9498357 DOI: 10.3390/genes13091645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Reindeer are native to harsh northern Eurasian environments which are characterized by long and cold winters, short summers, and limited pasture vegetation. Adipose tissues play a significant role in these animals by modulating energy metabolism, immunity, and reproduction. Here, we have investigated the transcriptome profiles of metacarpal, perirenal, and prescapular adipose tissues in Even reindeer and searched for genes that were differentially expressed in male and female individuals. A total of 15,551 genes were expressed, where the transcriptome profile of metacarpal adipose tissue was found to be distinct from that of perirenal and prescapular adipose tissues. Interestingly, 10 genes, including PRDM9, which is known to have an important role in adaptation and speciation in reindeer, were always upregulated in all three tissues of male reindeer.
Collapse
|
15
|
Sahota JS, Sharma B, Guleria K, Sambyal V. Candidate genes for infertility: an in-silico study based on cytogenetic analysis. BMC Med Genomics 2022; 15:170. [PMID: 35918717 PMCID: PMC9347124 DOI: 10.1186/s12920-022-01320-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/22/2022] [Indexed: 11/26/2022] Open
Abstract
Background The cause of infertility remains unclear in a significant proportion of reproductive-age couples who fail to conceive naturally. Chromosomal aberrations have been identified as one of the main genetic causes of male and female infertility. Structural chromosomal aberrations may disrupt the functioning of various genes, some of which may be important for fertility. The present study aims to identify candidate genes and putative functional interaction networks involved in male and female infertility using cytogenetic data from cultured peripheral blood lymphocytes of infertile patients. Methods Karyotypic analyses was done in 201 infertile patients (100 males and 101 females) and 201 age and gender matched healthy controls (100 males and 101 females) after 72 h peripheral lymphocyte culturing and GTG banding, followed by bioinformatic analysis using Cytoscape v3.8.2 and Metascape. Results Several chromosomal regions with a significantly higher frequency of structural aberrations were identified in the infertile males (5q2, 10q2, and 17q2) and females (6q2, 16q2, and Xq2). Segregation of the patients based on type of infertility (primary v/s secondary infertility) led to the identification of chromosomal regions with a significantly higher frequency of structural aberrations exclusively within the infertile males (5q2, 17q2) and females (16q2) with primary infertility. Cytoscape identified two networks specific to these regions: a male specific network with 99 genes and a female specific network with 109 genes. The top enriched GO terms within the male and female infertility networks were “skeletal system morphogenesis” and “mRNA transport” respectively. PSME3, PSMD3, and CDC27 were the top 3 hub genes identified within the male infertility network. Similarly, UPF3B, IRF8, and PSMB1 were the top 3 hub genes identified with the female infertility network. Among the hub genes identified in the male- and female-specific networks, PSMB1, PSMD3, and PSME3 are functional components of the proteasome complex. These hub genes have a limited number of reports related to their respective roles in maintenance of fertility in mice model and humans and require validation in further studies. Conclusion The candidate genes predicted in the present study can serve as targets for future research on infertility. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01320-x.
Collapse
Affiliation(s)
- Jatinder Singh Sahota
- Department of Human Genetics, Cytogenetics Laboratory, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India
| | - Bhavna Sharma
- Department of Human Genetics, Cytogenetics Laboratory, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India
| | - Kamlesh Guleria
- Department of Human Genetics, Cytogenetics Laboratory, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India
| | - Vasudha Sambyal
- Department of Human Genetics, Cytogenetics Laboratory, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India.
| |
Collapse
|
16
|
Ocañas SR, Ansere VA, Tooley KB, Hadad N, Chucair-Elliott AJ, Stanford DR, Rice S, Wronowski B, Pham KD, Hoffman JM, Austad SN, Stout MB, Freeman WM. Differential Regulation of Mouse Hippocampal Gene Expression Sex Differences by Chromosomal Content and Gonadal Sex. Mol Neurobiol 2022; 59:4669-4702. [PMID: 35589920 PMCID: PMC9119800 DOI: 10.1007/s12035-022-02860-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/25/2022] [Indexed: 01/23/2023]
Abstract
Common neurological disorders, like Alzheimer's disease (AD), multiple sclerosis (MS), and autism, display profound sex differences in prevalence and clinical presentation. However, sex differences in the brain with health and disease are often overlooked in experimental models. Sex effects originate, directly or indirectly, from hormonal or sex chromosomal mechanisms. To delineate the contributions of genetic sex (XX v. XY) versus gonadal sex (ovaries v. testes) to the epigenomic regulation of hippocampal sex differences, we used the Four Core Genotypes (FCG) mouse model which uncouples chromosomal and gonadal sex. Transcriptomic and epigenomic analyses of ~ 12-month-old FCG mouse hippocampus, revealed genomic context-specific regulatory effects of genotypic and gonadal sex on X- and autosome-encoded gene expression and DNA modification patterns. X-chromosomal epigenomic patterns, classically associated with X-inactivation, were established almost entirely by genotypic sex, independent of gonadal sex. Differences in X-chromosome methylation were primarily localized to gene regulatory regions including promoters, CpG islands, CTCF binding sites, and active/poised chromatin, with an inverse relationship between methylation and gene expression. Autosomal gene expression demonstrated regulation by both genotypic and gonadal sex, particularly in immune processes. These data demonstrate an important regulatory role of sex chromosomes, independent of gonadal sex, on sex-biased hippocampal transcriptomic and epigenomic profiles. Future studies will need to further interrogate specific CNS cell types, identify the mechanisms by which sex chromosomes regulate autosomes, and differentiate organizational from activational hormonal effects.
Collapse
Affiliation(s)
- Sarah R Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13thStreet, Oklahoma City, OK, 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Victor A Ansere
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13thStreet, Oklahoma City, OK, 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla B Tooley
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13thStreet, Oklahoma City, OK, 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Ana J Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13thStreet, Oklahoma City, OK, 73104, USA
| | - David R Stanford
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13thStreet, Oklahoma City, OK, 73104, USA
| | - Shannon Rice
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13thStreet, Oklahoma City, OK, 73104, USA
| | - Benjamin Wronowski
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kevin D Pham
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13thStreet, Oklahoma City, OK, 73104, USA
| | - Jessica M Hoffman
- Department of Biology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Steven N Austad
- Department of Biology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael B Stout
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13thStreet, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
17
|
Lacroix M, Beauchemin H, Fraszczak J, Ross J, Shooshtarizadeh P, Chen R, Moroy T. The X-linked helicase DDX3X is required for lymphoid differentiation and MYC-driven lymphomagenesis. Cancer Res 2022; 82:3172-3186. [PMID: 35815807 DOI: 10.1158/0008-5472.can-21-2454] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/29/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
The X-linked gene DDX3X encodes an RNA helicase that is mutated at high frequencies in several types of human B-cell lymphoma. Females have two active DDX3X alleles and males carry a DDX3Y homolog on the Y chromosome. We show here that pan-hematopoietic, homozygous deletion of Ddx3x in female mice perturbs erythropoiesis, causing early developmental arrest. However, both hemizygous male and heterozygous female embryos develop normally, suggesting that one Ddx3x allele is sufficient for fetal hematopoietic development in females and that the Ddx3y allele can compensate for the loss of Ddx3x in males. In adult mice, DDX3X deficiency altered hematopoietic progenitors, early lymphoid development, marginal zone and germinal center B-cells, and lymphomagenesis in a sex-dependent manner. Loss of both Ddx3x alleles abrogated MYC-driven lymphomagenesis in females, while Ddx3x-deletion in males did not affect the formation of B-cell lymphoma in both mouse models. Moreover, tumors that appeared in male mice lacking DDX3X showed upregulated expression of DDX3Y, indicating a critical requirement for DDX3 activity for lymphomagenesis. These data reveal sex-specific roles of DDX3X in erythro- and lymphopoiesis as well as in MYC-driven lymphomagenesis.
Collapse
Affiliation(s)
- Marion Lacroix
- IRCM (Institut de Recherches Cliniques de Montr�al), Montreal, Quebec, Canada
| | | | | | - Julie Ross
- Montreal Clinical Research Institute, Montreal, Quebec, Canada
| | | | | | - Tarik Moroy
- Institut de recherches cliniques de Montr�al, Montreal, Canada
| |
Collapse
|
18
|
Vogt PH, Rauschendorf MA, Zimmer J, Drummer C, Behr R. AZFa Y gene, DDX3Y, evolved novel testis transcript variants in primates with proximal 3´UTR polyadenylation for germ cell specific translation. Sci Rep 2022; 12:8954. [PMID: 35624115 PMCID: PMC9142519 DOI: 10.1038/s41598-022-12474-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/31/2022] [Indexed: 01/15/2023] Open
Abstract
Translational control is a major level of gene expression regulation in the male germ line. DDX3Y located in the AZFa region of the human Y chromosome encodes a conserved RNA helicase important for translational control at the G1-S phase of the cell cycle. In human, DDX3Y protein is expressed only in premeiotic male germ cells. In primates, DDX3Y evolved a second promoter producing novel testis-specific transcripts. Here, we show primate species-specific use of alternative polyadenylation (APA) sites for these testis-specific DDX3Y transcript variants. They have evolved subsequently in the 3´UTRs of the primates´ DDX3Y transcripts. Whereas a distal APA site (PAS4) is still used for polyadenylation of most DDX3Y testis transcripts in Callithrix jacchus; two proximal APAs (PAS1; PAS2) are used predominantly in Macaca mulatta, in Pan trogloydates and in human. This shift corresponds with a significant increase of DDX3Y protein expression in the macaque testis tissue. In chimpanzee and human, shift to predominant use of the most proximal APA site (PAS1) is associated with translation of these DDX3Y transcripts in only premeiotic male germ cells. We therefore assume evolution of a positive selection process for functional DDX3Y testis transcripts in these primates which increase their stability and translation efficiency to promote its cell cycle balancing function in the human male germ line.
Collapse
Affiliation(s)
- P. H. Vogt
- Division of Reproduction Genetics, Department of Gynecological Endocrinology and Fertility Disorders, University Women Hospital, Im Neuenheimer Feld 440, D-69120 Heidelberg, Germany
| | - M-A. Rauschendorf
- Molecular Health GmbH, Kurfürsten-Anlage 21, D-69115 Heidelberg, Germany
| | - J. Zimmer
- Division of Reproduction Genetics, Department of Gynecological Endocrinology and Fertility Disorders, University Women Hospital, Im Neuenheimer Feld 440, D-69120 Heidelberg, Germany
| | - C. Drummer
- grid.418215.b0000 0000 8502 7018Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, D-37077 Göttingen, Germany
| | - R. Behr
- grid.418215.b0000 0000 8502 7018Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, D-37077 Göttingen, Germany
| |
Collapse
|
19
|
Tran KTD, Valli-Pulaski H, Colvin A, Orwig KE. Male fertility preservation and restoration strategies for patients undergoing gonadotoxic therapies†. Biol Reprod 2022; 107:382-405. [PMID: 35403667 PMCID: PMC9382377 DOI: 10.1093/biolre/ioac072] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 11/22/2022] Open
Abstract
Medical treatments for cancers or other conditions can lead to permanent infertility. Infertility is an insidious disease that impacts not only the ability to have a biological child but also the emotional well-being of the infertile individuals, relationships, finances, and overall health. Therefore, all patients should be educated about the effects of their medical treatments on future fertility and about fertility preservation options. The standard fertility preservation option for adolescent and adult men is sperm cryopreservation. Sperms can be frozen and stored for a long period, thawed at a later date, and used to achieve pregnancy with existing assisted reproductive technologies. However, sperm cryopreservation is not applicable for prepubertal patients who do not yet produce sperm. The only fertility preservation option available to prepubertal boys is testicular tissue cryopreservation. Next-generation technologies are being developed to mature those testicular cells or tissues to produce fertilization-competent sperms. When sperm and testicular tissues are not available for fertility preservation, inducing pluripotent stem cells derived from somatic cells, such as blood or skin, may provide an alternative path to produce sperms through a process call in vitro gametogenesis. This review describes standard and experimental options to preserve male fertility as well as the experimental options to produce functional spermatids or sperms from immature cryopreserved testicular tissues or somatic cells.
Collapse
Affiliation(s)
- Kien T D Tran
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Hanna Valli-Pulaski
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Amanda Colvin
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Kyle E Orwig
- Correspondence: Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA 15213, USA. Tel: 412-641-2460; E-mail:
| |
Collapse
|
20
|
Vogt PH, Zimmer J, Bender U, Strowitzki T. AZFa candidate gene UTY and its X homologue UTX are expressed in human germ cells. REPRODUCTION AND FERTILITY 2022; 2:151-160. [PMID: 35128450 PMCID: PMC8812439 DOI: 10.1530/raf-20-0049] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
The Ubiquitous Transcribed Y (UTY a.k.a. KDM6C) AZFa candidate gene on the human Y chromosome and its paralog on the X chromosome, UTX (a.k.a. KDM6A), encode a histone lysine demethylase removing chromatin H3K27 methylation marks at genes transcriptional start sites for activation. Both proteins harbour the conserved Jumonji C (JmjC) domain, functional in chromatin metabolism, and an extended N-terminal tetratricopeptide repeat (TPR) block involved in specific protein interactions. Specific antisera for human UTY and UTX proteins were developed to distinguish the expression of both proteins in human germ cells by immunohistochemical experiments on appropriate tissue sections. In the male germ line, UTY was expressed in the fraction of A spermatogonia located at the basal membrane, probably including spermatogonia stem cells. UTX expression was more spread in all spermatogonia and in early spermatids. In female germ line, UTX expression was found in the primordial germ cells of the ovary. UTY was also expressed during fetal male germ cell development, whereas UTX expression was visible only at distinct gestation weeks. Based on these results and the conserved neighboured location of UTY and DDX3Y in Yq11 found in mammals of distinct lineages, we conclude that UTY, such as DDX3Y, is part of the Azoospermia factor a (AZFa) locus functioning in human spermatogonia to support the balance of their proliferation-differentiation rate before meiosis. Comparable UTY and DDX3Y expression was also found in gonadoblastoma and dysgerminoma cells found in germ cell nests of the dysgenetic gonads of individuals with disorders of sexual development and a Y chromosome in karyotype (DSD-XY). This confirms that AZFa overlaps with GBY, the Gonadoblastoma susceptibility Y locus, and includes the UTY gene.
Collapse
Affiliation(s)
- Peter H Vogt
- Division of Reproduction Genetics, Department of, Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Germany
| | - Jutta Zimmer
- Division of Reproduction Genetics, Department of, Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Germany
| | - Ulrike Bender
- Division of Reproduction Genetics, Department of, Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Germany
| | - Thomas Strowitzki
- Department of Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Germany
| |
Collapse
|
21
|
Zhang X, Liao Z, Tang S, Yuan Z, Li F, Yue X. A mutation modulating DDX3Y gene expression cosegregates with the major Y-chromosomal haplogroups and with testis size in Hu sheep. Anim Genet 2021; 53:193-202. [PMID: 34963194 DOI: 10.1111/age.13166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/12/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022]
Abstract
Variations in the Y-chromosome are usually correlated with male-specific traits. However, this condition has been described only sporadically, even in human genetics. The present study was conducted to clone the full-length gene sequence of ovine DEAD-box helicase 3, Y-linked (DDX3Y), and investigate the effect of the expression and variation of DDX3Y on the reproductive traits of Hu sheep. Consequently, we identified the full coding sequence and genomic sequence of ovine DDX3Y. Quantitative PCR (qPCR) analysis showed that ovine DDX3Y was highly expressed in testis, and the expression level increased during testicular development. Furthermore, individuals with larger testis at 6 months expressed significantly more DDX3Y mRNA in the testis than individuals with smaller testis. Notably, a novel SNP (g. 12657 C>A) in the 3' untranslated region was identified in Hu sheep and Tan sheep according to the investigation of the full DDX3Y genomic sequence of 1069 individuals from nine sheep breeds. Association analysis revealed that the SNP was significantly related to testis size in Hu sheep. Meanwhile, Hu rams with the derived C allele showed significantly higher expression levels of DDX3Y in testis than those with the ancestral A allele. In addition, data mining in a previous study showed that the C allele cosegregated with the globally major Y-chromosomal haplogroups y-HA and y-HC, and the A allele is found in all rams with haplogroups y-HB1, y-HB2 and y-HD. This study suggests that the association of the Y-chromosomal haplogroups with testis size in Hu sheep can be extrapolated to the sheep population worldwide.
Collapse
Affiliation(s)
- Xueying Zhang
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Zhi Liao
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Shijie Tang
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Zehu Yuan
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education, Yangzhou University, Yangzhou, China
| | - Fadi Li
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Xiangpeng Yue
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| |
Collapse
|
22
|
Venkataramanan S, Gadek M, Calviello L, Wilkins K, Floor SN. DDX3X and DDX3Y are redundant in protein synthesis. RNA (NEW YORK, N.Y.) 2021; 27:1577-1588. [PMID: 34535544 PMCID: PMC8594478 DOI: 10.1261/rna.078926.121] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
DDX3 is a DEAD-box RNA helicase that regulates translation and is encoded by the X- and Y-linked paralogs DDX3X and DDX3Y While DDX3X is ubiquitously expressed in human tissues and essential for viability, DDX3Y is male-specific and shows lower and more variable expression than DDX3X in somatic tissues. Heterozygous genetic lesions in DDX3X mediate a class of developmental disorders called DDX3X syndrome, while loss of DDX3Y is implicated in male infertility. One possible explanation for female-bias in DDX3X syndrome is that DDX3Y encodes a polypeptide with different biochemical activity. In this study, we use ribosome profiling and in vitro translation to demonstrate that the X- and Y-linked paralogs of DDX3 play functionally redundant roles in translation. We find that transcripts that are sensitive to DDX3X depletion or mutation are rescued by complementation with DDX3Y. Our data indicate that DDX3X and DDX3Y proteins can functionally complement each other in the context of mRNA translation in human cells. DDX3Y is not expressed in a large fraction of the central nervous system. These findings suggest that expression differences, not differences in paralog-dependent protein synthesis, underlie the sex-bias of DDX3X-associated diseases.
Collapse
Affiliation(s)
- Srivats Venkataramanan
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Margaret Gadek
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Lorenzo Calviello
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kevin Wilkins
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Stephen N Floor
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
23
|
Huang R, Zheng Z, Liu S, Yan P, Song D, Yin H, Hu P, Zhu X, Chang Z, Liu Y, Zhuang J, Meng T, Huang Z, Zhang J. Identification of prognostic and bone metastasis-related alternative splicing signatures in mesothelioma. Cancer Med 2021; 10:4478-4492. [PMID: 34041868 PMCID: PMC8267146 DOI: 10.1002/cam4.3977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 01/18/2023] Open
Abstract
Mesothelioma (MESO) is an infrequent tumor derived from mesothelial cells of pleura, peritoneum, pericardium, and tunica vaginalis testis. Despite advancement in technologies and better understanding of tumor progression mechanism, the prognosis of MESO remains poor. The role of alternative splicing events (ASEs) in the oncogenesis, tumor metastasis and drug resistance has been widely discussed in multiple cancers. But the prognosis and potential therapeutic value of ASEs in MESO were not clearly studied by now. We constructed a prognostic model using RNA sequencing data and matched ASE data of MESO patients obtained from the TCGA and TCGASpliceSeq database. A total of 3,993 ASEs were identified associated with overall survival using Cox regression analysis. Eight of them were finally figured out to institute the model by lasso regression analysis. The risk score of the model can predict the prognosis independently. Among the identified 390 splicing factors (SF), HSPA1A and DDX3Y was significantly associated with 43 OS-SEs. Among these OS-SEs, SNX5-58744-AT (p = 0.048) and SNX5-58745-AT (p = 0.048) were significantly associated with bone metastasis. Co-expression analysis of signal pathways and SNX5-58744-AT, SNX5-58745-AT was also depicted using GSVA. Finally, we proposed that splicing factor (SF) HSPA1A could regulate SNX5-58744-AT (R = -0.414) and SNX5-58745-AT (R = 0.414) through the pathway "Class I MHC mediated antigen processing and presentation" (R = 0.400). In this way, tumorigenesis and bone metastasis of MESO were controlled.
Collapse
Affiliation(s)
- Runzhi Huang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Tongji University School of Medicine, Shanghai, China
| | - Zixuan Zheng
- Tongji University School of Medicine, Shanghai, China
| | - Sijia Liu
- Tongji University School of Medicine, Shanghai, China
| | - Penghui Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Huabin Yin
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Peng Hu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaolong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengyan Chang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yihan Liu
- Tongji University School of Medicine, Shanghai, China
| | - Juanwei Zhuang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tong Meng
- Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zongqiang Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Zhang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Sánchez JM, Gómez-Redondo I, Browne JA, Planells B, Gutiérrez-Adán A, Lonergan P. MicroRNAs in amniotic fluid and maternal blood plasma associated with sex determination and early gonad differentiation in cattle†. Biol Reprod 2021; 105:345-358. [PMID: 33889937 PMCID: PMC8335352 DOI: 10.1093/biolre/ioab079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/22/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022] Open
Abstract
We hypothesized that sexually dimorphic differences exist in the expression of miRNAs in amniotic fluid (AF) and maternal blood plasma (MP) in association with the process of sex determination and gonad differentiation in cattle. Amniotic fluid and MP were collected from six pregnant heifers (three carrying a single male and three a single female embryo) following slaughter on Day 39 postinsemination, coinciding with the peak of SRY expression. Samples (six AF and six MP) were profiled using an miRNA Serum/Plasma Focus PCR Panel. Differentially expressed (DE) miRNAs were identified in AF (n = 5) and associated MP (n = 56) of male vs. female embryos (P < 0.05). Functional analysis showed that inflammatory and immune response were among the 13 biological processes enriched by miRNAs DE in MP in the male group (FDR < 0.05), suggesting that these sex-dependent DE miRNAs may be implicated in modulating the receptivity of the dam to a male embryo. Further, we compared the downstream targets of the sex-dependent DE miRNAs detected in MP with genes previously identified as DE in male vs. female genital ridges. The analyses revealed potential targets that might be important during this developmental stage such as SHROOM2, DDX3Y, SOX9, SRY, PPP1CB, JARID2, USP9X, KDM6A, and EIF2S3. Results from this study highlight novel aspects of sex determination and embryo–maternal communication in cattle such as the potential role of miRNAs in gonad development as well as in the modulation of the receptivity of the dam to a male embryo.
Collapse
Affiliation(s)
- José María Sánchez
- Animal and Crops Sciences, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland.,Departamento de Reproducción Animal, INIA, Madrid, Spain
| | | | - John A Browne
- Animal and Crops Sciences, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | - Pat Lonergan
- Animal and Crops Sciences, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
25
|
Pandey A, Jaiswal A, Tiwari M, Ali A, Sharma R. Yq AZF microdeletions in male infertility: An update on the phenotypic spectrum, epidemiology and diagnostics. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2021. [DOI: 10.4103/2305-0500.326718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
26
|
Punjani N, Kang C, Schlegel PN. Clinical implications of Y chromosome microdeletions among infertile men. Best Pract Res Clin Endocrinol Metab 2020; 34:101471. [PMID: 33214080 DOI: 10.1016/j.beem.2020.101471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Male factor infertility contributes significantly to couples facing difficulty achieving a pregnancy. Genetic factors, and specifically those related to the Y chromosome, may occur in up to 15% of men with oligozoospermia or azoospermia. A subset of loci within the Y chromosome, known as the azoospermia factors (AZFa, AZFb, and AZFc), have been associated with male infertility. Emerging evidence has demonstrated that microdeletions of at least a subset of these regions may also have impacts on systemic conditions. This review provides a brief review of male infertility and the structure of the Y chromosome, and further highlights the role of Y chromosome microdeletions in male infertility and other systemic disease.
Collapse
Affiliation(s)
- Nahid Punjani
- Division of Urology, Weill Cornell Medical College, New York, NY, USA
| | - Caroline Kang
- Division of Urology, Weill Cornell Medical College, New York, NY, USA.
| | - Peter N Schlegel
- Division of Urology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
27
|
Hwang YS, Suzuki S, Seita Y, Ito J, Sakata Y, Aso H, Sato K, Hermann BP, Sasaki K. Reconstitution of prospermatogonial specification in vitro from human induced pluripotent stem cells. Nat Commun 2020; 11:5656. [PMID: 33168808 PMCID: PMC7653920 DOI: 10.1038/s41467-020-19350-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Establishment of spermatogonia throughout the fetal and postnatal period is essential for production of spermatozoa and male fertility. Here, we establish a protocol for in vitro reconstitution of human prospermatogonial specification whereby human primordial germ cell (PGC)-like cells differentiated from human induced pluripotent stem cells are further induced into M-prospermatogonia-like cells and T1 prospermatogonia-like cells (T1LCs) using long-term cultured xenogeneic reconstituted testes. Single cell RNA-sequencing is used to delineate the lineage trajectory leading to T1LCs, which closely resemble human T1-prospermatogonia in vivo and exhibit gene expression related to spermatogenesis and diminished proliferation, a hallmark of quiescent T1 prospermatogonia. Notably, this system enables us to visualize the dynamic and stage-specific regulation of transposable elements during human prospermatogonial specification. Together, our findings pave the way for understanding and reconstructing human male germline development in vitro. Spermatogonia establishment in the fetal and postnatal period is essential for spermatozoa production. Here the authors present a protocol for in vitro reconstitution of human prospermatogonial specification and perform single cell RNA-sequencing to delineate lineage trajectories.
Collapse
Affiliation(s)
- Young Sun Hwang
- Institute for Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Shinnosuke Suzuki
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Yasunari Seita
- Institute for Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Bell Research Center for Reproductive Health and Cancer, Nagoya, Aichi, Japan
| | - Jumpei Ito
- Division of Systems Virology, Department of infectious Disease Control, International Research Center for infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, 1088639, Japan
| | - Yuka Sakata
- Institute for Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Hirofumi Aso
- Division of Systems Virology, Department of infectious Disease Control, International Research Center for infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, 1088639, Japan
| | - Kei Sato
- Division of Systems Virology, Department of infectious Disease Control, International Research Center for infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, 1088639, Japan
| | - Brian P Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Kotaro Sasaki
- Institute for Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
28
|
Haddad M, Stewart J, Xie P, Cheung S, Trout A, Keating D, Parrella A, Lawrence S, Rosenwaks Z, Palermo GD. Thoughts on the popularity of ICSI. J Assist Reprod Genet 2020; 38:101-123. [PMID: 33155089 PMCID: PMC7823003 DOI: 10.1007/s10815-020-01987-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/18/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose Intracytoplasmic sperm injection (ICSI) is the most widely utilized assisted reproductive technique (ART) worldwide. In this feature, we review the early assisted fertilization attempts that eventually led to the development of ICSI, and discuss its current utilization in cases of male and non-male factor infertility. Methods We researched the literature related to the development, indications, and current use of ICSI, such as sperm structural abnormalities, male genetic indications, surgically retrieved sperm, high sperm chromatin fragmentation, oocyte dysmorphism, and preimplantation genetic testing (PGT). We also describe the potential future applications of ICSI. Results This review summarizes the early micromanipulation techniques that led to the inception of ICSI. We also explore its current indications, including non-male factor infertility, where its use is more controversial. Finally, we consider the benefits of future advancements in reproductive biology that may incorporate ICSI, such as in vitro spermatogenesis, neogametogenesis, and heritable genome editing. Conclusion The versatility, consistency, and reliability of ICSI have made it the most prevalently utilized ART procedure worldwide.
Collapse
Affiliation(s)
- Mounia Haddad
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Joshua Stewart
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Philip Xie
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Stephanie Cheung
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Aysha Trout
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Derek Keating
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alessandra Parrella
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sherina Lawrence
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Zev Rosenwaks
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Gianpiero D Palermo
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
29
|
Marker-assisted selection vis-à-vis bull fertility: coming full circle-a review. Mol Biol Rep 2020; 47:9123-9133. [PMID: 33099757 DOI: 10.1007/s11033-020-05919-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/13/2020] [Indexed: 10/23/2022]
Abstract
Bull fertility is considered an indispensable trait, as far as farm economics is concerned since it is the successful conception in a cow that provides calf crop, along with the ensuing lactation. This ensures sustainability of a dairy farm. Traditionally, bull fertility did not receive much attention by the farm managers and breeding animals were solely evaluated based on phenotypic predictors, namely, sire conception rate and seminal parameters in bull. With the advent of the molecular era in animal breeding, attempts were made to unravel the genetic complexity of bull fertility by the identification of genetic markers related to the trait. Marker-Assisted Selection (MAS) is a methodology that aims at utilizing the genetic information at markers and selecting improved populations for important traits. Traditionally, MAS was pursued using a candidate gene approach for identifying markers related to genes that are already known to have a physiological function related to the trait but this approach had certain shortcomings like stringent criteria for significance testing. Now, with the availability of genome-wide data, the number of markers identified and variance explained in relation to bull fertility has gone up. So, this presents a unique opportunity to revisit MAS by selection based on the information of a large number of genome-wide markers and thus, improving the accuracy of selection.
Collapse
|
30
|
Vogt PH, Besikoglu B, Bettendorf M, Frank-Herrmann P, Zimmer J, Bender U, Knauer-Fischer S, Choukair D, Sinn P, Lau YFC, Heidemann PH, Strowitzki T. Gonadoblastoma Y locus genes expressed in germ cells of individuals with dysgenetic gonads and a Y chromosome in their karyotypes include DDX3Y and TSPY. Hum Reprod 2020; 34:770-779. [PMID: 30753444 DOI: 10.1093/humrep/dez004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/17/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Which Y genes mapped to the 'Gonadoblastoma Y (GBY)' locus on human Y chromosome are expressed in germ cells of individuals with some Differences of Sexual Development (DSD) and a Y chromosome in their karyotype (DSD-XY groups)? SUMMARY ANSWER The GBY candidate genes DDX3Y and TSPY are expressed in the germ cells of DSD-XY patients from distinct etiologies: patients with mixed gonadal dysgenesis (MGD) and sex chromosome mosaics (45,X0/46,XY; 46,XX/46,XY); patients with complete androgen insensitivity (CAIS), patients with complete gonadal dysgenesis (CGD; e.g. Swyer syndrome). WHAT IS KNOWN ALREADY A GBY locus was proposed to be present on the human Y chromosome because only DSD patients with a Y chromosome in their karyotype have a high-although variable-risk (up to 55%) for germ cell tumour development. GBY was mapped to the proximal part of the short and long Y arm. TSPY located in the proximal part of the short Y arm (Yp11.1) was found to be a strong GBY candidate gene. It is expressed in the germ cells of DSD-XY patients with distinct etiologies but also in foetal and pre-meiotic male spermatogonia. However, the GBY region extends to proximal Yq11 and therefore includes probably more than one candidate gene. STUDY DESIGN, SIZE, DURATION Protein expression of the putative GBY candidate gene in proximal Yq11, DDX3Y, is compared with that of TSPY in serial gonadal tissue sections of 40 DSD-XY individuals from the three DSD patient groups (MGD, Complete Androgen Insensitivity Syndrome [CAIS], CGD) with and without displaying malignancy. Expression of OCT3/4 in the same tissue samples marks the rate of pluripotent germ cells. PARTICIPANTS/MATERIALS, SETTING, METHOD A total of 145 DSD individuals were analysed for the Y chromosome to select the DSD-XY subgroup. PCR multiplex assays with Y gene specific marker set score for putative microdeletions in GBY Locus. Immunohistochemical experiments with specific antisera mark expression of the GBY candidate proteins, DDX3Y, TSPY, in serial sections of the gonadal tissue samples; OCT3/4 expression analyses in parallel reveal the pluripotent germ cell fraction. MAIN RESULTS AND THE ROLE OF CHANCE Similar DDX3Y and TSPY protein expression patterns were found in the germ cells of DSD-XY patients from each subgroup, independent of age. In CAIS patients OCT3/4 expression was often found only in a fraction of these germ cells. This suggest that GBY candidate proteins are also expressed in the non-malignant germ cells of DSD-XY individuals like in male spermatogonia. LIMITATIONS, REASONS FOR CAUTION Variation of the expression profiles of GBY candidate genes in the germ cells of some DSD-XY individuals suggests distinct transcriptional and translational control mechanisms which are functioning during expression of these Y genes in the DSD-XY germ cells. Their proposed GBY tumour susceptibility function to transform these germ cells to pre-malignant GB/Germ Cell Neoplasia in Situ (GB/GCNIS) cells seems therefore to be limited and depending on their state of pluripotency. WIDER IMPLICATIONS OF THE FINDINGS These experimental findings are of general importance for each individual identified in the clinic with DSD and a Y chromosome in the karyotype. To judge their risk of germ cell tumour development, OCT3/4 expression analyses on their gonadal tissue section is mandatory to reveal the fraction of germ cells still being pluripotent. Comparative expression analysis of the GBY candidate genes can be helpful to reveal the fraction of germ cells with genetically still activated Y chromosomes contributing to further development of malignancy if at high expression level. STUDY FUNDING/COMPETING INTEREST(S) This research project was supported by a grant (01GM0627) from the BMBF (Bundesministerium für Bildung und Forschung), Germany to P.H.V. and B.B. The authors have no competing interests.
Collapse
Affiliation(s)
- P H Vogt
- Division of Reproduction Genetics, Department of Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Heidelberg, Germany
| | - B Besikoglu
- Division of Reproduction Genetics, Department of Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Heidelberg, Germany.,Novum, Center for Reproductive Medicine, Akazienallee 8, Essen, Germany
| | - M Bettendorf
- Division of Paediatric Endocrinology and Diabetes, Children Hospital, University of Heidelberg, Heidelberg, Germany
| | - P Frank-Herrmann
- Department of Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Heidelberg, Germany
| | - J Zimmer
- Division of Reproduction Genetics, Department of Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Heidelberg, Germany
| | - U Bender
- Division of Reproduction Genetics, Department of Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Heidelberg, Germany
| | - S Knauer-Fischer
- Division of Paediatric Endocrinology and Diabetes, Children Hospital, University of Heidelberg, Heidelberg, Germany
| | - D Choukair
- Division of Paediatric Endocrinology and Diabetes, Children Hospital, University of Heidelberg, Heidelberg, Germany
| | - P Sinn
- Division of Gynaecopathology, Department of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany
| | - Y-F C Lau
- Division of Cell & Developmental Genetics, Department of Medicine, VAMC 111C5, San Francisco, CA, USA
| | - P H Heidemann
- Children Hospital Augsburg I, Academic Hospital of University of Munich, Stenglinstraβe 2, Augsburg, Germany
| | - T Strowitzki
- Department of Gynaecol. Endocrinology & Infertility Disorders, Women Hospital, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
31
|
Fang F, Li Z, Zhao Q, Ye Z, Gu X, Pan F, Li H, Xiang W, Xiong C. Induced Pluripotent Stem Cells Derived From Two Idiopathic Azoospermia Patients Display Compromised Differentiation Potential for Primordial Germ Cell Fate. Front Cell Dev Biol 2020; 8:432. [PMID: 32671061 PMCID: PMC7331483 DOI: 10.3389/fcell.2020.00432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022] Open
Abstract
At present, the etiology of most non-obstructive azoospermia (NOA) remains unclear. In vitro generation of patient-specific induced pluripotent stem cells (iPSCs) is an effective approach for exploring the mechanisms of human disease. Here, we established iPSCs from two patients with idiopathic NOA and differentiated them into primordial germ cell-like cells (PGCLCs) in vitro. Compared with iPSCs derived from normal fertile men, the NOA patient-specific iPSCs show decreased efficiency of PGCLC formation in vitro. Particularly, the embryoids derived from NOA patient-specific iPSCs show defects in the expression of early primordial germ cell (PGC) genes. The transcriptome analysis reveals the expression patterns of key human PGC genes are generally similar in PGCLCs differentiated from all iPSC lines, and the differentially expressed genes were enriched with gene ontology (GO) of cell cycle and apoptosis regulation. Moreover, the PGCLCs derived from NOA patient-specific iPSCs might have initiated epigenetic reprogramming at a very early stage. Thus, the NOA patient-specific iPSCs exhibit poor response to germ cell induction in vitro, which may be related to the regulation of apoptotic process. These findings provide a foundation for future research on mechanism of male infertility.
Collapse
Affiliation(s)
- Fang Fang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zili Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Tongji Reproductive Medicine Hospital, Wuhan, China
| | - Qian Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Ye
- Wuhan Tongji Reproductive Medicine Hospital, Wuhan, China
| | - Xiuli Gu
- Wuhan Tongji Reproductive Medicine Hospital, Wuhan, China
| | - Feng Pan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Honggang Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Tongji Reproductive Medicine Hospital, Wuhan, China
| | - Wenpei Xiang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Tongji Reproductive Medicine Hospital, Wuhan, China
| | - Chengliang Xiong
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Tongji Reproductive Medicine Hospital, Wuhan, China
| |
Collapse
|
32
|
Discrete roles of RNA helicases in human male germline and spermatogenesis. J Appl Genet 2020; 61:415-419. [PMID: 32328902 PMCID: PMC7413869 DOI: 10.1007/s13353-020-00558-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/05/2022]
Abstract
RNA helicases are known from their ability to bind and unwind double-stranded RNA initiating RNA processing events. These evolutionary conserved RNA binding proteins are broadly expressed in a variety of tissues; however, we can distinguish those, which represent tissue-specific expression pattern and play unique roles in certain cell lineages. For instance, some RNA helicases mediate transcriptomic changes triggering cell differentiation which results in specification and establishment of germline in a developing embryo. Others act as safeguards responsible for maintenance of DNA integrity in germ cell. In this article, we focus on selected DEAD/DEAH-box RNA helicases involved in germline development and spermatogenesis presenting their diverse functions and implications for male fertility.
Collapse
|
33
|
The Drosophila RNA Helicase Belle (DDX3) Non-Autonomously Suppresses Germline Tumorigenesis Via Regulation of a Specific mRNA Set. Cells 2020; 9:cells9030550. [PMID: 32111103 PMCID: PMC7140462 DOI: 10.3390/cells9030550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/28/2022] Open
Abstract
DDX3 subfamily DEAD-box RNA helicases are essential developmental regulators of RNA metabolism in eukaryotes. belle, the single DDX3 ortholog in Drosophila, is required for fly viability, fertility, and germline stem cell maintenance. Belle is involved both in translational activation and repression of target mRNAs in different tissues; however, direct targets of Belle in the testes are essentially unknown. Here we showed that belle RNAi knockdown in testis cyst cells caused a disruption of adhesion between germ and cyst cells and generation of tumor-like clusters of stem-like germ cells. Ectopic expression of β-integrin in cyst cells rescued early stages of spermatogenesis in belle knockdown testes, indicating that integrin adhesion complexes are required for the interaction between somatic and germ cells in a cyst. To address Belle functions in spermatogenesis in detail we performed cross-linking immunoprecipitation and sequencing (CLIP-seq) analysis and identified multiple mRNAs that interacted with Belle in the testes. The set of Belle targets includes transcripts of proteins that are essential for preventing the tumor-like clusters of germ cells and for sustaining spermatogenesis. By our hypothesis, failures in the translation of a number of mRNA targets additively contribute to developmental defects observed in the testes with belle knockdowns both in cyst cells and in the germline.
Collapse
|
34
|
Jia Y, Niu ZG, Li WY, Qin Q, Sun TT, Zhang F, Liu SR. A fertile male with a single sY86 deletion on the Y chromosome. Asian J Androl 2019; 22:333-334. [PMID: 31535627 PMCID: PMC7275800 DOI: 10.4103/aja.aja_94_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Yin Jia
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zi-Guang Niu
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China.,Department of Laboratory Medicine, Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Wei-Yu Li
- Obstetrics and Gynecology Hospital, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Qin Qin
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Ting-Ting Sun
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Shan-Rong Liu
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
35
|
Kerr CL, Bol GM, Vesuna F, Raman V. Targeting RNA helicase DDX3 in stem cell maintenance and teratoma formation. Genes Cancer 2019; 10:11-20. [PMID: 30899416 PMCID: PMC6420792 DOI: 10.18632/genesandcancer.187] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
DDX3 is an RNA helicase that has antiapoptotic properties, and promotes proliferation and transformation. Besides the role of DDX3 in transformed cells, there is evidence to indicate that DDX3 expression is at its highest levels during early embryonic development and is also expressed in germ cells of adults. Even though there is a distinct pattern of DDX3 expression during embryonic development and in adults, very little is known regarding its role in embryonic stem cells and pluripotency. In this work, we examined the relationship between DDX3 and human embryonic stem cells and its differentiated lineages. DDX3 expression was analyzed by immunohistochemistry in human embryonic stem cells and embryonal carcinoma cells. From the data obtained, it was evident that DDX3 was overexpressed in undifferentiated stem cells compared to differentiated cells. Moreover, when DDX3 expression was abrogated in multiple stem cells, proliferation was decreased, but differentiation was facilitated. Importantly, this resulted in reduced potency to induce teratoma formation. Taken together, these findings indicate a distinct role for DDX3 in stem cell maintenance.
Collapse
Affiliation(s)
- Candace L Kerr
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guus M Bol
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, University Medical Center Utrecht Cancer Center, GA Utrecht, The Netherlands
| | - Farhad Vesuna
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, University Medical Center Utrecht Cancer Center, GA Utrecht, The Netherlands
| | - Venu Raman
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, University Medical Center Utrecht Cancer Center, GA Utrecht, The Netherlands
| |
Collapse
|
36
|
New insights into the genetics of spermatogenic failure: a review of the literature. Hum Genet 2019; 138:125-140. [PMID: 30656449 DOI: 10.1007/s00439-019-01974-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/09/2019] [Indexed: 12/23/2022]
Abstract
Genetic anomalies are known to affect about 15% of infertile patients with azoospermia or severe oligozoospermia. Despite a throughout diagnostic work-up, in up to the 72% of the male partners of infertile couples, no etiological factor can be found; hence, the cause of infertility remains unclear. Recently, several novel genetic causes of spermatogenic failure (SPGF) have been described. The aim of this review was to collect all the available evidence of SPGF genetics, matching data from in-vitro and animal models with those in human beings to provide a comprehensive and updated overview of the genes capable of affecting spermatogenesis. By reviewing the literature, we provided a list of 60 candidate genes for SPGF. Their investigation by Next Generation Sequencing in large cohorts of patients with apparently idiopathic infertility would provide new interesting data about their racial- and ethnic-related prevalence in infertile patients, likely raising the diagnostic yields. We propose a phenotype-based approach to identify the genes to look for.
Collapse
|
37
|
Matsumura T, Endo T, Isotani A, Ogawa M, Ikawa M. An azoospermic factor gene, Ddx3y and its paralog, Ddx3x are dispensable in germ cells for male fertility. J Reprod Dev 2019; 65:121-128. [PMID: 30613052 PMCID: PMC6473106 DOI: 10.1262/jrd.2018-145] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
About 10% of male infertile patients show abnormalities in spermatogenesis. The microdeletion of azoospermia factor a (AZFa) region of the Y chromosome is thought to be a
cause of spermatogenic failure. However, candidate gene responsible for the spermatogenic failure in AZFa deleted patients has not been elucidated yet. Using mice, we
explored the function of Ddx3y, a strong candidate gene in the Azfa region, and Ddx3x, a Ddx3y paralog on the X
chromosome, in spermatogenesis. We first generated Ddx3y KO male mice using CRISPR/Cas9 and found that the Ddx3y KO male mice show normal spermatogenesis,
produce morphologically normal spermatozoa, and sire healthy offspring. Because Ddx3x KO males were embryonic lethal, we next generated chimeric mice, which contain
Ddx3x and Ddx3y double KO (dKO) germ cells, and found that the dKO germ cells can differentiate into spermatozoa and transmit their mutant alleles to
offspring by normal mating. We conclude that Ddx3x and Ddx3y are dispensable for spermatogenesis at least in mice. Unlike human, mice have an additional
Ddx3y paralog D1pas1, that has been reported to be essential for spermatogenesis. These findings suggest that human and mouse DDX3 related proteins have
distinct differences in their functions.
Collapse
Affiliation(s)
- Takafumi Matsumura
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Tsutomu Endo
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.,Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Ayako Isotani
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.,Graduate School of Biological Sciences, Nara Institute of Science and Technology, Nara 630-0192, Japan
| | - Masaki Ogawa
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.,Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan.,The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
38
|
Jedidi I, Ouchari M, Yin Q. Sex chromosomes-linked single-gene disorders involved in human infertility. Eur J Med Genet 2018; 62:103560. [PMID: 31402110 DOI: 10.1016/j.ejmg.2018.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 10/01/2018] [Accepted: 10/24/2018] [Indexed: 10/28/2022]
Abstract
Human infertility is a healthcare problem that has a worldwide impact. Genetic causes of human infertility include chromosomal aneuploidies and rearrangements and single-gene defects. The sex chromosomes (X and Y) are critical players in human fertility since they contain several genes essential for sex determination and reproductive traits for both men and women. This paper provides a review of the most common sex chromosomes-linked single-gene disorders involved in human infertility and their corresponding phenotypes. In addition to the Y-linked SRY gene, which mutations may cause XY gonadal dysgenesis and sex reversal, the deletions of genes present in AZF regions of the Y chromosome (DAZ, RBMY, DBY and USP9Y genes) are implicated in varying degrees of spermatogenic dysfunction. Furthermore, a list of X-linked genes (KAL1, NR0B1, AR, TEX11, FMR1, PGRMC1, BMP15 and POF1 and 2 regions genes (XPNPEP2, POF1B, DACH2, CHM and DIAPH2)) were reported to have critical roles in pubertal and reproductive deficiencies in humans, affecting only men, only women or both sexes. Mutations in these genes may be transmitted to the offspring by a dominant or a recessive inheritance.
Collapse
Affiliation(s)
- Ines Jedidi
- Faculty of Medicine of Sousse, Sousse, Tunisia.
| | - Mouna Ouchari
- Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Qinan Yin
- Clinical Center, National Institutes of Health, Bethesda, MD, USA; Department of Obstetrics and Gynecology, China Meitan General Hospital, Beijing, China
| |
Collapse
|
39
|
Fang F, Angulo B, Xia N, Sukhwani M, Wang Z, Carey CC, Mazurie A, Cui J, Wilkinson R, Wiedenheft B, Irie N, Surani MA, Orwig KE, Reijo Pera RA. A PAX5-OCT4-PRDM1 developmental switch specifies human primordial germ cells. Nat Cell Biol 2018; 20:655-665. [PMID: 29713018 PMCID: PMC5970969 DOI: 10.1038/s41556-018-0094-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/23/2018] [Indexed: 02/02/2023]
Abstract
Dysregulation of genetic pathways during human germ cell development leads to infertility. Here, we analysed bona fide human primordial germ cells (hPGCs) to probe the developmental genetics of human germ cell specification and differentiation. We examined the distribution of OCT4 occupancy in hPGCs relative to human embryonic stem cells (hESCs). We demonstrated that development, from pluripotent stem cells to germ cells, is driven by switching partners with OCT4 from SOX2 to PAX5 and PRDM1. Gain- and loss-of-function studies revealed that PAX5 encodes a critical regulator of hPGC development. Moreover, an epistasis analysis indicated that PAX5 acts upstream of OCT4 and PRDM1. The PAX5-OCT4-PRDM1 proteins form a core transcriptional network that activates germline and represses somatic programmes during human germ cell differentiation. These findings illustrate the power of combined genome editing, cell differentiation and engraftment for probing human developmental genetics that have historically been difficult to study.
Collapse
Affiliation(s)
- Fang Fang
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT, USA.
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA.
| | - Benjamin Angulo
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Ninuo Xia
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, School of Medicine, Magee Women's Research Institute, Pittsburgh, PA, USA
| | - Zhengyuan Wang
- Genomic Medicine Division, Hematology Branch, NHLBI/NIH, Rockville, MD, USA
| | - Charles C Carey
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Aurélien Mazurie
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Jun Cui
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Royce Wilkinson
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Blake Wiedenheft
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Naoko Irie
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - M Azim Surani
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, School of Medicine, Magee Women's Research Institute, Pittsburgh, PA, USA
| | - Renee A Reijo Pera
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| |
Collapse
|
40
|
Nailwal M, Chauhan JB. Computational Analysis of High-Risk SNPs in Human DBY Gene Responsible for Male Infertility: A Functional and Structural Impact. Interdiscip Sci 2018. [PMID: 29520635 DOI: 10.1007/s12539-018-0290-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND DEAD-box helicase 3, Y-linked (DBY) is a candidate gene of the AZF region which is involved in spermatogenesis process. Mutations in the DBY gene may disrupt the spermatogenesis and lead to infertility in men. Identification of functionally neutral mutation from the disease-causing mutation is the biggest challenge in human genetic variation analysis. Owing to the importance of DBY in male infertility, functional analysis was carried out to reveal the association between genetic mutation and phenotypic variation through various in silico approaches. METHODS The present study analyzed the functional consequences of the nsSNPs in human DBY gene using SIFT, PolyPhen 2, PROVEAN, SNAP2, PMut, nsSNPAnalyzer, PhD-SNP and SNPs&GO along with stability analysis through I-Mutant2.0, MuPro and iPTREE-STAB. The conservational analysis of amino acid residues, biophysical properties and conserved domains of the DBY protein was analyzed using various computational tools. The 3D structure of the protein was generated using SPARKS-X and validated using RAMPAGE. RESULTS Out of 1130 SNPs reported in dbSNP, only one nsSNP (G300D) was found to have a functional effect on stability as well as the function of the DBY protein. The results showed the presence of G300 in the putative structure of DBY domain. CONCLUSION To the best of our knowledge, this is the first study to detect pathologically significant nsSNPs (G300D) through a computational approach in the DBY which can be useful for development in potent drug discovery studies.
Collapse
Affiliation(s)
- Mili Nailwal
- P.G. Department of Genetics, Ashok and Rita Patel Institute of Integrated Study and Research in Biotechnology and Allied Sciences (ARIBAS), New Vallabh Vidyanagar, Dist-Anand, Gujarat, 388121, India
| | - Jenabhai B Chauhan
- P.G. Department of Genetics, Ashok and Rita Patel Institute of Integrated Study and Research in Biotechnology and Allied Sciences (ARIBAS), New Vallabh Vidyanagar, Dist-Anand, Gujarat, 388121, India.
| |
Collapse
|
41
|
Colaco S, Modi D. Genetics of the human Y chromosome and its association with male infertility. Reprod Biol Endocrinol 2018; 16:14. [PMID: 29454353 PMCID: PMC5816366 DOI: 10.1186/s12958-018-0330-5] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/06/2018] [Indexed: 12/12/2022] Open
Abstract
The human Y chromosome harbors genes that are responsible for testis development and also for initiation and maintenance of spermatogenesis in adulthood. The long arm of the Y chromosome (Yq) contains many ampliconic and palindromic sequences making it predisposed to self-recombination during spermatogenesis and hence susceptible to intra-chromosomal deletions. Such deletions lead to copy number variation in genes of the Y chromosome resulting in male infertility. Three common Yq deletions that recur in infertile males are termed as AZF (Azoospermia Factor) microdeletions viz. AZFa, AZFb and AZFc. As estimated from data of nearly 40,000 Y chromosomes, the global prevalence of Yq microdeletions is 7.5% in infertile males; however the European infertile men are less susceptible to Yq microdeletions, the highest prevalence is in Americans and East Asian infertile men. In addition, partial deletions of the AZFc locus have been associated with infertility but the effect seems to be ethnicity dependent. Analysis of > 17,000 Y chromosomes from fertile and infertile men has revealed an association of gr/gr deletion with male infertility in Caucasians and Mongolian men, while the b2/b3 deletion is associated with male infertility in African and Dravidian men. Clinically, the screening for Yq microdeletions would aid the clinician in determining the cause of male infertility and decide a rational management strategy for the patient. As these deletions are transmitted to 100% of male offspring born through assisted reproduction, testing of Yq deletions will allow the couples to make an informed choice regarding the perpetuation of male infertility in future generations. With the emerging data on association of Yq deletions with testicular cancers and neuropsychiatric conditions long term follow-up data is urgently needed for infertile men harboring Yq deletions. If found so, the information will change the current the perspective of androgenetics from infertility and might have broad implication in men health.
Collapse
Affiliation(s)
- Stacy Colaco
- Department of Molecular and Cellular Biology, ICMR-National Institute for Research in Reproductive Health, JM Street, Parel, Mumbai, Maharashtra, 400012, India
| | - Deepak Modi
- Department of Molecular and Cellular Biology, ICMR-National Institute for Research in Reproductive Health, JM Street, Parel, Mumbai, Maharashtra, 400012, India.
| |
Collapse
|
42
|
Ruthig VA, Nielsen T, Riel JM, Yamauchi Y, Ortega EA, Salvador Q, Ward MA. Testicular abnormalities in mice with Y chromosome deficiencies. Biol Reprod 2017; 96:694-706. [PMID: 28339606 DOI: 10.1095/biolreprod.116.144006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 01/10/2017] [Indexed: 11/01/2022] Open
Abstract
We recently investigated mice with Y chromosome gene contribution limited to two, one, or no Y chromosome genes in respect to their ability to produce haploid round spermatids and live offspring following round spermatid injection. Here we explored the normalcy of germ cells and Sertoli cells within seminiferous tubules, and the interstitial tissue of the testis in these mice. We performed quantitative analysis of spermatogenesis and interstitial tissue on Periodic acid-Schiff and hematoxylin-stained mouse testis sections. The seminiferous epithelium of mice with limited Y gene contribution contained various cellular abnormalities, the total number of which was higher than in the males with an intact Y chromosome. The distribution of specific abnormality types varied among tested genotypes. The males with limited Y genes also had an increased population of testicular macrophages and internal vasculature structures. The data indicate that Y chromosome gene deficiencies in mice are associated with cellular abnormalities of the seminiferous epithelium and some changes within the testicular interstitium.
Collapse
Affiliation(s)
- Victor A Ruthig
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Torbjoern Nielsen
- Center for Advanced Research in Sleep Medicine, CIUSSS-NÎM - Hôpital du Sacré-Coeur de Montréal, Montréal, Que., Canada.,Department of Psychiatry, Université de Montréal, Montréal, Que., Canada H3T 1J4
| | - Jonathan M Riel
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Yasuhiro Yamauchi
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Egle A Ortega
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | | | - Monika A Ward
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| |
Collapse
|
43
|
Meyfour A, Pooyan P, Pahlavan S, Rezaei-Tavirani M, Gourabi H, Baharvand H, Salekdeh GH. Chromosome-Centric Human Proteome Project Allies with Developmental Biology: A Case Study of the Role of Y Chromosome Genes in Organ Development. J Proteome Res 2017; 16:4259-4272. [PMID: 28914051 DOI: 10.1021/acs.jproteome.7b00446] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
One of the main goals of Chromosome-Centric Human Proteome Project is to identify protein evidence for missing proteins (MPs). Here, we present a case study of the role of Y chromosome genes in organ development and how to overcome the challenges facing MPs identification by employing human pluripotent stem cell differentiation into cells of different organs yielding unprecedented biological insight into adult silenced proteins. Y chromosome is a male-specific sex chromosome which escapes meiotic recombination. From an evolutionary perspective, Y chromosome has preserved 3% of ancestral genes compared to 98% preservation of the X chromosome based on Ohno's law. Male specific region of Y chromosome (MSY) contains genes that contribute to central dogma and govern the expression of various targets throughout the genome. One of the most well-known functions of MSY genes is to decide the male-specific characteristics including sex, testis formation, and spermatogenesis, which are majorly formed by ampliconic gene families. Beyond its role in sex-specific gonad development, MSY genes in coexpression with their X counterparts, as single copy and broadly expressed genes, inhibit haplolethality and play a key role in embryogenesis. The role of X-Y related gene mutations in the development of hereditary syndromes suggests an essential contribution of sex chromosome genes to development. MSY genes, solely and independent of their X counterparts and/or in association with sex hormones, have a considerable impact on organ development. In this Review, we present major recent findings on the contribution of MSY genes to gonad formation, spermatogenesis, and the brain, heart, and kidney development and discuss how Y chromosome proteome project may exploit developmental biology to find missing proteins.
Collapse
Affiliation(s)
- Anna Meyfour
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research , 81589-68433 Tehran, Iran.,Proteomics Research Center, Department of Basic Science, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences , 19839-63113 Tehran, Iran
| | - Paria Pooyan
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research , 81589-68433 Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research , 81589-68433 Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Department of Basic Science, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences , 19839-63113 Tehran, Iran
| | - Hamid Gourabi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute , 19395-4644 Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research , 81589-68433 Tehran, Iran.,Department of Developmental Biology, University of Science and Culture , 19395-4644 Tehran, Iran
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research , 81589-68433 Tehran, Iran.,Department of Systems Biology, Agricultural Biotechnology Research Institute of Iran , 31535-1897 Karaj, Iran
| |
Collapse
|
44
|
Oud MS, Ramos L, O'Bryan MK, McLachlan RI, Okutman Ö, Viville S, Vries PF, Smeets DF, Lugtenberg D, Hehir‐Kwa JY, Gilissen C, de Vorst M, Vissers LE, Hoischen A, Meijerink AM, Fleischer K, Veltman JA, Noordam MJ. Validation and application of a novel integrated genetic screening method to a cohort of 1,112 men with idiopathic azoospermia or severe oligozoospermia. Hum Mutat 2017; 38:1592-1605. [DOI: 10.1002/humu.23312] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/04/2017] [Accepted: 08/08/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Manon S. Oud
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
| | - Liliana Ramos
- Department of Gynaecology and Obstetrics Radboudumc Nijmegen The Netherlands
| | - Moira K. O'Bryan
- The Development and Stem Cells Program of Monash Biomedicine Discovery Institute Monash University Clayton Australia
- Department of Anatomy and Developmental Biology Monash University Clayton Australia
| | | | - Özlem Okutman
- Laboratory of Genetic Diagnostics UF3472‐Genetics of Infertility University Hospital Strasbourg Strasbourg France
| | - Stephane Viville
- Laboratory of Genetic Diagnostics UF3472‐Genetics of Infertility University Hospital Strasbourg Strasbourg France
| | - Petra F. Vries
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
| | - Dominique F.C.M. Smeets
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
| | - Dorien Lugtenberg
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
| | - Jayne Y. Hehir‐Kwa
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
| | - Christian Gilissen
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
| | - Maartje de Vorst
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
| | - Lisenka E.L.M. Vissers
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
| | - Alexander Hoischen
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI) Radboudumc Nijmegen The Netherlands
| | - Aukje M. Meijerink
- Department of Gynaecology and Obstetrics Radboudumc Nijmegen The Netherlands
| | - Kathrin Fleischer
- Department of Gynaecology and Obstetrics Radboudumc Nijmegen The Netherlands
| | - Joris A. Veltman
- Department of Human Genetics Donders Institute for Brain Cognition and Behavior Radboudumc Nijmegen The Netherlands
- Institute of Genetic Medicine Newcastle University Newcastle upon Tyne United Kingdom
- Department of Genetics and Cell Biology Maastricht UMC+ Maastricht The Netherlands
| | - Michiel J. Noordam
- Department of Genetics and Cell Biology Maastricht UMC+ Maastricht The Netherlands
| |
Collapse
|
45
|
Abstract
The properties of the human Y chromosome - namely, male specificity, haploidy and escape from crossing over - make it an unusual component of the genome, and have led to its genetic variation becoming a key part of studies of human evolution, population history, genealogy, forensics and male medical genetics. Next-generation sequencing (NGS) technologies have driven recent progress in these areas. In particular, NGS has yielded direct estimates of mutation rates, and an unbiased and calibrated molecular phylogeny that has unprecedented detail. Moreover, the availability of direct-to-consumer NGS services is fuelling a rise of 'citizen scientists', whose interest in resequencing their own Y chromosomes is generating a wealth of new data.
Collapse
|
46
|
Miao N, Wang X, Feng Y, Gong Y. Male-biased miR-92 from early chicken embryonic gonads directly targets ATRX and DDX3X. Gene 2017; 626:326-336. [PMID: 28554548 DOI: 10.1016/j.gene.2017.05.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/11/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022]
Abstract
MiR-17-92 cluster consists of multifunctional miRNAs related to gonadal development in mammals. Our preliminary data showed that gga-miR-92 was male-biased in chicken embryonic gonads at E5.5 and E6.5. MiR-92(a-2) and two putative targets (ATRX and DDX3X) were highly conserved and located on mammalian Chromosome X but on autosomes in chicken. Here, we studied the expression and interaction of miR-92 and the targets (ATRX and DDX3X) in chicken embryonic gonads. What's more, male-biased miR-92 shows an opposite expression tendency with ATRX and DDX3X in eight embryonic stages and different tissues at E10.5 by qRT-PCR. To verify the regulation relationship between miR-92 and two targets, we performed dual-luciferase reporter assay in DF1, overexpression and inhibition of miR-92 in chicken embryonic fibroblasts (CEFs). The results show that miR-92 directly targets ATRX and DDX3X by binding the 3' un-translated region (3'-UTR), and the over-expression and inhibition of miR-92 negatively regulates ATRX and DDX3X. After the identification of the expression of their downstream genes (AMH and WNT4) in mRNA level, we found that there is no regulatory relationship between ATRX and DDX3X. The overall results indicate that miR-92 may perform roles in early chicken gonadogenesis by regulating the expressions of ATRX and DDX3X, respectively.
Collapse
Affiliation(s)
- Nan Miao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; Institute of Genomics, College of Biomedical, Huaqiao University, 668 Jimei Road, Xiamen 361021, People's Republic of China
| | - Xin Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Yanping Feng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| | - Yanzhang Gong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| |
Collapse
|
47
|
Spermatogenic failure and the Y chromosome. Hum Genet 2017; 136:637-655. [PMID: 28456834 DOI: 10.1007/s00439-017-1793-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/30/2017] [Indexed: 12/29/2022]
Abstract
The Y chromosome harbors a number of genes essential for testis development and function. Its highly repetitive structure predisposes this chromosome to deletion/duplication events and is responsible for Y-linked copy-number variations (CNVs) with clinical relevance. The AZF deletions remove genes with predicted spermatogenic function en block and are the most frequent known molecular causes of impaired spermatogenesis (5-10% of azoospermic and 2-5% of severe oligozoospermic men). Testing for this deletion has both diagnostic and prognostic value for testicular sperm retrieval in azoospermic men. The most dynamic region on the Yq is the AZFc region, presenting numerous NAHR hotspots leading to partial losses or gains of the AZFc genes. The gr/gr deletion (a partial AZFc deletion) negatively affects spermatogenic efficiency and it is a validated, population-dependent risk factor for oligozoospermia. In certain populations, the Y background may play a role in the phenotypic expression of partial AZFc rearrangements and similarly it may affect the predisposition to specific deletions/duplication events. Also, the Yp contains a gene array, TSPY1, with potential effect on germ cell proliferation. Despite intensive investigations during the last 20 years on the role of this sex chromosome in spermatogenesis, a number of clinical and basic questions remain to be answered. This review is aimed at providing an overview of the role of Y chromosome-linked genes, CNVs, and Y background in spermatogenesis.
Collapse
|
48
|
Helsel AR, Oatley MJ, Oatley JM. Glycolysis-Optimized Conditions Enhance Maintenance of Regenerative Integrity in Mouse Spermatogonial Stem Cells during Long-Term Culture. Stem Cell Reports 2017; 8:1430-1441. [PMID: 28392219 PMCID: PMC5425612 DOI: 10.1016/j.stemcr.2017.03.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 02/08/2023] Open
Abstract
The application of spermatogonial stem cell (SSC) transplantation for regenerating male fertility requires amplification of SSC number in vitro during which the integrity to re-establish spermatogenesis must be preserved. Conventional conditions supporting proliferation of SSCs from mouse pups have been the basis for developing methodology with adult human cells but are unrefined. We found that the integrity to regenerate spermatogenesis after transplantation declines with advancing time in primary cultures of pup SSCs and that the efficacy of deriving cultures from adult SSCs is limited with conventional conditions. To address these deficiencies, we optimized the culture environment to favor glycolysis as the primary bioenergetics process. In these conditions, regenerative integrity of pup and adult SSCs was significantly improved and the efficiency of establishing primary cultures was 100%. Collectively, these findings suggest that SSCs are primed for conditions favoring glycolytic activity, and matching culture environments to their bioenergetics is critical for maintaining functional integrity. Regenerative integrity of SSCs declines over time in conventional culture Glycolysis-optimized (GO) culture improves regenerative integrity of SSCs GO conditions enhance the long-term culture of SSCs from adult mice
Collapse
Affiliation(s)
- Aileen R Helsel
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Melissa J Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Jon M Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
49
|
Kotov AA, Olenkina OM, Godneeva BK, Adashev VE, Olenina LV. Progress in understanding the molecular functions of DDX3Y (DBY) in male germ cell development and maintenance. Biosci Trends 2017; 11:46-53. [PMID: 28190795 DOI: 10.5582/bst.2016.01216] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Human DDX3 paralogs are housed on the X chromosome (DDX3X) as well as in the non- recombining region Yq11 of the Y-chromosome (DDX3Y or DBY). A gene encoding RNA helicase DDX3Y is located in the AZoospermia Factor a (AZFa) region of the Y-chromosome and expressed only in male germ cells. Deletions encompassing the DDX3Y gene lead to azoospermia and cause Sertoli Cell-Only Syndrome (SCOS) in humans. SCOS is characterized by a complete germ cell lack with preservation of somatic Sertoli cells. This review summarizes current advances in the study of DDX3Y functions in maintenance and development of early male germ cells. Data obtained from a mouse xenotransplantation model reveals that DDX3Y expression is enough to drive germ cell differentiation of AZFa-deleted human induced pluripotent stem cells (iPSCs) and for activation of the specific set of germline developmental genes. Results achieved using the testes of Drosophila demonstrate that DDX3Y homolog Belle is required cell-autonomously for mitotic progression and survival of germline stem cells and spermatogonia as the upstream regulator of mitotic cyclin expression.
Collapse
Affiliation(s)
- Alexei A Kotov
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences
| | | | | | | | | |
Collapse
|
50
|
|