1
|
Li M, Zhang X, Yan J, Shu H, Li Z, Ye C, Chen L, Feng C, Zheng Y. Non-invasive ultrasound localization microscopy (ULM) in azoospermia: connecting testicular microcirculation to spermatogenic functions. Theranostics 2024; 14:4967-4982. [PMID: 39267788 PMCID: PMC11388075 DOI: 10.7150/thno.99668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Rationale: Azoospermia is a significant reproductive challenge. Differentiating between non-obstructive azoospermia (NOA) and obstructive azoospermia (OA) is crucial as each type requires distinct management strategies. Testicular microcirculation plays a profound role in spermatogenic functions. However, current diagnostic methods are limited in their ability to effectively elucidate this crucial connection. Methods: We employed ultrasound localization microscopy (ULM) to visualize testicular microcirculation in NOA and OA patients and quantified the testicular hemodynamic parameters. Pearson correlation analysis was conducted to investigate the inner connection between parameters of testicular microcirculation and clinical spermatogenic functions. We conducted multiple logistic regression analysis to establish a new diagnostic model that integrates follicle-stimulating hormone (FSH) and mean vascular diameter to distinguish NOA from OA. Results: Our findings demonstrated significant differences in vascular parameters between NOA and OA, with NOA characterized by lower mean vascular diameter (p < 0.001), vessel density (p < 0.001), and fractal number (p < 0.001). Testicular volume showed a moderate positive correlation with mean vascular diameter (r = 0.419, p < 0.01) and vessel density (r = 0.415, p < 0.01); Mean vascular diameter exhibited negative correlations with both FSH (r = -0.214, p < 0.05) and age (r = -0.240, p < 0.05); FSH (r = -0.202, p < 0.05) and luteinizing hormone (LH) (r = -0.235, p < 0.05) were negatively correlated with mean blood flow velocity. The diagnostic model demonstrated an area under the curve (AUC) of 0.968. We also reported a method to map the vascular pressure distribution derived from the blood flow velocity generated by ULM. Conclusions: ULM provides a non-invasive and detailed assessment of testicular microvascular dynamics. The ULM-derived vascular parameters are able to connect testicular microcirculation to spermatogenic functions. The combination of FSH and mean vascular diameter enhances diagnostic precision and holds potential for distinguishing NOA from OA.
Collapse
Affiliation(s)
- Maoyao Li
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yishan Road 600, Shanghai, 200233, China
| | - Xingxuan Zhang
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yishan Road 600, Shanghai, 200233, China
| | - Jipeng Yan
- Department of Bioengineering, Imperial College London, Exhibition Road, London, SW7 2AZ, U.K
| | - Huiquan Shu
- Department of Reproductive Medicine, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China
| | - Zitong Li
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yishan Road 600, Shanghai, 200233, China
| | - Chujun Ye
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yishan Road 600, Shanghai, 200233, China
| | - Lei Chen
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yishan Road 600, Shanghai, 200233, China
| | - Chao Feng
- Department of Reproductive Medicine, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China
| | - Yuanyi Zheng
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yishan Road 600, Shanghai, 200233, China
| |
Collapse
|
2
|
Kavoussi PK, Gherabi N, Saleh R. Clinical predictors of successful outcomes for couples with nonobstructive azoospermic male partners undergoing micro-TESE. Asian J Androl 2024:00129336-990000000-00209. [PMID: 39040009 DOI: 10.4103/aja202436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/09/2024] [Indexed: 07/24/2024] Open
Abstract
ABSTRACT Nonobstructive azoospermia (NOA) is the most challenging and complex clinical scenario for infertile men. Besides circumstances such as hypogonadotropic hypogonadism, surgical sperm retrieval is typically necessary, and microdissection testicular sperm extraction (micro-TESE) is the procedure of choice for men with NOA desiring to father children with their own gametes. Micro-TESE results in the highest numbers of sperm cells retrieved for use with in vitro fertilization/intracytoplasmic sperm injection (ICSI) in comparison to all other techniques for surgical sperm retrieval in men with NOA. Several factors may affect sperm retrieval rate and ICSI outcomes, including the patient's age, testicular volume, histopathological and genetic profile, and serum hormone levels. This article aims to review the medical literature describing predictors of successful micro-TESE and the outcomes of ICSI in men with NOA.
Collapse
Affiliation(s)
- Parviz K Kavoussi
- Department of Reproductive Urology, Austin Fertility and Reproductive Medicine/Westlake IVF, Austin, TX 78746, USA
| | - Nazim Gherabi
- Department of Medicine, Algiers 1 University, Algiers 16000, Algeria
| | - Ramadan Saleh
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag 83534, Egypt
- Ajyal IVF Center, Ajyal Hospital, Sohag 83534, Egypt
| |
Collapse
|
3
|
Japari A, Moorthy D, Rambhatla A. Andrology laboratory technique for analysis of semen in men with azoospermia. Asian J Androl 2024:00129336-990000000-00189. [PMID: 38759095 DOI: 10.4103/aja202429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/25/2024] [Indexed: 05/19/2024] Open
Abstract
ABSTRACT Discovery of spermatozoa during the 17th century led to developing technologies for semen analysis in the early 1900s, and then, standard techniques were implemented during the 20th century. Semen analysis has a pivotal role in the male infertility evaluation, and azoospermia is an important finding. Azoospermia is identified in 15% of infertile men. However, the accurate laboratory assessment of azoospermia poses certain technical challenges. Laboratories currently perform semen assessment with great variability; thus, a standard method should be used. Planning suitable management and determining the cause of infertility require a precise evaluation of azoospermia. This review aims to address the definition of azoospermia and highlight laboratory methods in the assessments of azoospermia. Basic methods such as centrifugation, repeat pellet analysis, and staining and advanced methods such as genetic testing and biomarkers have been discussed. These methods have helped in standardizing the protocol for accurate azoospermia assessments with less variability.
Collapse
Affiliation(s)
- Andrian Japari
- Fertility Clinic, Telogorejo Hospital, Semarang 50241, Indonesia
- Global Andrology Forum, Moreland Hills, OH 44022, USA
| | - Dharani Moorthy
- Global Andrology Forum, Moreland Hills, OH 44022, USA
- Swarupa Fertility and IVF Centre, Vijayawada, Andhra Pradesh 520002, India
| | - Amarnath Rambhatla
- Global Andrology Forum, Moreland Hills, OH 44022, USA
- Department of Urology, Vattikuti Urology Institute, Henry Ford Health System, Detroit, MI 48075, USA
| |
Collapse
|
4
|
Fietz D, Sgaier R, O’Donnell L, Stanton PG, Dagley LF, Webb AI, Schuppe HC, Diemer T, Pilatz A. Proteomic biomarkers in seminal plasma as predictors of reproductive potential in azoospermic men. Front Endocrinol (Lausanne) 2024; 15:1327800. [PMID: 38654926 PMCID: PMC11035875 DOI: 10.3389/fendo.2024.1327800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Azoospermia, characterized by an absence of sperm in the ejaculate, represents the most severe form of male infertility. While surgical sperm retrieval in obstructive azoospermia (OA) is successful in the majority of cases, patients with non-obstructive azoospermia (NOA) show retrieval rates of only about 50% and thus frequently have unnecessary surgery. Surgical intervention could be avoided if patients without preserved spermatogenesis are identified preoperatively. This prospective study aimed to discover biomarkers in seminal plasma that could be employed for a non-invasive differential diagnosis of OA/NOA in order to rationalize surgery recommendations and improve success rates. Methods All patients signed written informed consent, underwent comprehensive andrological evaluation, received human genetics to exclude relevant pathologies, and patients with azoospermia underwent surgical sperm retrieval. Using label-free LC-MS/MS, we compared the proteomes of seminal plasma samples from fertile men (healthy controls (HC), n=8) and infertile men diagnosed with 1) OA (n=7), 2) NOA with successful sperm retrieval (mixed testicular atrophy (MTA), n=8), and 3) NOA without sperm retrieval (Sertoli cell-only phenotype (SCO), n=7). Relative abundance changes of two candidate markers of sperm retrieval, HSPA2 and LDHC, were confirmed by Western Blot. Results We found the protein expression levels of 42 proteins to be significantly down-regulated (p ≤ 0.05) in seminal plasma from SCO NOA patients relative to HC whereas only one protein was down-regulated in seminal plasma from MTA patients. Analysis of tissue and cell expression suggested that the testis-specific proteins LDHC, PGK2, DPEP3, and germ-cell enriched heat-shock proteins HSPA2 and HSPA4L are promising biomarkers of spermatogenic function. Western blotting revealed a significantly lower abundance of LDHC and HSPA2 in the seminal plasma of men with NOA (SCO and MTA) compared to controls. Discussion The results indicate that certain testis-specific proteins when measured in seminal plasma, could serve as indicators of the presence of sperm in the testis and predict the success of sperm retrieval. Used in conjunction with conventional clinical assessments, these proteomic biomarkers may assist in the non-invasive diagnosis of idiopathic male infertility.
Collapse
Affiliation(s)
- Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Raouda Sgaier
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| | - Liza O’Donnell
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Peter G. Stanton
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Laura F. Dagley
- Advanced Technology and Biology Division, Walter and Eliza Hall Institute, Parkville, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Andrew I. Webb
- Advanced Technology and Biology Division, Walter and Eliza Hall Institute, Parkville, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Hans-Christian Schuppe
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| | - Thorsten Diemer
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| | - Adrian Pilatz
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
5
|
Khanmohammadi N, Malek F, Takzaree N, Malekzadeh M, Khanehzad M, Akanji OD, Rastegar T. Sertoli Cell-Conditioned Medium Induces Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells to Male Germ-Like Cells in Busulfan-Induced Azoospermic Mouse Model. Reprod Sci 2024; 31:375-392. [PMID: 37737972 DOI: 10.1007/s43032-023-01332-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023]
Abstract
Non-obstructive azoospermia is a severe form of male infertility, with limited effective treatments. Bone marrow mesenchymal stem cells (BMSCs) can differentiate to different cell lines; therefore, transplantation of these cells is used for treatment of several diseases. Since these cells require induction factors to differentiate into germ cells, we co-transplanted bone marrow stem cells (BMSCs) with Sertoli cell-conditioned medium (SCCM) into the testis of azoospermic mice. This study was carried out in two sections, in vitro and in vivo. For in vitro study, differentiating factors (c-kit and ID4) were examined after 15 days of co-culture of bone marrow cells with Sertoli cell-conditioned medium, while for in vivo study, the azoospermia model was first created by intraperitoneal administration of a single-dose busulfan (40 mg/kg) followed by single-dose CdCl2 (2 mg/kg) after 4 weeks. Mice were divided into 4 groups including control (azoospermia), BMSC, SCCM, and BMSC + SCCM. Eight weeks after transplantation, samples were assessed for proliferation and differentiation via the expression level of MVH, ID4, SCP3, Tp1, Tp2, and Prm1 differentiation markers. The results showed that BMSC co-cultured with SCCM in vitro differentiated BMSC to germ-like cells. Similarly, in vivo studies revealed a higher level of BMSC differentiation into germ-like cells with significant higher expression of differentiation markers in transplanted groups compared to the control. This study confirmed the role of SCCM as an inductive factor for BMSC differentiation to germ cells both in vivo and in vitro conditions.
Collapse
Affiliation(s)
- Nasrin Khanmohammadi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Malek
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Takzaree
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrnoush Malekzadeh
- Department of Anatomy, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Khanehzad
- Department of Anatomy, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Tayebeh Rastegar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Romano M, Cirillo F, Ravaioli N, Morenghi E, Negri L, Ozgur B, Albani E, Levi-Setti PE. Reproductive and obstetric outcomes in TESE-ICSI cycles: A comparison between obstructive and non-obstructive azoospermia. Andrology 2023. [PMID: 38108554 DOI: 10.1111/andr.13568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE Comparison of intracytoplasmic sperm injection cycles with testicular sperm extraction in obstructive azoospermia and non-obstructive azoospermia are limited, and few studies have addressed obstetric and neonatal outcomes. DESIGN This study analyzed couples who underwent testicular sperm extraction-intracytoplasmic sperm injection cycles for obstructive azoospermia and non-obstructive azoospermia to determine whether impaired spermatogenesis in non-obstructive azoospermia patients would lead to worse reproductive outcomes and higher rates of pregnancy complications and fetal anomalies. This study is a retrospective, single-center analysis of all testicular sperm cycles performed between January 1, 2001 and December 31, 2020. RESULTS A total of 392 couples were considered in the study, leading to 1066 induction cycles, 620 (58.2%) from patients with obstructive azoospermia and 446 (41.8%) from non-obstructive azoospermia. The cumulative delivery rate did not significantly differ between the two groups (34% vs. 31%; p = 0.326). The miscarriage rate was similar between obstructive azoospermia and non-obstructive azoospermia patients. Fertilization rate instead showed a statistically significant difference (obstructive azoospermia: 66.1 ± 25.7 vs. non-obstructive azoospermia: 56.1 ± 27.0; p < 0.001). The overall maternal complication rate in the non-obstructive azoospermia group was higher (10.7% vs. 18.4%; p = 0.035), but there was no statistical significance for each pathology. There was no statistical difference in gestational age between the two groups for both single and twin pregnancies. Seven cases of congenital defects occurred in the obstructive azoospermia group, while two cases occurred in the non-obstructive azoospermia group. CONCLUSIONS Despite impaired spermatogenesis in non-obstructive azoospermia patients, there were no substantial differences in reproductive outcomes compared to patients with obstructive azoospermia, even in terms of obstetric safety and neonatal well-being.
Collapse
Affiliation(s)
- Massimo Romano
- Department of Gynecology, Division of Gynecology and Reproductive Medicine, Fertility Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Federico Cirillo
- Department of Gynecology, Division of Gynecology and Reproductive Medicine, Fertility Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Noemi Ravaioli
- Department of Gynecology and Obstetrics, Hospital of Lugo (RA), Lugo, Italy
| | - Emanuela Morenghi
- Biostatistics Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luciano Negri
- Department of Gynecology, Division of Gynecology and Reproductive Medicine, Fertility Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Bulbul Ozgur
- Department of Gynecology, Division of Gynecology and Reproductive Medicine, Fertility Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Elena Albani
- Department of Gynecology, Division of Gynecology and Reproductive Medicine, Fertility Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Paolo Emanuele Levi-Setti
- Department of Gynecology, Division of Gynecology and Reproductive Medicine, Fertility Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| |
Collapse
|
7
|
Meng Q, Shao B, Zhao D, Fu X, Wang J, Li H, Zhou Q, Gao T. Loss of SUN1 function in spermatocytes disrupts the attachment of telomeres to the nuclear envelope and contributes to non-obstructive azoospermia in humans. Hum Genet 2023; 142:531-541. [PMID: 36933034 DOI: 10.1007/s00439-022-02515-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/13/2022] [Indexed: 03/19/2023]
Abstract
One of the most severe forms of infertility in humans, caused by gametogenic failure, is non-obstructive azoospermia (NOA). Approximately, 20-30% of men with NOA may have single-gene mutations or other genetic variables that cause this disease. While a range of single-gene mutations associated with infertility has been identified in prior whole-exome sequencing (WES) studies, current insight into the precise genetic etiology of impaired human gametogenesis remains limited. In this paper, we described a proband with NOA who experienced hereditary infertility. WES analyses identified a homozygous variant in the SUN1 (Sad1 and UNC84 domain containing 1) gene [c. 663C > A: p.Tyr221X] that segregated with infertility. SUN1 encodes a LINC complex component essential for telomeric attachment and chromosomal movement. Spermatocytes with the observed mutations were incapable of repairing double-strand DNA breaks or undergoing meiosis. This loss of SUN1 functionality contributes to significant reductions in KASH5 levels within impaired chromosomal telomere attachment to the inner nuclear membrane. Overall, our results identify a potential genetic driver of NOA pathogenesis and provide fresh insight into the role of the SUN1 protein as a regulator of prophase I progression in the context of human meiosis.
Collapse
Affiliation(s)
- Qingxia Meng
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Gusu School, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, China
| | - Binbin Shao
- Department of Reproduction, The Affiliated Obstetrics and Gynecology Hospital With, Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Dan Zhao
- Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xu Fu
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Gusu School, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, China
| | - Jiaxiong Wang
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Gusu School, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, China
| | - Hong Li
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Gusu School, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, China.
| | - Qiao Zhou
- Department of Reproduction, The Affiliated Obstetrics and Gynecology Hospital With, Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China.
| | - Tingting Gao
- Changzhou Medical Center, Changzhou Maternal and Child Health Care Hospital, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
8
|
Balagannavar G, Basavaraju K, Bajpai AK, Davuluri S, Kannan S, S Srini V, S Chandrashekar D, Chitturi N, K Acharya K. Transcriptomic analysis of the Non-Obstructive Azoospermia (NOA) to address gene expression regulation in human testis. Syst Biol Reprod Med 2023; 69:196-214. [PMID: 36883778 DOI: 10.1080/19396368.2023.2176268] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
There is a need to understand the molecular basis of testes under Non-Obstructive Azoospermia (NOA), a state of failed spermatogenesis. There has been a lack of attention to the transcriptome at the level of alternatively spliced mRNAs (iso-mRNAs) and the mechanism of gene expression regulation. Hence, we aimed to establish a reliable iso-mRNA profile of NOA-testes, and explore molecular mechanisms - especially those related to gene expression regulation. We sequenced mRNAs from testicular samples of donors with complete spermatogenesis (control samples) and a failure of spermatogenesis (NOA samples). We identified differentially expressed genes and their iso-mRNAs via standard NGS data analyses. We then listed these iso-mRNAs hierarchically based on the extent of consistency of differential quantities across samples and groups, and validated the lists via RT-qPCRs (for 80 iso-mRNAs). In addition, we performed extensive bioinformatic analysis of the splicing features, domains, interactions, and functions of differentially expressed genes and iso-mRNAs. Many top-ranking down-regulated genes and iso-mRNAs, i.e., those down-regulated more consistently across the NOA samples, are associated with mitosis, replication, meiosis, cilium, RNA regulation, and post-translational modifications such as ubiquitination and phosphorylation. Most down-regulated iso-mRNAs correspond to full-length proteins that include all expected domains. The predominance of alternative promoters and termination sites in these iso-mRNAs indicate their gene expression regulation via promoters and UTRs. We compiled a new, comprehensive list of human transcription factors (TFs) and used it to identify TF-'TF gene' interactions with potential significance in down-regulating genes under the NOA condition. The results indicate that RAD51 suppression by HSF4 prevents SP1-activation, and SP1, in turn, could regulate multiple TF genes. This potential regulatory axis and other TF interactions identified in this study could explain the down-regulation of multiple genes in NOA-testes. Such molecular interactions may also have key regulatory roles during normal human spermatogenesis.
Collapse
Affiliation(s)
- Govindkumar Balagannavar
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India.,Research Scholar, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Kavyashree Basavaraju
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India.,BdataA: Biological data Analyzers' Association (virtual organization http://startbioinfo.com/BdataA/), India
| | - Akhilesh Kumar Bajpai
- BdataA: Biological data Analyzers' Association (virtual organization http://startbioinfo.com/BdataA/), India
| | - Sravanthi Davuluri
- BdataA: Biological data Analyzers' Association (virtual organization http://startbioinfo.com/BdataA/), India
| | - Shruthi Kannan
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India
| | - Vasan S Srini
- Manipal Fertility, Manipal Hospital, Bengaluru, Karnataka, India
| | | | - Neelima Chitturi
- BdataA: Biological data Analyzers' Association (virtual organization http://startbioinfo.com/BdataA/), India
| | - Kshitish K Acharya
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India.,BdataA: Biological data Analyzers' Association (virtual organization http://startbioinfo.com/BdataA/), India
| |
Collapse
|
9
|
Tanaka A, Watanabe S. How to improve the clinical outcome of round spermatid injection (ROSI) into the oocyte: Correction of epigenetic abnormalities. Reprod Med Biol 2023; 22:e12503. [PMID: 36789269 PMCID: PMC9909386 DOI: 10.1002/rmb2.12503] [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: 09/09/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 02/11/2023] Open
Abstract
Background First successful human round spermatid injection (ROSI) was conducted by Tesarik et al. in 1996 for the sole treatment of nonobstructive azoospermic men whose most advanced spermatogenic cells were elongating round spermatids. Nine offsprings from ROSI were reported between 1996 and 2000. No successful deliveries were reported for 15 years after that. Tanaka et al. reported 90 babies born after ROSI and their follow-up studies in 2015 and 2018 showed no significant differences in comparison with those born after natural conception in terms of physical and cognitive abilities. However, clinical outcomes remain low. Method Clinical and laboratory data of successful cases in the precursor ROSI groups and those of Tanaka et al. were reviewed. Results Differences were found between the two groups in terms of identification of characteristics of round spermatid and oocyte activation. Additionally, epigenetic abnormalities were identified as underlying causes for poor ROSI results, besides correct identification of round spermatid and adequate oocyte activation. Correction of epigenetic errors could lead to optimal embryonic development. Conclusion Correction of epigenetic abnormalities has a probability to improve the clinical outcome of ROSI.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Obstetrics and GynecologySaint Mother ClinicKitakyushuJapan
- Department of Obstetrics and GynecologyJuntendo University School of MedicineBunkyo‐kuJapan
| | - Seiji Watanabe
- Department of Anatomical ScienceHirosaki University Graduate School of MedicineAomoriJapan
| |
Collapse
|
10
|
Guzmán-Jiménez A, González-Muñoz S, Cerván-Martín M, Rivera-Egea R, Garrido N, Luján S, Santos-Ribeiro S, Castilla JA, Gonzalvo MC, Clavero A, Vicente FJ, Maldonado V, Villegas-Salmerón J, Burgos M, Jiménez R, Pinto MG, Pereira I, Nunes J, Sánchez-Curbelo J, López-Rodrigo O, Pereira-Caetano I, Marques PI, Carvalho F, Barros A, Bassas L, Seixas S, Gonçalves J, Lopes AM, Larriba S, Palomino-Morales RJ, Carmona FD, Bossini-Castillo L. Contribution of TEX15 genetic variants to the risk of developing severe non-obstructive oligozoospermia. Front Cell Dev Biol 2022; 10:1089782. [PMID: 36589743 PMCID: PMC9797780 DOI: 10.3389/fcell.2022.1089782] [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: 11/04/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Severe spermatogenic failure (SPGF) represents one of the most relevant causes of male infertility. This pathological condition can lead to extreme abnormalities in the seminal sperm count, such as severe oligozoospermia (SO) or non-obstructive azoospermia (NOA). Most cases of SPGF have an unknown aetiology, and it is known that this idiopathic form of male infertility represents a complex condition. In this study, we aimed to evaluate whether common genetic variation in TEX15, which encodes a key player in spermatogenesis, is involved in the susceptibility to idiopathic SPGF. Materials and Methods: We designed a genetic association study comprising a total of 727 SPGF cases (including 527 NOA and 200 SO) and 1,058 unaffected men from the Iberian Peninsula. Following a tagging strategy, three tag single-nucleotide polymorphisms (SNPs) of TEX15 (rs1362912, rs323342, and rs323346) were selected for genotyping using TaqMan probes. Case-control association tests were then performed by logistic regression models. In silico analyses were also carried out to shed light into the putative functional implications of the studied variants. Results: A significant increase in TEX15-rs1362912 minor allele frequency (MAF) was observed in the group of SO patients (MAF = 0.0842) compared to either the control cohort (MAF = 0.0468, OR = 1.90, p = 7.47E-03) or the NOA group (MAF = 0.0472, OR = 1.83, p = 1.23E-02). The genotype distribution of the SO population was also different from those of both control (p = 1.14E-02) and NOA groups (p = 4.33-02). The analysis of functional annotations of the human genome suggested that the effect of the SO-associated TEX15 variants is likely exerted by alteration of the binding affinity of crucial transcription factors for spermatogenesis. Conclusion: Our results suggest that common variation in TEX15 is involved in the genetic predisposition to SO, thus supporting the notion of idiopathic SPGF as a complex trait.
Collapse
Affiliation(s)
- Andrea Guzmán-Jiménez
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Sara González-Muñoz
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Miriam Cerván-Martín
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Rocío Rivera-Egea
- Andrology Laboratory and Sperm Bank, IVIRMA Valencia, Valencia, Spain,IVI Foundation, Health Research Institute La Fe, Valencia, Spain
| | - Nicolás Garrido
- IVI Foundation, Health Research Institute La Fe, Valencia, Spain,Servicio de Urología. Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Saturnino Luján
- Servicio de Urología. Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Samuel Santos-Ribeiro
- IVI-RMA Lisbon, Lisbon, Portugal,Department of Obstetrics and Gynecology, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - José A. Castilla
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain,Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de Las Nieves, Granada, Spain,CEIFER Biobanco—GAMETIA, Granada, Spain
| | - M. Carmen Gonzalvo
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain,Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de Las Nieves, Granada, Spain
| | - Ana Clavero
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain,Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de Las Nieves, Granada, Spain
| | - F. Javier Vicente
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain,UGC de Urología, HU Virgen de las Nieves, Granada, Spain
| | - Vicente Maldonado
- UGC de Obstetricia y Ginecología, Complejo Hospitalario de Jaén, Jaén, Spain
| | - Javier Villegas-Salmerón
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Miguel Burgos
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Rafael Jiménez
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Maria Graça Pinto
- Centro de Medicina Reprodutiva, Maternidade Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - Isabel Pereira
- Departamento de Obstetrícia, Ginecologia e Medicina da Reprodução, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisboa, Portugal
| | - Joaquim Nunes
- Departamento de Obstetrícia, Ginecologia e Medicina da Reprodução, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisboa, Portugal
| | - Josvany Sánchez-Curbelo
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, Barcelona, Spain
| | - Olga López-Rodrigo
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, Barcelona, Spain
| | - Iris Pereira-Caetano
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Patricia Isabel Marques
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Filipa Carvalho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Serviço de Genética, Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Alberto Barros
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Serviço de Genética, Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Lluís Bassas
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, Barcelona, Spain
| | - Susana Seixas
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - João Gonçalves
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,ToxOmics—Centro de Toxicogenómica e Saúde Humana, Nova Medical School, Lisbon, Portugal
| | - Alexandra M. Lopes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,CGPP-IBMC—Centro de Genética Preditiva e Preventiva, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sara Larriba
- Human Molecular Genetics Group, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Rogelio J. Palomino-Morales
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain,Departamento de Bioquímica y Biología Molecular I, Universidad de Granada, Granada, Spain
| | - F. David Carmona
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain,*Correspondence: F. David Carmona, ; Lara Bossini-Castillo,
| | - Lara Bossini-Castillo
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain,*Correspondence: F. David Carmona, ; Lara Bossini-Castillo,
| | | | | |
Collapse
|
11
|
Immune and spermatogenesis-related loci are involved in the development of extreme patterns of male infertility. Commun Biol 2022; 5:1220. [PMID: 36357561 PMCID: PMC9649734 DOI: 10.1038/s42003-022-04192-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/28/2022] [Indexed: 11/12/2022] Open
Abstract
We conducted a genome-wide association study in a large population of infertile men due to unexplained spermatogenic failure (SPGF). More than seven million genetic variants were analysed in 1,274 SPGF cases and 1,951 unaffected controls from two independent European cohorts. Two genomic regions were associated with the most severe histological pattern of SPGF, defined by Sertoli cell-only (SCO) phenotype, namely the MHC class II gene HLA-DRB1 (rs1136759, P = 1.32E-08, OR = 1.80) and an upstream locus of VRK1 (rs115054029, P = 4.24E-08, OR = 3.14), which encodes a protein kinase involved in the regulation of spermatogenesis. The SCO-associated rs1136759 allele (G) determines a serine in the position 13 of the HLA-DRβ1 molecule located in the antigen-binding pocket. Overall, our data support the notion of unexplained SPGF as a complex trait influenced by common variation in the genome, with the SCO phenotype likely representing an immune-mediated condition. A GWAS in a large case-control cohort of European ancestry identifies two genomic regions, the MHC class II gene HLA-DRB1 and an upstream locus of VRK1, that are associated with the most severe phenotype of spermatogenic failure.
Collapse
|
12
|
Wu X, Yun D, Sang M, Liu J, Zhou L, Shi J, Wang L, Bu T, Li L, Huang Y, Lin D, Sun F, Cheng CY. Defects of microtubule cytoskeletal organization in NOA human testes. Reprod Biol Endocrinol 2022; 20:154. [PMID: 36329464 PMCID: PMC9632130 DOI: 10.1186/s12958-022-01026-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022] Open
Abstract
The importance of actin and microtubule (MT) cytoskeletons in testis function in rodents is known to some extent, but its role in the etiology of azoospermia in humans remains unexplored. Here, we examined if MT cytoskeleton was defective in NOA (non-obstructive azoospermia) testes versus normal human testes based on histopathological, immunofluorescence (IF), and scRNA-Seq transcriptome profiling. Testis biopsy samples from n = 6 normal men versus n = 3 Sertoli cell only (SCO) and n = 3 MA (meiotic arrest) of NOA patients were used for histopathological analysis. IF analysis was also used to examine MT organization across the seminiferous epithelium, investigating the likely involvement of microtubule-associated proteins (MAPs). scRNA-Seq transcriptome profiling datasets from testes of 3 SCO patients versus 3 normal men in public domain in Gene Expression Omnibus (GEO) Sample (GSM) with identifiers were analyzed to examine relevant genes that regulate MT dynamics. NOA testes of MA and SCO patients displayed notable defects in MT organization across the epithelium with extensive truncation, mis-alignments and appeared as collapsed structures near the base of the tubules. These changes are in contrast to MTs in testes of normal men. scRNA-Seq analyses revealed considerable loss of spermatogenesis capacity in SCO testes of NOA patients versus normal men. An array of genes that support MT dynamics displayed considerable changes in expression and in spatial distribution. In summary, defects in MT cytoskeleton were noted in testes of NOA (SCO) patients, possibly mediated by defective spatial expression and/or distribution of MAPs. These changes, in turn, may impede spermatogenesis in SCO testes of NOA patients.
Collapse
Affiliation(s)
- Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Damin Yun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Mengmeng Sang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Jianpeng Liu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Liwei Zhou
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Jie Shi
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Lingling Wang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Tiao Bu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Linxi Li
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - YingYing Huang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Dengfeng Lin
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China.
- Center for Biomedical Research, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, 1230 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
13
|
Li JP, Du CQ, Liu ZB, Zhang FB, Li LJ, Wu JG, Tian YH, Liang ZY, Chen C, Jin F. Development and validation predictive models of sperm retrieval for azoospermic men undergoing testicular sperm aspiration: a multicentre, retrospective, cohort study. J Assist Reprod Genet 2022; 39:1779-1787. [PMID: 35870097 PMCID: PMC9428087 DOI: 10.1007/s10815-022-02531-y] [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: 02/21/2022] [Accepted: 05/26/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Testicular sperm aspiration (TESA) is widely used to retrieve sperm from testis. Diagnostic testicular biopsy should not be routinely performed for azoospermia. Therefore, a good predictive model is needed before TESA. METHODS A total of 1972 azoospermia patients constituted the modelling set, and 260 azoospermia patients from two other centres constituted the validation set. An integrated predictive model was built using logistic regression. Receiver operating characteristic (ROC), calibration and decision curve analyses were performed to evaluate the performance of follicle-stimulating hormone (FSH), semen volume, testicular volume and the integrated model. RESULTS The FSH level was the best univariate predictor for successful sperm retrieval (SSR) and was better than semen volume and testicular volume alone (p<0.001, threshold 6.17 IU/L, modelling set area under receiver operating characteristic curve (AUC) 0.80, accuracy 0.79; validation set AUC 0.87, accuracy 0.78). The integrated predictive model had excellent accuracy for predicting SSR (modelling set: AUC 0.93, accuracy 0.89; validation set: AUC 0.96, accuracy: 0.89). Calibration curve analysis indicated that the integrated model calibration was good and better than that of FSH, semen volume and testicular volume alone. Decision curve analysis indicated with a threshold probability between 0.05 and 0.98, the integrated model added more benefit than treating either all or no patients. CONCLUSIONS The integrated model has excellent discrimination and good calibration. It can help azoospermic men make better decisions before TESA. It should be noted that TESA is not the first-line treatment for non-obstructive azoospermia because of a low sperm retrieval rate.
Collapse
Affiliation(s)
- Jing-Ping Li
- grid.13402.340000 0004 1759 700XDepartment of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China 310006
| | - Cong-Qi Du
- grid.13402.340000 0004 1759 700XReproductive Medicine Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Zu-Bo Liu
- grid.513202.7Reproductive Medicine Centre, Jinhua People’s Hospital, Jinhua, People’s Republic of China
| | - Feng-Bin Zhang
- grid.13402.340000 0004 1759 700XDepartment of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China 310006
| | - Le-Jun Li
- grid.13402.340000 0004 1759 700XDepartment of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China 310006
| | - Jing-Gen Wu
- grid.13402.340000 0004 1759 700XDepartment of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China 310006
| | - Yong-Hong Tian
- grid.13402.340000 0004 1759 700XDepartment of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China 310006
| | - Zhong-Yan Liang
- grid.13402.340000 0004 1759 700XDepartment of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China 310006
| | - Chong Chen
- grid.13402.340000 0004 1759 700XDepartment of Ultrasound, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Fan Jin
- grid.13402.340000 0004 1759 700XDepartment of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China 310006
| |
Collapse
|
14
|
Common Variation in the PIN1 Locus Increases the Genetic Risk to Suffer from Sertoli Cell-Only Syndrome. J Pers Med 2022; 12:jpm12060932. [PMID: 35743717 PMCID: PMC9225465 DOI: 10.3390/jpm12060932] [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: 04/20/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022] Open
Abstract
We aimed to analyze the role of the common genetic variants located in the PIN1 locus, a relevant prolyl isomerase required to control the proliferation of spermatogonial stem cells and the integrity of the blood–testis barrier, in the genetic risk of developing male infertility due to a severe spermatogenic failure (SPGF). Genotyping was performed using TaqMan genotyping assays for three PIN1 taggers (rs2287839, rs2233678 and rs62105751). The study cohort included 715 males diagnosed with SPGF and classified as suffering from non-obstructive azoospermia (NOA, n = 505) or severe oligospermia (SO, n = 210), and 1058 controls from the Iberian Peninsula. The allelic frequency differences between cases and controls were analyzed by the means of logistic regression models. A subtype specific genetic association with the subset of NOA patients classified as suffering from the Sertoli cell-only (SCO) syndrome was observed with the minor alleles showing strong risk effects for this subset (ORaddrs2287839 = 1.85 (1.17–2.93), ORaddrs2233678 = 1.62 (1.11–2.36), ORaddrs62105751 = 1.43 (1.06–1.93)). The causal variants were predicted to affect the binding of key transcription factors and to produce an altered PIN1 gene expression and isoform balance. In conclusion, common non-coding single-nucleotide polymorphisms located in PIN1 increase the genetic risk to develop SCO.
Collapse
|
15
|
Adamczewska D, Słowikowska-Hilczer J, Walczak-Jędrzejowska R. The Fate of Leydig Cells in Men with Spermatogenic Failure. Life (Basel) 2022; 12:570. [PMID: 35455061 PMCID: PMC9028943 DOI: 10.3390/life12040570] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/25/2022] [Accepted: 04/08/2022] [Indexed: 11/18/2022] Open
Abstract
The steroidogenic cells in the testicle, Leydig cells, located in the interstitial compartment, play a vital role in male reproductive tract development, maintenance of proper spermatogenesis, and overall male reproductive function. Therefore, their dysfunction can lead to all sorts of testicular pathologies. Spermatogenesis failure, manifested as azoospermia, is often associated with defective Leydig cell activity. Spermatogenic failure is the most severe form of male infertility, caused by disorders of the testicular parenchyma or testicular hormone imbalance. This review covers current progress in knowledge on Leydig cells origin, structure, and function, and focuses on recent advances in understanding how Leydig cells contribute to the impairment of spermatogenesis.
Collapse
Affiliation(s)
| | | | - Renata Walczak-Jędrzejowska
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, 92-213 Lodz, Poland; (D.A.); (J.S.-H.)
| |
Collapse
|
16
|
Pathogenic variations in Germ Cell Nuclear Acidic Peptidase (GCNA) are associated with human male infertility. Eur J Hum Genet 2021; 29:1781-1788. [PMID: 34413498 PMCID: PMC8632907 DOI: 10.1038/s41431-021-00946-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/29/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023] Open
Abstract
Infertility affects one in six couples, half of which are caused by a male factor. Male infertility can be caused by both, qualitative and quantitative defects, leading to Oligo- astheno-terato-zoospermia (OAT; impairment in ejaculate sperm cell concentration, motility and morphology). Azoospermia defined as complete absence of sperm cells in the ejaculation. While hundreds of genes are involved in spermatogenesis the genetic etiology of men's infertility remains incomplete.We identified a hemizygous stop gain pathogenic variation (PV) in the X-linked Germ Cell Nuclear Acidic Peptidase (GCNA), in an Azoospermic patient by exome sequencing. Assessment of the prevalence of pathogenic variations in this gene in infertile males by exome sequence data of 11 additional unrelated patients identified a probable hemizygous causative missense PV in GCNA in a severe OAT patient. Expression of GCNA in the patients' testes biopsies and the stage of spermatogonial developmental arrest were determined by immunofluorescence and immunohistochemistry. The Azoospermic patient presented spermatogenic maturation arrest with an almost complete absence of early and late primary spermatocytes and thus the complete absence of sperm. GCNA is critical for genome integrity and its loss results in genomic instability and infertility in Drosophila, C. elegans, zebrafish, and mouse. PVs in GCNA appear to be incompatible with male fertility in humans as well: A stop-gain PV caused Azoospermia and a missense PV caused severe OAT with very low fertilization rates and no pregnancy in numerous IVF treatments.
Collapse
|
17
|
Kocamanoglu F, Ayas B, Bolat MS, Abur U, Bolat R, Asci R. Endocrine, sexual and reproductive functions in patients with Klinefelter syndrome compared to non-obstructive azoospermic patients. Int J Clin Pract 2021; 75:e14294. [PMID: 33928735 DOI: 10.1111/ijcp.14294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/27/2021] [Indexed: 11/27/2022] Open
Abstract
AIMS We aimed to investigate fertilisation rates, quality of embryo, pregnancy and live birth rates, endocrine, sexual function, psychological status and quality of life of cases diagnosed with Klinefelter syndrome (KS). METHODS Clinical findings, hormone values and semen analyses in patients with nonmosaic KS (Group 1, n = 121) and those with non-genetic nonobstructive azoospermia (NOA) (Group 2, n = 178) were retrospectively analysed. Sperm retrieval outcomes with microdissection testicular sperm extraction (micro-TESE), fertilisation rates and embryo quality, pregnancy, abortion and live birth rates were compared. Sexual functions were assessed using IIEF-15, quality of life was evaluated and psychological status was assessed. RESULTS There was no difference in terms of age between groups. Sperm retrieval rates was 38% and 55.6% in Groups 1 and 2, respectively (P = .012). Sperm retrieval rates were higher in Group 1 before 31.5 years than in Group 2 (AUC = 0.620 and 0.578). Compared to Group 2, the fertilisation rate was low in Group 1, whereas embryo quality was similar. Live birth rates were 12.5% and 23% in Groups 1 and 2, respectively (P = .392). The education level, libido, erectile functions and general health satisfaction were lower in Group 1 than in Group 2 (P < .005). Depression and anxiety levels were higher in Group 2 than Group 1 (P < .001). CONCLUSION Higher sperm retrieval rate has been achieved in Group 1 younger than 31.5 years. Similar embryo quality is provided between groups. Sexual dysfunction and psychiatric problems were higher in Group 1, with lower satisfaction and general health than Group 2. Patients with KS should be monitored not only with their reproductive functions but also with their general health status.
Collapse
Affiliation(s)
| | - Bulent Ayas
- Department of IVF Center, Ondokuz Mayis University, Samsun, Turkey
| | | | - Ummet Abur
- Department of Medical Genetics, Ondokuz Mayis University, Samsun, Turkey
| | - Recep Bolat
- Psychiatry Clinic, Cevdet Aykan Mental Health Hospital, Tokat, Turkey
| | - Ramazan Asci
- Department of Urology, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
18
|
Mechanisms of Central Hypogonadism. Int J Mol Sci 2021; 22:ijms22158217. [PMID: 34360982 PMCID: PMC8348115 DOI: 10.3390/ijms22158217] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 02/01/2023] Open
Abstract
Reproductive function depends upon an operational hypothalamo–pituitary–gonadal (HPG) axis. Due to its role in determining survival versus reproductive strategies, the HPG axis is vulnerable to a diverse plethora of signals that ultimately manifest with Central Hypogonadism (CH) in all its many guises. Acquired CH can result from any pituitary or hypothalamic lesion, including its treatment (such as surgical resection and/or radiotherapy). The HPG axis is particularly sensitive to the suppressive effects of hyperprolactinaemia that can occur for many reasons, including prolactinomas, and as a side effect of certain drug therapies. Physiologically, prolactin (combined with the suppressive effects of autonomic neural signals from suckling) plays a key role in suppressing the gonadal axis and establishing temporary CH during lactation. Leptin is a further key endocrine regulator of the HPG axis. During starvation, hypoleptinaemia (from diminished fat stores) results in activation of hypothalamic agouti-related peptide neurons that have a dual purpose to enhance appetite (important for survival) and concomitantly suppresses GnRH neurons via effects on neural kisspeptin release. Obesity is associated with hyperleptinaemia and leptin resistance that may also suppress the HPG axis. The suppressibility of the HPG axis also leaves it vulnerable to the effects of external signals that include morphine, anabolic-androgenic steroids, physical trauma and stress, all of which are relatively common causes of CH. Finally, the HPG axis is susceptible to congenital malformations, with reports of mutations within >50 genes that manifest with congenital CH, including Kallmann Syndrome associated with hyposmia or anosmia (reduction or loss of the sense of smell due to the closely associated migration of GnRH with olfactory neurons during embryogenesis). Analogous to the HPG axis itself, patients with CH are often vulnerable, and their clinical management requires both sensitivity and empathy.
Collapse
|
19
|
Wu X, Gao S, Wang L, Bu T, Wu S, Zhou L, Shi J, Wu D, Sun F, Cheng CY. Role of laminin and collagen chains in human spermatogenesis - Insights from studies in rodents and scRNA-Seq transcriptome profiling. Semin Cell Dev Biol 2021; 121:125-132. [PMID: 34325997 DOI: 10.1016/j.semcdb.2021.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022]
Abstract
Studies have demonstrated that biologically active fragments are generated from the basement membrane and the Sertoli cell-spermatid adhesion site known as apical ectoplasmic specialization (apical ES, a testis-specific actin-based anchoring junction) in the rat testis. These bioactive fragments or peptides are produced locally across the seminiferous epithelium through proteolytic cleavage of constituent proteins at the basement membrane and the apical ES. Studies have shown that they are being used to modulate and coordinate cellular functions across the seminiferous epithelium during different stages of the epithelial cycle of spermatogenesis. In this review, we briefly summarize recent findings based on studies using rat testes as a study model regarding the role of these bioactive peptides that serve as a local regulatory network to support spermatogenesis. We also used scRNA-Seq transcriptome datasets in the public domain for OA (obstructive azoospermia) and NAO (non-obstructive azoospermia) human testes versus testes from normal men for analysis in this review. It was shown that there are differential expression of different collagen chains and laminin chains in these testes, suggesting the possibility of a similar local regulatory network in the human testis to support spermatogenesis, and the possible disruption of such network in men is associated with OA and/or NOA.
Collapse
Affiliation(s)
- Xiaolong Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China; The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Sheng Gao
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Lingling Wang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China; The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Tiao Bu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Siwen Wu
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Liwei Zhou
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Jie Shi
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Di Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Fei Sun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China.
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA.
| |
Collapse
|
20
|
Punjani N, Kang C, Lamb DJ, Schlegel PN. Current updates and future perspectives in the evaluation of azoospermia: A systematic review. Arab J Urol 2021; 19:206-214. [PMID: 34552771 PMCID: PMC8451618 DOI: 10.1080/2090598x.2021.1954415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/29/2021] [Indexed: 11/24/2022] Open
Abstract
Objectives: To provide a summary of the current evaluation of azoospermia and insights into future perspectives in the evaluation and counselling of men with azoospermia. Methods: A search of PubMed, Cochrane Reviews and Web of Science databases was performed for full-text English-language articles published between 1943 and 2020 focussing on 'future perspectives', 'azoospermia' and 'evaluation'. Results: Azoospermia represents a severe form of male infertility characterised by sperm production so impaired that there are no sperm present in the ejaculate. The current evaluation of azoospermia focusses on patient history and physical examination with selected adjunctive laboratory investigations including serum hormones, a karyotype and screening for Y chromosome microdeletions. Future diagnostics are focussed on identifying the underlying genetic aetiologies for azoospermia, as well as a greater emphasis on screening for systemic illness that men with severe infertility may be predisposed to develop. Conclusion: Azoospermia represents an extreme form of male infertility, and evaluation relies heavily on history and physical examination, as genetic evaluations for these individuals remain limited. Future evaluation will focus on next-generation sequencing and more rigorous evaluation for possible co-existing and future risk of systemic disease. ABBREVIATIONS: ADGRG2, adhesion G protein-coupled receptor G2; ASRM: American Society of Reproductive Medicine; AZF: azoospermia factor; CBAVD: congenital bilateral absence of the vas deferens; CFTR: cystic fibrosis transmembrane conductance regulator; CRKL: CRK-like proto-oncogene; E2F1: E2F transcription factor 1; HAUS7: HAUS augmin-like complex subunit 7; HR: hazard ratio; KS: Klinefelter syndrome; MAZ, MYC-associated zinc finger protein; NGS: next-generation sequencing; NOA: non-obstructive azoospermia; OA: obstructive azoospermia; RHOX: reproductive homeobox on the X chromosome; SH2: SRC homology 2; TAF7L: TATA-box binding protein associated factor 7-like; TEX11: testis-expressed 11; WES: whole-exome sequencing.
Collapse
Affiliation(s)
- Nahid Punjani
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
| | - Caroline Kang
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
| | - Dolores J. Lamb
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
- Center for Reproductive Genomics, Weill Cornell Medical College, New York, NY, USA
| | - Peter N. Schlegel
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
- Center for Reproductive Genomics, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
21
|
Duffy JMN, Bhattacharya S, Bhattacharya S, Bofill M, Collura B, Curtis C, Evers JLH, Giudice LC, Farquharson RG, Franik S, Hickey M, Hull ML, Jordan V, Khalaf Y, Legro RS, Lensen S, Mavrelos D, Mol BW, Niederberger C, Ng EHY, Puscasiu L, Repping S, Sarris I, Showell M, Strandell A, Vail A, van Wely M, Vercoe M, Vuong NL, Wang AY, Wang R, Wilkinson J, Youssef MA, Farquhar CM. Standardizing definitions and reporting guidelines for the infertility core outcome set: an international consensus development study† ‡. Hum Reprod 2021; 35:2735-2745. [PMID: 33252643 PMCID: PMC7744157 DOI: 10.1093/humrep/deaa243] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/08/2020] [Indexed: 01/21/2023] Open
Abstract
STUDY QUESTION Can consensus definitions for the core outcome set for infertility be identified in order to recommend a standardized approach to reporting? SUMMARY ANSWER Consensus definitions for individual core outcomes, contextual statements and a standardized reporting table have been developed. WHAT IS KNOWN ALREADY Different definitions exist for individual core outcomes for infertility. This variation increases the opportunities for researchers to engage with selective outcome reporting, which undermines secondary research and compromises clinical practice guideline development. STUDY DESIGN, SIZE, DURATION Potential definitions were identified by a systematic review of definition development initiatives and clinical practice guidelines and by reviewing Cochrane Gynaecology and Fertility Group guidelines. These definitions were discussed in a face-to-face consensus development meeting, which agreed consensus definitions. A standardized approach to reporting was also developed as part of the process. PARTICIPANTS/MATERIALS, SETTING, METHODS Healthcare professionals, researchers and people with fertility problems were brought together in an open and transparent process using formal consensus development methods. MAIN RESULTS AND THE ROLE OF CHANCE Forty-four potential definitions were inventoried across four definition development initiatives, including the Harbin Consensus Conference Workshop Group and International Committee for Monitoring Assisted Reproductive Technologies, 12 clinical practice guidelines and Cochrane Gynaecology and Fertility Group guidelines. Twenty-seven participants, from 11 countries, contributed to the consensus development meeting. Consensus definitions were successfully developed for all core outcomes. Specific recommendations were made to improve reporting. LIMITATIONS, REASONS FOR CAUTION We used consensus development methods, which have inherent limitations. There was limited representation from low- and middle-income countries. WIDER IMPLICATIONS OF THE FINDINGS A minimum data set should assist researchers in populating protocols, case report forms and other data collection tools. The generic reporting table should provide clear guidance to researchers and improve the reporting of their results within journal publications and conference presentations. Research funding bodies, the Standard Protocol Items: Recommendations for Interventional Trials statement, and over 80 specialty journals have committed to implementing this core outcome set. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by the Catalyst Fund, Royal Society of New Zealand, Auckland Medical Research Fund and Maurice and Phyllis Paykel Trust. Siladitya Bhattacharya reports being the Editor-in-Chief of Human Reproduction Open and an editor of the Cochrane Gynaecology and Fertility Group. J.L.H.E. reports being the Editor Emeritus of Human Reproduction. R.S.L. reports consultancy fees from Abbvie, Bayer, Ferring, Fractyl, Insud Pharma and Kindex and research sponsorship from Guerbet and Hass Avocado Board. B.W.M. reports consultancy fees from Guerbet, iGenomix, Merck, Merck KGaA and ObsEva. C.N. reports being the Editor-in-Chief of Fertility and Sterility and Section Editor of the Journal of Urology, research sponsorship from Ferring, and a financial interest in NexHand. E.H.Y.N. reports research sponsorship from Merck. A.S. reports consultancy fees from Guerbet. J.W. reports being a statistical editor for the Cochrane Gynaecology and Fertility Group. A.V. reports that he is a Statistical Editor of the Cochrane Gynaecology & Fertility Review Group and of the journal Reproduction. His employing institution has received payment from Human Fertilisation and Embryology Authority for his advice on review of research evidence to inform their 'traffic light' system for infertility treatment 'add-ons'. N.L.V. reports consultancy and conference fees from Ferring, Merck and Merck Sharp and Dohme. The remaining authors declare no competing interests in relation to the work presented. All authors have completed the disclosure form. TRIAL REGISTRATION NUMBER Core Outcome Measures in Effectiveness Trials Initiative: 1023.
Collapse
Affiliation(s)
- J M N Duffy
- King's Fertility, Fetal Medicine Research Institute, London, UK.,Institute for Women's Health, University College London, London, UK
| | - S Bhattacharya
- School of Medicine, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - S Bhattacharya
- School of Medicine, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - M Bofill
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - B Collura
- RESOLVE, The National Infertility Association, VA, USA
| | - C Curtis
- Fertility New Zealand, Auckland, New Zealand.,School of Psychology, University of Waikato, Hamilton, New Zealand
| | - J L H Evers
- Maastricht University Medical Centre, Maastricht, The Netherlands
| | - L C Giudice
- Center for Research, Innovation and Training in Reproduction and Infertility, Center for Reproductive Sciences, University of California, San Francisco, CA, USA.,International Federation of Fertility Societies, Philadelphia, PA, USA
| | - R G Farquharson
- Department of Obstetrics and Gynaecology, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - S Franik
- Department of Obstetrics and Gynaecology, Münster University Hospital, Münster, Germany
| | - M Hickey
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - M L Hull
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - V Jordan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Y Khalaf
- Department of Women and Children's Health, King's College London, Guy's Hospital, London
| | - R S Legro
- Department of Obstetrics and Gynaecology, Penn State College of Medicine, PA, USA
| | - S Lensen
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - D Mavrelos
- Reproductive Medicine Unit, University College Hospital, London, UK
| | - B W Mol
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - C Niederberger
- Department of Urology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - E H Y Ng
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong.,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, China
| | - L Puscasiu
- Pharmacy, Sciences and Technology, University of Medicine, Targu Mures, Romania
| | - S Repping
- Amsterdam University Medical Centers, Amsterdam, The Netherlands.,National Health Care Institute, Diemen, The Netherlands
| | - I Sarris
- King's Fertility, Fetal Medicine Research Institute, London, UK
| | - M Showell
- Cochrane Gynaecology and Fertility Group, University of Auckland, Auckland, New Zealand
| | - A Strandell
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - A Vail
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - M van Wely
- Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - M Vercoe
- Cochrane Gynaecology and Fertility Group, University of Auckland, Auckland, New Zealand
| | - N L Vuong
- Department of Obstetrics and Gynaecology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - A Y Wang
- Faculty of Health, University of Technology, Sydney, Broadway, Australia
| | - R Wang
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - J Wilkinson
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - M A Youssef
- Department of Obstetrics & Gynaecology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - C M Farquhar
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand.,Cochrane Gynaecology and Fertility Group, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
22
|
The Scenario of Adoption and Foster Care in Relation to the Reproductive Medicine Practice in Asia. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18073466. [PMID: 33810525 PMCID: PMC8037075 DOI: 10.3390/ijerph18073466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 11/17/2022]
Abstract
In vitro fertilization (IVF) is a hallmark of reproductive medicine. However, the inconclusive outcome leads to marital disharmonies; thus, the choices of gamete donation and surrogacy (GD/S) are often offered. In restricted countries, the child-rearing choice through foster/adoption care is promising, but the uptake remains low. We explore the current reproductive services and adoption scenarios in Asian countries to delineate this issue. The web and literature search using PubMed and Ichushi was conducted in Japanese and English using the keywords "adoption", "foster care", "reproductive medicine", including the interview with the respective Asian clinicians. We found that an established adoption system was seen in China, Malaysia, and the Philippines, mainly due to the restriction of GD/S. Although GD/S were allowed in Thailand, Singapore, and India, the different local affordability of IVF cost led to various adoption system scenarios. Nevertheless, the country's economic aspect does influence the establishment of adoption care, mainly due to financial support from local government. Otherwise, the significant barrier was the cultural/religious background leading to low adoption rates. We concluded that the adoption option should always be highlighted as an alternative strategy as it synergistically contributes to children's and infertile couples' welfare.
Collapse
|
23
|
[Testicular sperm extraction in male infertility : Indications, success rates, practical implementation, and possible complications]. Urologe A 2021; 60:921-931. [PMID: 33660059 DOI: 10.1007/s00120-021-01480-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Unwanted childlessness is a burden on a couple's relationship. The therapeutic spectrum of male infertility has increased significantly in recent years so that even azoospermia patients can be given biological paternity by testicular sperm extraction (TESE). OBJECTIVES The indications, success rates, practical implementation, and possible complications of conventional and microscopic TESE in male infertility are presented in this review. METHODS A nonsystematic search of the relevant literature was carried out. RESULTS In obstructive azoospermia (OA), primarily desobstructive surgical procedures are used, while TESE is the surgical procedure of choice in nonobstructive azoospermia (NOA). In the latter, sperm extraction can be performed conventionally or microscopically (mTESE) assisted, whereby the latter offers an advantage in terms of sperm detection rate in the case of small testicular volumes (<12 ml), chemotherapy, Klinefelter's disease and AZFc microdeletions. The sperm detection rate of TESE is about 50%. Postoperative controls are useful because of the possible induction of symptomatic hypogonadism. CONCLUSION Before performing TESE, determining the hormone status and human genetic clarification are necessary. Any costs incurred and the possibility of missing sperm proof must be discussed. Close cooperation between andrologists, gynecologists, reproductive physicians, and human geneticists is necessary. All in all, TESE is a safe surgical procedure with a low complication rate.
Collapse
|
24
|
Agarwal A, Baskaran S, Parekh N, Cho CL, Henkel R, Vij S, Arafa M, Panner Selvam MK, Shah R. Male infertility. Lancet 2021; 397:319-333. [PMID: 33308486 DOI: 10.1016/s0140-6736(20)32667-2] [Citation(s) in RCA: 495] [Impact Index Per Article: 165.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
It is estimated that infertility affects 8-12% of couples globally, with a male factor being a primary or contributing cause in approximately 50% of couples. Causes of male subfertility vary highly, but can be related to congenital, acquired, or idiopathic factors that impair spermatogenesis. Many health conditions can affect male fertility, which underscores the need for a thorough evaluation of patients to identify treatable or reversible lifestyle factors or medical conditions. Although semen analysis remains the cornerstone for evaluating male infertility, advanced diagnostic tests to investigate sperm quality and function have been developed to improve diagnosis and management. The use of assisted reproductive techniques has also substantially improved the ability of couples with infertility to have biological children. This Seminar aims to provide a comprehensive overview of the assessment and management of men with infertility, along with current controversies and future endeavours.
Collapse
Affiliation(s)
- Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA.
| | - Saradha Baskaran
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Neel Parekh
- Department of Urology, Cleveland Clinic, Cleveland, OH, USA
| | - Chak-Lam Cho
- SH Ho Urology Center, Department of Surgery, Chinese University of Hong Kong, Hong Kong
| | - Ralf Henkel
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA; Department of Medical Bioscience, University of Western Cape, Bellville, South Africa; Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Sarah Vij
- Department of Urology, Cleveland Clinic, Cleveland, OH, USA
| | - Mohamed Arafa
- Male Infertility Unit, Urology Department, Hamad Medical Corporation, Doha, Qatar; Andrology Department, Cairo University, Cairo, Egypt
| | | | - Rupin Shah
- Department of Urology, Lilavati Hospital and Research Center, Mumbai, India
| |
Collapse
|
25
|
Duffy JMN, Adamson GD, Benson E, Bhattacharya S, Bhattacharya S, Bofill M, Brian K, Collura B, Curtis C, Evers JLH, Farquharson RG, Fincham A, Franik S, Giudice LC, Glanville E, Hickey M, Horne AW, Hull ML, Johnson NP, Jordan V, Khalaf Y, Knijnenburg JML, Legro RS, Lensen S, MacKenzie J, Mavrelos D, Mol BW, Morbeck DE, Nagels H, Ng EHY, Niederberger C, Otter AS, Puscasiu L, Rautakallio-Hokkanen S, Sadler L, Sarris I, Showell M, Stewart J, Strandell A, Strawbridge C, Vail A, van Wely M, Vercoe M, Vuong NL, Wang AY, Wang R, Wilkinson J, Wong K, Wong TY, Farquhar CM. Top 10 priorities for future infertility research: an international consensus development study. Fertil Steril 2021; 115:180-190. [PMID: 33272617 DOI: 10.1016/j.fertnstert.2020.11.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/05/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022]
Abstract
STUDY QUESTION Can the priorities for future research in infertility be identified? SUMMARY ANSWER The top 10 research priorities for the four areas of male infertility, female and unexplained infertility, medically assisted reproduction, and ethics, access, and organization of care for people with fertility problems were identified. WHAT IS KNOWN ALREADY Many fundamental questions regarding the prevention, management, and consequences of infertility remain unanswered. This is a barrier to improving the care received by those people with fertility problems. STUDY DESIGN, SIZE, DURATION Potential research questions were collated from an initial international survey, a systematic review of clinical practice guidelines, and Cochrane systematic reviews. A rationalized list of confirmed research uncertainties was prioritized in an interim international survey. Prioritized research uncertainties were discussed during a consensus development meeting. Using a formal consensus development method, the modified nominal group technique, diverse stakeholders identified the top 10 research priorities for each of the categories male infertility, female and unexplained infertility, medically assisted reproduction, and ethics, access, and organization of care. PARTICIPANTS/MATERIALS, SETTING, METHODS Healthcare professionals, people with fertility problems, and others (healthcare funders, healthcare providers, healthcare regulators, research funding bodies and researchers) were brought together in an open and transparent process using formal consensus methods advocated by the James Lind Alliance. MAIN RESULTS AND THE ROLE OF CHANCE The initial survey was completed by 388 participants from 40 countries, and 423 potential research questions were submitted. Fourteen clinical practice guidelines and 162 Cochrane systematic reviews identified a further 236 potential research questions. A rationalized list of 231 confirmed research uncertainties were entered into an interim prioritization survey completed by 317 respondents from 43 countries. The top 10 research priorities for each of the four categories male infertility, female and unexplained infertility (including age-related infertility, ovarian cysts, uterine cavity abnormalities, and tubal factor infertility), medically assisted reproduction (including ovarian stimulation, IUI, and IVF), and ethics, access, and organization of care, were identified during a consensus development meeting involving 41 participants from 11 countries. These research priorities were diverse and seek answers to questions regarding prevention, treatment, and the longer-term impact of infertility. They highlight the importance of pursuing research which has often been overlooked, including addressing the emotional and psychological impact of infertility, improving access to fertility treatment, particularly in lower resource settings, and securing appropriate regulation. Addressing these priorities will require diverse research methodologies, including laboratory-based science, qualitative and quantitative research, and population science. LIMITATIONS, REASONS FOR CAUTION We used consensus development methods, which have inherent limitations, including the representativeness of the participant sample, methodological decisions informed by professional judgement, and arbitrary consensus definitions. WIDER IMPLICATIONS OF THE FINDINGS We anticipate that identified research priorities, developed to specifically highlight the most pressing clinical needs as perceived by healthcare professionals, people with fertility problems, and others, will help research funding organizations and researchers to develop their future research agenda. STUDY FUNDING/ COMPETING INTEREST(S) The study was funded by the Auckland Medical Research Foundation, Catalyst Fund, Royal Society of New Zealand, and Maurice and Phyllis Paykel Trust. Geoffrey Adamson reports research sponsorship from Abbott, personal fees from Abbott and LabCorp, a financial interest in Advanced Reproductive Care, committee membership of the FIGO Committee on Reproductive Medicine, International Committee for Monitoring Assisted Reproductive Technologies, International Federation of Fertility Societies, and World Endometriosis Research Foundation, and research sponsorship of the International Committee for Monitoring Assisted Reproductive Technologies from Abbott and Ferring. Siladitya Bhattacharya reports being the Editor-in-Chief of Human Reproduction Open and editor for the Cochrane Gynaecology and Fertility Group. Hans Evers reports being the Editor Emeritus of Human Reproduction. Andrew Horne reports research sponsorship from the Chief Scientist's Office, Ferring, Medical Research Council, National Institute for Health Research, and Wellbeing of Women and consultancy fees from Abbvie, Ferring, Nordic Pharma, and Roche Diagnostics. M. Louise Hull reports grants from Merck, grants from Myovant, grants from Bayer, outside the submitted work and ownership in Embrace Fertility, a private fertility company. Neil Johnson reports research sponsorship from Abb-Vie and Myovant Sciences and consultancy fees from Guerbet, Myovant Sciences, Roche Diagnostics, and Vifor Pharma. José Knijnenburg reports research sponsorship from Ferring and Theramex. Richard Legro reports consultancy fees from Abbvie, Bayer, Ferring, Fractyl, Insud Pharma and Kindex and research sponsorship from Guerbet and Hass Avocado Board. Ben Mol reports consultancy fees from Guerbet, iGenomix, Merck, Merck KGaA and ObsEva. Ernest Ng reports research sponsorship from Merck. Craig Niederberger reports being the Co Editor-in-Chief of Fertility and Sterility and Section Editor of the Journal of Urology, research sponsorship from Ferring, and retains a financial interest in NexHand. Jane Stewart reports being employed by a National Health Service fertility clinic, consultancy fees from Merck for educational events, sponsorship to attend a fertility conference from Ferring, and being a clinical subeditor of Human Fertility. Annika Strandell reports consultancy fees from Guerbet. Jack Wilkinson reports being a statistical editor for the Cochrane Gynaecology and Fertility Group. Andy Vail reports that he is a Statistical Editor of the Cochrane Gynaecology & Fertility Review Group and of the journal Reproduction. His employing institution has received payment from HFEA for his advice on review of research evidence to inform their 'traffic light' system for infertility treatment 'add-ons'. Lan Vuong reports consultancy and conference fees from Ferring, Merck and Merck Sharp and Dohme. The remaining authors declare no competing interests in relation to the present work. All authors have completed the disclosure form. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- J M N Duffy
- King's Fertility, Fetal Medicine Research Institute, London, UK; Institute for Women's Health, University College London, London, UK.
| | - G D Adamson
- ARC Fertility, Cupertino, California, United States
| | - E Benson
- Patient and Public Participation Group, Priority Setting Partnership for Infertility, University of Auckland, Auckland, New Zealand
| | - S Bhattacharya
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - S Bhattacharya
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - M Bofill
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - K Brian
- Women's Network, Royal College of Obstetricians and Gynecologists, London, UK
| | - B Collura
- Resolve: The National Infertility Association, Virginia, United States
| | - C Curtis
- School of Psychology, University of Waikato, Hamilton, New Zealand
| | - J L H Evers
- Centre for Reproductive Medicine and Biology, University Medical Centre Maastricht, Maastricht, The Netherlands
| | - R G Farquharson
- Department of Obstetrics and Gynaecology, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | | | - S Franik
- Department of Obstetrics and Gynaecology, Münster University Hospital, Münster, Germany
| | - L C Giudice
- Center for Research, Innovation and Training in Reproduction and Infertility, Center for Reproductive Sciences, University of California, San Francisco, California, United States; International Federation of Fertility Societies, Mount Royal, New Jersey, United States
| | - E Glanville
- Auckland District Health Board, Auckland, New Zealand
| | - M Hickey
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - A W Horne
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - M L Hull
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - N P Johnson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - V Jordan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Y Khalaf
- Department of Women and Children's Health, Kings College London, London, UK
| | | | - R S Legro
- Department of Obstetrics and Gynaecology, Penn State College of Medicine, Pennsylvania
| | - S Lensen
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | | | - D Mavrelos
- Reproductive Medicine Unit, University College Hospital, London, UK
| | - B W Mol
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - D E Morbeck
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand; Fertility Associates, Auckland, New Zealand
| | - H Nagels
- Cochrane Gynaecology and Fertility, University of Auckland, Auckland, New Zealand
| | - E H Y Ng
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, China
| | - C Niederberger
- Department of Urology, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | | | - L Puscasiu
- Pharmacy, Science, and Technology, University of Medicine, Targu Mures, Romania; Center for Reproductive Medicine, Amsterdam Reproduction and Development Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | | | - L Sadler
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand; Auckland District Health Board, Auckland, New Zealand
| | - I Sarris
- King's Fertility, Fetal Medicine Research Institute, London, UK
| | - M Showell
- Cochrane Gynaecology and Fertility, University of Auckland, Auckland, New Zealand
| | - J Stewart
- British Fertility Society, Middlesex, UK
| | - A Strandell
- Sahlgrenska Academy, Dept of Obstetrics and Gynecology, University of Gothenburg, Sahlgrenska University Hospital, Göteborg, Sweden
| | | | - A Vail
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - M van Wely
- Center for Reproductive Medicine, Amsterdam Reproduction and Development Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - M Vercoe
- Cochrane Gynaecology and Fertility, University of Auckland, Auckland, New Zealand
| | - N L Vuong
- Department of Obstetrics and Gynaecology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - A Y Wang
- Australian Centre for Public and Population Health Research, Faculty of Health, University of Technology Sydney, Australia
| | - R Wang
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - J Wilkinson
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - K Wong
- School of Psychology, University of Waikato, Hamilton, New Zealand
| | - T Y Wong
- Auckland District Health Board, Auckland, New Zealand
| | - C M Farquhar
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand; Cochrane Gynaecology and Fertility, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
26
|
Yang J, Wu X, Yang B, Liu Y, Cheng R, Gong Z, Sun F. Mn(ii)-Catalysed ortho-alkenylation of aromatic amines and its application in reproductive diseases. RSC Adv 2020; 11:164-167. [PMID: 35423065 PMCID: PMC8690850 DOI: 10.1039/d0ra10172a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/03/2022] Open
Abstract
A Mn(ii)-catalysed ortho-alkenylation of aromatic amines and its application in reproductive diseases were developed. The use of MnCl2 was critical for the ortho-alkenylation of aromatic amines. The general applicability of this procedure was highlighted by the synthesis of 27 vinylanilines, with good regioselectivities. The value of our approach in practical applications was investigated by studying the effects of one of the compounds 3m on 8 week-old adult male rats with azoospermia as a mammalian model. The results show that a small amount of sperm will gradually be produced in the epididymis and testes by treatment of 8 week-old adult male rats with azoospermia with 1 mg kg−13m after two weeks, while treatment with 10 mg kg−13m led to obvious sperm production. Notably, if we increase the dose to 100 mg kg−1, there will be a lot of sperm production in the epididymis and testes after two weeks of treatment. The results of this study will be of great significance in research on drugs for treating azoospermia and oligospermia diseases. A Mn(ii)-catalysed ortho-alkenylation of aromatic amines and its application in reproductive diseases were developed.![]()
Collapse
Affiliation(s)
- Jinfei Yang
- Medical School, Institute of Reproductive Medicine, Nantong University Nantong 226019 China
| | - Xiaolong Wu
- Medical School, Institute of Reproductive Medicine, Nantong University Nantong 226019 China
| | - Banghua Yang
- Medical School, Institute of Reproductive Medicine, Nantong University Nantong 226019 China
| | - Yirong Liu
- Medical School, Institute of Reproductive Medicine, Nantong University Nantong 226019 China
| | - Rui Cheng
- Medical School, Institute of Reproductive Medicine, Nantong University Nantong 226019 China
| | - Zijun Gong
- Medical School, Institute of Reproductive Medicine, Nantong University Nantong 226019 China
| | - Fei Sun
- Medical School, Institute of Reproductive Medicine, Nantong University Nantong 226019 China
| |
Collapse
|
27
|
Duffy JMN, Adamson GD, Benson E, Bhattacharya S, Bhattacharya S, Bofill M, Brian K, Collura B, Curtis C, Evers JLH, Farquharson RG, Fincham A, Franik S, Giudice LC, Glanville E, Hickey M, Horne AW, Hull ML, Johnson NP, Jordan V, Khalaf Y, Knijnenburg JML, Legro RS, Lensen S, MacKenzie J, Mavrelos D, Mol BW, Morbeck DE, Nagels H, Ng EHY, Niederberger C, Otter AS, Puscasiu L, Rautakallio-Hokkanen S, Sadler L, Sarris I, Showell M, Stewart J, Strandell A, Strawbridge C, Vail A, van Wely M, Vercoe M, Vuong NL, Wang AY, Wang R, Wilkinson J, Wong K, Wong TY, Farquhar CM. Top 10 priorities for future infertility research: an international consensus development study† ‡. Hum Reprod 2020; 35:2715-2724. [PMID: 33252677 PMCID: PMC7744161 DOI: 10.1093/humrep/deaa242] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/05/2020] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Can the priorities for future research in infertility be identified? SUMMARY ANSWER The top 10 research priorities for the four areas of male infertility, female and unexplained infertility, medically assisted reproduction and ethics, access and organization of care for people with fertility problems were identified. WHAT IS KNOWN ALREADY Many fundamental questions regarding the prevention, management and consequences of infertility remain unanswered. This is a barrier to improving the care received by those people with fertility problems. STUDY DESIGN, SIZE, DURATION Potential research questions were collated from an initial international survey, a systematic review of clinical practice guidelines and Cochrane systematic reviews. A rationalized list of confirmed research uncertainties was prioritized in an interim international survey. Prioritized research uncertainties were discussed during a consensus development meeting. Using a formal consensus development method, the modified nominal group technique, diverse stakeholders identified the top 10 research priorities for each of the categories male infertility, female and unexplained infertility, medically assisted reproduction and ethics, access and organization of care. PARTICIPANTS/MATERIALS, SETTING, METHODS Healthcare professionals, people with fertility problems and others (healthcare funders, healthcare providers, healthcare regulators, research funding bodies and researchers) were brought together in an open and transparent process using formal consensus methods advocated by the James Lind Alliance. MAIN RESULTS AND THE ROLE OF CHANCE The initial survey was completed by 388 participants from 40 countries, and 423 potential research questions were submitted. Fourteen clinical practice guidelines and 162 Cochrane systematic reviews identified a further 236 potential research questions. A rationalized list of 231 confirmed research uncertainties was entered into an interim prioritization survey completed by 317 respondents from 43 countries. The top 10 research priorities for each of the four categories male infertility, female and unexplained infertility (including age-related infertility, ovarian cysts, uterine cavity abnormalities and tubal factor infertility), medically assisted reproduction (including ovarian stimulation, IUI and IVF) and ethics, access and organization of care were identified during a consensus development meeting involving 41 participants from 11 countries. These research priorities were diverse and seek answers to questions regarding prevention, treatment and the longer-term impact of infertility. They highlight the importance of pursuing research which has often been overlooked, including addressing the emotional and psychological impact of infertility, improving access to fertility treatment, particularly in lower resource settings and securing appropriate regulation. Addressing these priorities will require diverse research methodologies, including laboratory-based science, qualitative and quantitative research and population science. LIMITATIONS, REASONS FOR CAUTION We used consensus development methods, which have inherent limitations, including the representativeness of the participant sample, methodological decisions informed by professional judgment and arbitrary consensus definitions. WIDER IMPLICATIONS OF THE FINDINGS We anticipate that identified research priorities, developed to specifically highlight the most pressing clinical needs as perceived by healthcare professionals, people with fertility problems and others, will help research funding organizations and researchers to develop their future research agenda. STUDY FUNDING/COMPETING INTEREST(S) The study was funded by the Auckland Medical Research Foundation, Catalyst Fund, Royal Society of New Zealand and Maurice and Phyllis Paykel Trust. G.D.A. reports research sponsorship from Abbott, personal fees from Abbott and LabCorp, a financial interest in Advanced Reproductive Care, committee membership of the FIGO Committee on Reproductive Medicine, International Committee for Monitoring Assisted Reproductive Technologies, International Federation of Fertility Societies and World Endometriosis Research Foundation, and research sponsorship of the International Committee for Monitoring Assisted Reproductive Technologies from Abbott and Ferring. Siladitya Bhattacharya reports being the Editor-in-Chief of Human Reproduction Open and editor for the Cochrane Gynaecology and Fertility Group. J.L.H.E. reports being the Editor Emeritus of Human Reproduction. A.W.H. reports research sponsorship from the Chief Scientist's Office, Ferring, Medical Research Council, National Institute for Health Research and Wellbeing of Women and consultancy fees from AbbVie, Ferring, Nordic Pharma and Roche Diagnostics. M.L.H. reports grants from Merck, grants from Myovant, grants from Bayer, outside the submitted work and ownership in Embrace Fertility, a private fertility company. N.P.J. reports research sponsorship from AbbVie and Myovant Sciences and consultancy fees from Guerbet, Myovant Sciences, Roche Diagnostics and Vifor Pharma. J.M.L.K. reports research sponsorship from Ferring and Theramex. R.S.L. reports consultancy fees from AbbVie, Bayer, Ferring, Fractyl, Insud Pharma and Kindex and research sponsorship from Guerbet and Hass Avocado Board. B.W.M. reports consultancy fees from Guerbet, iGenomix, Merck, Merck KGaA and ObsEva. E.H.Y.N. reports research sponsorship from Merck. C.N. reports being the Co Editor-in-Chief of Fertility and Sterility and Section Editor of the Journal of Urology, research sponsorship from Ferring and retains a financial interest in NexHand. J.S. reports being employed by a National Health Service fertility clinic, consultancy fees from Merck for educational events, sponsorship to attend a fertility conference from Ferring and being a clinical subeditor of Human Fertility. A.S. reports consultancy fees from Guerbet. J.W. reports being a statistical editor for the Cochrane Gynaecology and Fertility Group. A.V. reports that he is a Statistical Editor of the Cochrane Gynaecology & Fertility Review Group and the journal Reproduction. His employing institution has received payment from Human Fertilisation and Embryology Authority for his advice on review of research evidence to inform their 'traffic light' system for infertility treatment 'add-ons'. N.L.V. reports consultancy and conference fees from Ferring, Merck and Merck Sharp and Dohme. The remaining authors declare no competing interests in relation to the present work. All authors have completed the disclosure form. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- J M N Duffy
- King’s Fertility, Fetal Medicine Research Institute, London, UK
- Institute for Women’s Health, University College London, London, UK
| | | | - E Benson
- Patient and Public Participation Group, Priority Setting Partnership for Infertility, University of Auckland, Auckland, New Zealand
| | - S Bhattacharya
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - S Bhattacharya
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - M Bofill
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - K Brian
- Women’s Network, Royal College of Obstetricians and Gynecologists, London, UK
| | - B Collura
- Resolve: The National Infertility Association, VA, USA
| | - C Curtis
- School of Psychology, University of Waikato, Hamilton, New Zealand
| | - J L H Evers
- Centre for Reproductive Medicine and Biology, University Medical Centre Maastricht, Maastricht, The Netherlands
| | - R G Farquharson
- Department of Obstetrics and Gynaecology, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | | | - S Franik
- Department of Obstetrics and Gynaecology, Münster University Hospital, Münster, Germany
| | - L C Giudice
- Center for Research, Innovation and Training in Reproduction and Infertility, Center for Reproductive Sciences, University of California, San Francisco, CA, USA
- International Federation of Fertility Societies, Mount Royal, NJ, USA
| | - E Glanville
- Auckland District Health Board, Auckland, New Zealand
| | - M Hickey
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - A W Horne
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - M L Hull
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - N P Johnson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - V Jordan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Y Khalaf
- Department of Women and Children’s Health, Kings College London, London, UK
| | | | - R S Legro
- Department of Obstetrics and Gynaecology, Penn State College of Medicine, PA, USA
| | - S Lensen
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | | | - D Mavrelos
- Reproductive Medicine Unit, University College Hospital, London, UK
| | - B W Mol
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - D E Morbeck
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Fertility Associates, Auckland, New Zealand
| | - H Nagels
- Cochrane Gynaecology and Fertility, University of Auckland, Auckland, New Zealand
| | - E H Y Ng
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, China
| | - C Niederberger
- Department of Urology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | - L Puscasiu
- ARC Fertility, Cupertino, CA, USA
- Institute for Women’s Health, University College London, London, UK
- Center for Reproductive Medicine, Amsterdam Reproduction and Development Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | | | - L Sadler
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Auckland District Health Board, Auckland, New Zealand
| | - I Sarris
- King’s Fertility, Fetal Medicine Research Institute, London, UK
| | - M Showell
- Cochrane Gynaecology and Fertility, University of Auckland, Auckland, New Zealand
| | - J Stewart
- British Fertility Society, Middlesex, UK
| | - A Strandell
- Sahlgrenska Academy, Department of Obstetrics and Gynecology, University of Gothenburg, Sahlgrenska University Hospital, Göteborg, Sweden
| | | | - A Vail
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - M van Wely
- Center for Reproductive Medicine, Amsterdam Reproduction and Development Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - M Vercoe
- Cochrane Gynaecology and Fertility, University of Auckland, Auckland, New Zealand
| | - N L Vuong
- Department of Obstetrics and Gynaecology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - A Y Wang
- Australian Centre for Public and Population Health Research, Faculty of Health, University of Technology, Sydney, Australia
| | - R Wang
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - J Wilkinson
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - K Wong
- School of Psychology, University of Waikato, Hamilton, New Zealand
| | - T Y Wong
- Auckland District Health Board, Auckland, New Zealand
| | - C M Farquhar
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Cochrane Gynaecology and Fertility, University of Auckland, Auckland, New Zealand
| |
Collapse
|
28
|
Tharakan T, Salonia A, Corona G, Dhillo W, Minhas S, Jayasena C. The Role of Hormone Stimulation in Men With Nonobstructive Azoospermia Undergoing Surgical Sperm Retrieval. J Clin Endocrinol Metab 2020; 105:5893978. [PMID: 32810280 DOI: 10.1210/clinem/dgaa556] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Nonobstructive azoospermia, (NOA) is the most common cause of azoospermia. NOA is characterized by hypergonadotropic hypogonadism, testicular failure, and impaired spermatogenesis. The recent development of surgical sperm retrieval techniques such as microsurgical testicular sperm extraction (mTESE) has, for the first time, allowed some men with NOA to father biological children. It is common practice for endocrine stimulation therapies such as gonadotropins, selective estrogen receptor modulators (SERMs), and aromatase inhibitors to be used prior to mTESE to increase intratesticular testosterone synthesis with the aim of improving sperm retrieval rates; however, there is currently a paucity of data underpinning their safety and efficacy. We present 2 cases of men with NOA undergoing endocrine stimulation therapy and mTESE. We also discuss the current evidence and controversies associated with the use of hormonal stimulation therapy in couples affected by this severe form of male infertility.
Collapse
Affiliation(s)
- Tharu Tharakan
- Section of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Urology, Imperial Healthcare NHS Trust, Charing Cross Hospital, London, United Kingdom
| | - Andrea Salonia
- Division of Experimental Oncology/Unite of Urology, URI, IRCCS Ospedale, San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Giovanni Corona
- Andrology Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Waljit Dhillo
- Section of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Suks Minhas
- Department of Urology, Imperial Healthcare NHS Trust, Charing Cross Hospital, London, United Kingdom
| | - Channa Jayasena
- Section of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| |
Collapse
|
29
|
Abstract
The understanding of male factors of infertility has grown exponentially in the past ten years. While clear guidelines for obstructive azoospermia have been developed, management of non-obstructive azoospermia has lagged. Specifically, management of Kallmann Syndrome and central non-obstructive azoospermia has been limited by a lack of understanding of the molecular pathogenesis and investigational trials exploring the best option for management and fertility in these patients. This review aims to summarize our current understanding of the causes of central hypogonadotropic hypogonadism with a focus on genetic etiologies while also discussing options that endocrinologists and urologists can utilize to successfully treat this group of infertile men.
Collapse
Affiliation(s)
| | | | - Bobby B Najari
- NYU Langone Department of Urology, Department of Population Health, New York, NY, USA.
| |
Collapse
|
30
|
Duffy JMN, Bhattacharya S, Bhattacharya S, Bofill M, Collura B, Curtis C, Evers JLH, Giudice LC, Farquharson RG, Franik S, Hickey M, Hull ML, Jordan V, Khalaf Y, Legro RS, Lensen S, Mavrelos D, Mol BW, Niederberger C, Ng EHY, Puscasiu L, Repping S, Sarris I, Showell M, Strandell A, Vail A, van Wely M, Vercoe M, Vuong NL, Wang AY, Wang R, Wilkinson J, Youssef MA, Farquhar CM. Standardizing definitions and reporting guidelines for the infertility core outcome set: an international consensus development study. Fertil Steril 2020; 115:201-212. [PMID: 33272619 DOI: 10.1016/j.fertnstert.2020.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/08/2020] [Accepted: 07/22/2020] [Indexed: 01/21/2023]
Abstract
STUDY QUESTION Can consensus definitions for the core outcome set for infertility be identified in order to recommend a standardized approach to reporting? SUMMARY ANSWER Consensus definitions for individual core outcomes, contextual statements, and a standardized reporting table have been developed. WHAT IS KNOWN ALREADY Different definitions exist for individual core outcomes for infertility. This variation increases the opportunities for researchers to engage with selective outcome reporting, which undermines secondary research and compromises clinical practice guideline development. STUDY DESIGN, SIZE, DURATION Potential definitions were identified by a systematic review of definition development initiatives and clinical practice guidelines and by reviewing Cochrane Gynaecology and Fertility Group guidelines. These definitions were discussed in a face-to-face consensus development meeting, which agreed consensus definitions. A standardized approach to reporting was also developed as part of the process. PARTICIPANTS/MATERIALS, SETTING, METHODS Healthcare professionals, researchers, and people with fertility problems were brought together in an open and transparent process using formal consensus development methods. MAIN RESULTS AND THE ROLE OF CHANCE Forty-four potential definitions were inventoried across four definition development initiatives, including the Harbin Consensus Conference Workshop Group and International Committee for Monitoring Assisted Reproductive Technologies, 12 clinical practice guidelines, and Cochrane Gynaecology and Fertility Group guidelines. Twenty-seven participants, from 11 countries, contributed to the consensus development meeting. Consensus definitions were successfully developed for all core outcomes. Specific recommendations were made to improve reporting. LIMITATIONS, REASONS FOR CAUTION We used consensus development methods, which have inherent limitations. There was limited representation from low- and middle-income countries. WIDER IMPLICATIONS OF THE FINDINGS A minimum data set should assist researchers in populating protocols, case report forms, and other data collection tools. The generic reporting table should provide clear guidance to researchers and improve the reporting of their results within journal publications and conference presentations. Research funding bodies, the Standard Protocol Items: Recommendations for Interventional Trials statement, and over 80 specialty journals have committed to implementing this core outcome set. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by the Catalyst Fund, Royal Society of New Zealand, Auckland Medical Research Fund, and Maurice and Phyllis Paykel Trust. Siladitya Bhattacharya reports being the Editor-in-Chief of Human Reproduction Open and an editor of the Cochrane Gynaecology and Fertility group. Hans Evers reports being the Editor Emeritus of Human Reproduction. Richard Legro reports consultancy fees from Abbvie, Bayer, Ferring, Fractyl, Insud Pharma and Kindex and research sponsorship from Guerbet and Hass Avocado Board. Ben Mol reports consultancy fees from Guerbet, iGenomix, Merck, Merck KGaA and ObsEva. Craig Niederberger reports being the Editor-in-Chief of Fertility and Sterility and Section Editor of the Journal of Urology, research sponsorship from Ferring, and a financial interest in NexHand. Ernest Ng reports research sponsorship from Merck. Annika Strandell reports consultancy fees from Guerbet. Jack Wilkinson reports being a statistical editor for the Cochrane Gynaecology and Fertility group. Andy Vail reports that he is a Statistical Editor of the Cochrane Gynaecology & Fertility Review Group and of the journal Reproduction. His employing institution has received payment from HFEA for his advice on review of research evidence to inform their 'traffic light' system for infertility treatment 'add-ons'. Lan Vuong reports consultancy and conference fees from Ferring, Merck and Merck Sharp and Dohme. The remaining authors declare no competing interests in relation to the work presented. All authors have completed the disclosure form. TRIAL REGISTRATION NUMBER Core Outcome Measures in Effectiveness Trials Initiative: 1023.
Collapse
Affiliation(s)
- J M N Duffy
- King's Fertility, Fetal Medicine Research Institute, London, UK; Institute for Women's Health, University College London, London, UK.
| | - S Bhattacharya
- School of Medicine, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - S Bhattacharya
- School of Medicine, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - M Bofill
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - B Collura
- RESOLVE: The National Infertility Association, Virginia, United States
| | - C Curtis
- Fertility New Zealand, Auckland, New Zealand; School of Psychology, University of Waikato, Hamilton, New Zealand
| | - J L H Evers
- Maastricht University Medical Centre, Maastricht, The Netherlands
| | - L C Giudice
- Center for Research, Innovation and Training in Reproduction and Infertility, Center for Reproductive Sciences, University of California, San Francisco, California, United States; International Federation of Fertility Societies, Philadelphia, Pennsylvania, United States
| | - R G Farquharson
- Department of Obstetrics and Gynaecology, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - S Franik
- Department of Obstetrics and Gynaecology, Münster University Hospital, Münster, Germany
| | - M Hickey
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - M L Hull
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - V Jordan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Y Khalaf
- Department of Women and Children's Health, King's College London, Guy's Hospital, London
| | - R S Legro
- Department of Obstetrics and Gynaecology, Penn State College of Medicine, Pennsylvania
| | - S Lensen
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - D Mavrelos
- Reproductive Medicine Unit, University College Hospital, London, UK
| | - B W Mol
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - C Niederberger
- Department of Urology, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - E H Y Ng
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, China
| | - L Puscasiu
- University of Medicine, Pharmacy, Sciences and Technology, Targu Mures, Romania
| | - S Repping
- Amsterdam University Medical Centers, Amsterdam, The Netherlands; National Health Care Institute, Diemen, The Netherlands
| | - I Sarris
- King's Fertility, Fetal Medicine Research Institute, London, UK
| | - M Showell
- Cochrane Gynaecology and Fertility Group, University of Auckland, Auckland, New Zealand
| | - A Strandell
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - A Vail
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - M van Wely
- Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - M Vercoe
- Cochrane Gynaecology and Fertility Group, University of Auckland, Auckland, New Zealand
| | - N L Vuong
- Department of Obstetrics and Gynaecology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - A Y Wang
- Faculty of Health, University of Technology, Sydney, Broadway, Australia
| | - R Wang
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - J Wilkinson
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - M A Youssef
- Department of Obstetrics & Gynaecology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - C M Farquhar
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand; Cochrane Gynaecology and Fertility Group, University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Arshad MA, Majzoub A, Esteves SC. Predictors of surgical sperm retrieval in non-obstructive azoospermia: summary of current literature. Int Urol Nephrol 2020; 52:2015-2038. [PMID: 32519242 DOI: 10.1007/s11255-020-02529-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/20/2022]
Abstract
Intracytoplasmic sperm injection (ICSI), combined with surgical sperm retrieval (SR) techniques, is the sole option for patients with non-obstructive azoospermia to achieve fertility; however, with suboptimal results. Given the variability in clinical presentation, the potential role of factors that can predict the likelihood of successful testicular SR needs to be clarified. This article summarizes the current evidence concerning the variables predicting SR success in non-obstructive azoospermic patients with spermatogenic failure. For this, we used 60 articles, including 46 original papers and six meta-analyses. Clinical and laboratory factors, as well as adjuvant therapies and surgical retrieval methods, were the factors most commonly investigated. We found that Klinefelter syndrome, Y chromosome microdeletions in regions AZFa/b, and Sertoli cell-only histopathology were associated with reduced SR success. By contrast, testis volume > 12.5 ml, history of cryptorchidism, use of micro-TESE as the sperm retrieval method, and adjuvant therapy were associated with improved SR success. None of the predictors, alone or combined, provide definitive information about the chances of harvesting sperm in men with non-obstructive azoospermia, except for Y chromosome microdeletions in regions AZFa/b. In the latter, SR success is virtually nil. We conclude that SR outcomes in men with non-obstructive azoospermia are difficult to predict based on the existing variables. Although several predictors can be used for patient counseling, their clinical value is limited to either ensure SR success or discourage reproductive urologists from recommending SR to men with non-obstructive azoospermia seeking fertility. A notable exception includes the deletions involving the regions AZFa and/or AZFb of the Y chromosome; the affected patients should be counseled against undergoing SR.
Collapse
Affiliation(s)
- Muhammad A Arshad
- Fatima Memorial Hospital College of Medicine and Dentistry, Lahore, Pakistan
- Nishter Hospital, Multan, Pakistan
| | - Ahmad Majzoub
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Weill Cornell Medicine- Qatar, Doha, Qatar
| | - Sandro C Esteves
- ANDROFERT, Andrology and Human Reproduction Clinic, Av. Dr. Heitor Penteado, 1464, Campinas, São Paulo, 13075-460, Brazil.
- Department of Surgery (Division of Urology), University of Campinas (UNICAMP), Campinas, Brazil.
- Faculty of Health, Department of Clinical Sciences, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
32
|
Xi Q, Zhang H, Zhang X, Jiang Y, Wang R, Liu R, Zhang H. Analysis of TATA-box binding protein associated factor 4b gene mutations in a Chinese population with nonobstructive azoospermia. Medicine (Baltimore) 2020; 99:e20561. [PMID: 32502024 PMCID: PMC7306362 DOI: 10.1097/md.0000000000020561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Nonobstructive azoospermia (NOA) is a severe form of male infertility. The molecular basis of NOA is still poorly understood. The aim of this study was to explore the associations between single nucleotide polymorphisms (SNPs) of the TATA-box binding protein associated factor 4b (TAF4B) gene and NOA. A total of 100 Han Chinese patients with NOA and 100 healthy men as controls were recruited. Targeted gene capture sequencing was performed. A total of 11 TAF4B SNPs were screened in the NOA and control subjects. Six synonymous and 4 nonsynonymous variants were detected. The c.11G>T (p.G4V) mutation was detected only in NOA patients. Polymorphism Phenotyping v2 and Sorting Intolerant From Tolerant analysis indicated that the p.G4V mutation influenced the protein structure of TAF4B. Haplotype analysis showed that the candidate SNPs did not independently associate with NOA and were found at extremely low frequencies in the subject population. Mutation Taster analysis indicated that the c.11G>T/p.G4V mutation was damaging. WebLogo analysis showed that the residue at amino acid 4 was relatively conserved. The p.Gly4Val substitution may affect the structure of the TAF4B protein. The c.11G>T mutation of the TAF4B gene may be associated with NOA in a Chinese population. Bioinformatics analysis indicated this variation may play an important role in the process of spermatogenesis.
Collapse
Affiliation(s)
- Qi Xi
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun
| | - Hao Zhang
- Center for Reproductive Medicine, Yanbian University Hospital, Yanji, China
| | - Xinyue Zhang
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun
| | - Yuting Jiang
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun
| | - Ruixue Wang
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun
| | - Ruizhi Liu
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun
| | - Hongguo Zhang
- Center for Reproductive Medicine and Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun
| |
Collapse
|
33
|
Davis H, Sosulski N, Civetta A. Reproductive isolation caused by azoospermia in sterile male hybrids of Drosophila. Ecol Evol 2020; 10:5922-5931. [PMID: 32607201 PMCID: PMC7319132 DOI: 10.1002/ece3.6329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/16/2020] [Accepted: 04/14/2020] [Indexed: 11/12/2022] Open
Abstract
Recently diverged populations in the early stages of speciation offer an opportunity to understand mechanisms of isolation and their relative contributions. Drosophila willistoni is a tropical species with broad distribution from Argentina to the southern United States, including the Caribbean islands. A postzygotic barrier between northern populations (North America, Central America, and the northern Caribbean islands) and southern populations (South American and the southern Caribbean islands) has been recently documented and used to propose the existence of two different subspecies. Here, we identify premating isolation between populations regardless of their subspecies status. We find no evidence of postmating prezygotic isolation and proceeded to characterize hybrid male sterility between the subspecies. Sterile male hybrids transfer an ejaculate that is devoid of sperm but causes elongation and expansion of the female uterus. In sterile male hybrids, bulging of the seminal vesicle appears to impede the movement of the sperm toward the sperm pump, where sperm normally mixes with accessory gland products. Our results highlight a unique form of hybrid male sterility in Drosophila that is driven by a mechanical impediment to transfer sperm rather than by an abnormality of the sperm itself. Interestingly, this form of sterility is reminiscent of a form of infertility (azoospermia) that is caused by lack of sperm in the semen due to blockages that impede the sperm from reaching the ejaculate.
Collapse
Affiliation(s)
- Hunter Davis
- Department of BiologyUniversity of WinnipegWinnipegMBCanada
| | | | | |
Collapse
|
34
|
Babakhanzadeh E, Khodadadian A, Nazari M, Dehghan Tezerjani M, Aghaei SM, Ghasemifar S, Hosseinnia M, Mazaheri M. Deficient Expression of DGCR8 in Human Testis is Related to Spermatogenesis Dysfunction, Especially in Meiosis I. Int J Gen Med 2020; 13:185-192. [PMID: 32523370 PMCID: PMC7237130 DOI: 10.2147/ijgm.s255431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/06/2020] [Indexed: 12/28/2022] Open
Abstract
Introduction DiGeorge syndrome critical region gene 8 (DGCR8) contributes to miRNA biogenesis, and defects in its expression could lead to defects in spermatogenesis. Methods Here, we assess gene and protein expression levels of DGCR8 in the testicular biopsy specimens obtained from men with obstructive azoospermia (OA, n = 19) and various types of non-obstructive azoospermia (NOA) including maturation arrest (MA, n = 17), Sertoli cell-only syndrome (SCOS, n = 20) and hypospermatogenesis (HYPO, 18). Also, samples of men with NOA were divided into two groups based on successful and unsuccessful sperm recovery, NOA+ in 21 patients and NOA− in 34 patients. Results Examinations disclosed a severe decrease in DGCR8 in samples with MA and SCOS in comparison to OA samples (P < 0.001). Also, the results showed DGCR8 has significantly lower expression in testis tissues of NOA− group in comparison to NOA+ group (p<0.05). Western blot analysis confirmed that the DGCR8 protein was not expressed in SCOS samples and had a very low expression in MA and HYPO samples. Discussion The results of this survey showed that DGCR8 is an important gene for the entire spermatogenesis pathway. Moreover, DGCR8 gene plays an important role in the diagnosis of NOA subgroups, and also the expression changes in it might contribute to SCOS or MA phenotypes. This gene with considering other related genes can also be a predictor of sperm retrieval.
Collapse
Affiliation(s)
- Emad Babakhanzadeh
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Medical Genetics Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Khodadadian
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Majid Nazari
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Seyed Mohsen Aghaei
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sina Ghasemifar
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mehdi Hosseinnia
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mahta Mazaheri
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
35
|
Babakhanzadeh E, Khodadadian A, Rostami S, Alipourfard I, Aghaei M, Nazari M, Hosseinnia M, Mehrjardi MYV, Jamshidi Y, Ghasemi N. Testicular expression of TDRD1, TDRD5, TDRD9 and TDRD12 in azoospermia. BMC MEDICAL GENETICS 2020; 21:33. [PMID: 32059713 PMCID: PMC7023801 DOI: 10.1186/s12881-020-0970-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/10/2020] [Indexed: 11/20/2022]
Abstract
Background Tudor domain-containing proteins (TDRDs) play a critical role in piRNA biogenesis and germ cell development. piRNAs, small regulatory RNAs, act by silencing of transposons during germline development and it has recently been shown in animal model studies that defects in TDRD genes can lead to sterility in males. Methods Here we evaluate gene and protein expression levels of four key TDRDs (TDRD1, TDRD5, TDRD9 and TDRD12) in testicular biopsy samples obtained from men with obstructive azoospermia (OA, n = 29), as controls, and various types of non-obstructive azoospermia containing hypospermatogenesis (HP, 28), maturation arrest (MA, n = 30), and Sertoli cell-only syndrome (SCOS, n = 32) as cases. One-way ANOVA test followed by Dunnett’s multiple comparison post-test was used to determine inter-group differences in TDRD gene expression among cases and controls. Results The results showed very low expression of TDRD genes in SCOS specimens. Also, the expression of TDRD1 and TDRD9 genes were lower in MA samples compared to OA samples. The expression of TDRD5 significantly reduced in SCOS, MA and HP specimens than the OA specimens. Indeed, TDRD12 exhibited a very low expression in HP specimens in comparison to OA specimens. All these results were confirmed by Western blot technique. Conclusion TDRDs could be very important in male infertility, which should be express in certain stages of spermatogenesis.
Collapse
Affiliation(s)
- Emad Babakhanzadeh
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Medical Genetics Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Khodadadian
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Saadi Rostami
- Department of Cellular and Molecular Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Iraj Alipourfard
- Center of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria.,School of Pharmacy, Faculty of Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Mohsen Aghaei
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Majid Nazari
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mehdi Hosseinnia
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mohammad Yahya Vahidi Mehrjardi
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Medical Genetics Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Yalda Jamshidi
- Genetics Centre, Molecular and Clinical Sciences Institute, St George's University of London, London, UK
| | - Nasrin Ghasemi
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. .,Abortion Research Centre, Yazd Reproductive Sicences Institue, Shahid sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
36
|
Rehman R, Zahid N, Amjad S, Baig M, Gazzaz ZJ. Relationship Between Smoking Habit and Sperm Parameters Among Patients Attending an Infertility Clinic. Front Physiol 2019; 10:1356. [PMID: 31736779 PMCID: PMC6834764 DOI: 10.3389/fphys.2019.01356] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 10/14/2019] [Indexed: 01/04/2023] Open
Abstract
Background This study aimed to estimate stress markers, oxidative stress (OS), reproductive hormones and sperm parameters in male smokers and non-smokers and observe the impact of oxidative stress markers and smoking on sperm count, motility and morphology in a selected population of Karachi, Pakistan. Methods This cross-sectional study was conducted from July 2017 to July 2018 at Aga Khan University (AKU), in Karachi, Pakistan. The subjects were recruited from the Sindh Institute of Reproductive Medicine (SIRM), Karachi based on defined inclusion criteria. The subjects were categorized into fertile and infertile based on cut off values of sperm parameters as recommended by the WHO i.e., sperm count/ejaculate of 39 × 106/ml, sperm motility 40% and normal morphology 4%. Two hundred eleven fertile and 165 infertile male subjects were included in the study. Serum cortisol, adrenaline, superoxide dismutase (SOD), and glutathione peroxidase (GPX) were analyzed by ELISA kits. Data was analyzed on SPSS-22. A p-value of <0.05 was considered statistically significant. Results Age, Body Mass Index (BMI), and body fat were similar among smokers and non-smokers. Age was significantly lower, while mean BMI and body fat were significantly higher among infertile smokers vs. fertile smokers (p-value < 0.05). The testosterone levels were significantly reduced among smokers as compared to non- smokers (p-value < 0.05). The median cortisol levels were increased as well as GPX, and steroid hormone-binding globulin (SHBG) were significantly reduced among smokers as compared to non-smokers. Additionally, the same findings with a significant difference have also been observed among infertile smokers as compared to fertile smokers (p-value < 0.05). This study has shown that the semen parameters (total count, motility, and morphology) are decreased in infertile smokers as compared to infertile non-smokers. Furthermore, the multivariate analysis showed that smoking causes a significant decrease in sperm count and morphology but it did not have any significant effect on motility. Conclusion Smoking has a significant effect on fertility, specifically sperm count and normal morphology of sperm. This might be due to OS produced by smoking, which has devastating effects on semen parameters, thus reducing male fertility. Infertility specialist should counsel their patients about the ill effects of smoking on their fertility status and should advise maintaining a healthy lifestyle, including normal weight and avoiding smoking, to prevent future health problems. Hence smokers should quit smoking for their next generation.
Collapse
Affiliation(s)
- Rehana Rehman
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Nida Zahid
- Department of Surgery, The Aga Khan University, Karachi, Pakistan
| | - Sofia Amjad
- Department of Physiology, Ziauddin University, Karachi, Pakistan
| | - Mukhtiar Baig
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zohair Jamil Gazzaz
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
37
|
Öztekin Ü, Caniklioğlu M, Sarı S, Selmi V, Gürel A, Işıkay L. Evaluation of Male Infertility Prevalence with Clinical Outcomes in Middle Anatolian Region. Cureus 2019; 11:e5122. [PMID: 31523553 PMCID: PMC6741393 DOI: 10.7759/cureus.5122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/10/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE the aim of this study is to determine the prevalence of male factor infertility with the clinical patterns of patients in our region. MATERIALS AND METHODS this is a descriptive retrospective study of 406 infertility cases presented at our urology clinic from February 2018 to February 2019. We assessed hormone and physical examination data, semen analysis results, the contribution of male and female factors to infertility, and types of infertility (i.e., primary or secondary). RESULTS the age of the male patients ranged from 18 to 50 years, with a mean of 30 ± 5 years. Asthenozoospermia was the leading cause of male factor infertility in 77 patients (19%). Male factors as the sole cause of infertility were found in 185 (45.6%) couples. Female factors as the sole cause were found in 32 couples (7.9%). Primary infertility was determined in 314 (77.3%) patients, and 92 (22.7%) had secondary infertility. CONCLUSION according to our results, the male infertility rate was high among couples reporting infertility. Couples should be informed about the causes of infertility, which may be due factors attributed to either sex.
Collapse
Affiliation(s)
- Ünal Öztekin
- Urology, Bozok University Faculty of Medicine, Yozgat, TUR
| | | | - Sercan Sarı
- Urology, Bozok University Faculty of Medicine, Yozgat, TUR
| | - Volkan Selmi
- Urology, Bozok University Faculty of Medicine, Yozgat, TUR
| | - Abdullah Gürel
- Urology, Bozok University Faculty of Medicine, Yozgat, TUR
| | - Levent Işıkay
- Urology, Bozok University Faculty of Medicine, Yozgat, TUR
| |
Collapse
|
38
|
Huang M, Zhu M, Jiang T, Wang Y, Wang C, Jin G, Guo X, Sha J, Dai J, Wang X, Hu Z. Fine mapping the MHC region identified rs4997052 as a new variant associated with nonobstructive azoospermia in Han Chinese males. Fertil Steril 2018; 111:61-68. [PMID: 30502936 DOI: 10.1016/j.fertnstert.2018.08.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/14/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To investigate the association between genetic variants in the major histocompatibility complex (MHC) region and nonobstructive azoospermia (NOA) susceptibility. DESIGN MHC region fine-mapping analysis based on previous NOA genome-wide association study (GWAS) data. SETTING Medical university. PATIENT(S) Nine hundred and eighty-one men with NOA and 1,657 normal fertile male controls. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) The MHC region imputation assessed with SNP2HLA software, taking the specific Han-MHC database as a reference panel; statistical significance of the MHC variants calculated using logistic regression models; functional annotation based on online public databases; and phenotypic variances explained by specific groups of genetic variants estimated using the fixed effects model from individual associations. RESULT(S) Two independent risk loci, rs7194 (odds ratio [OR] 1.37) at MHC class II molecules and rs4997052 (OR 1.30) at MHC class I molecules, were identified. Functional annotation showed rs7194 may tag the effect of multiple amino acid residues and the expression of HLA-DQB1 and HLA-DRB1; while rs4997052 showed the effect of amino acid changes of HLA-B at position 116 as well as the expression of HLA-B and CCHCR1, which coexpressed with genes enriched in pathways of spermatogenesis and male gamete generation. The novel variant rs4997052 identified in our study can explain another approximately 0.66% of the phenotypic variances of NOA. CONCLUSION(S) We fine-mapped the MHC region and identified two loci that independently drove NOA susceptibility. These results provide a deeper understanding of the association mechanisms of MHC and NOA risk.
Collapse
Affiliation(s)
- Mingtao Huang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Meng Zhu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Tingting Jiang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yifeng Wang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Cheng Wang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Guangfu Jin
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Juncheng Dai
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaoming Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhibin Hu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, People's Republic of China.
| |
Collapse
|
39
|
DNA Flow cytometric analysis of the human testicular tissues to investigate the status of spermatogenesis in azoospermic patients. Sci Rep 2018; 8:11117. [PMID: 30042518 PMCID: PMC6057995 DOI: 10.1038/s41598-018-29369-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/09/2018] [Indexed: 11/09/2022] Open
Abstract
A single, rapid and reproducible diagnostic test to predict the type of azoospermia and outcome of sperm retrieval is not yet available. So the feasibility of employing DNA flow cytometry for rapid investigation of the status of spermatogenesis in the patients with azoospermia was investigated. Testicular biopsies of 44 patients with azoospermia undergoing sperm-retrieval surgery and 4 controls were analyzed by flow cytometry to ascertain their testicular germ-cell patterns. The observed germ-cell pattern was further confirmed by RT-PCR analysis of the cell-specific markers and histology for some patients. The patients with Obstructive Azoospermia (OA) exhibited normal spermatogenesis similar to the control fertile patients showing the presence of diploid, double-diploid and haploid cells. The non-obstructive azoospermia (NOA) patients exhibited disrupted spermatogenesis with arrest at the pre-meiotic (only diploid cells present) or meiotic (diploid and double-diploid cells present) stages. The germ-cell pattern, as ascertained by flow cytometry, provided a clear picture of the intra-testicular spermatogenesis and the presence of spermatozoa in the patients’ testes, which was prognostic of their sperm-retrieval. DNA flow cytometry test to ascertain the testicular germ-cell pattern is simple in execution, analysis and interpretation, requires small amount of tissue and provides quantitative data about the status of spermatogenesis in patients. This test would allow comparable analysis of the status of spermatogenesis in patients across clinics and may form the basis for deciding future treatment and intervention strategies.
Collapse
|
40
|
Comizzoli P, Paulson EE, McGinnis LK. The mutual benefits of research in wild animal species and human-assisted reproduction. J Assist Reprod Genet 2018; 35:551-560. [PMID: 29470702 PMCID: PMC5949116 DOI: 10.1007/s10815-018-1136-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/06/2018] [Indexed: 01/08/2023] Open
Abstract
Studying the reproductive biology of wild animal species produces knowledge beneficial to their management and conservation. However, wild species also share intriguing similarities in reproductive biology with humans, thereby offering alternative models for better understanding the etiology of infertility and developing innovative treatments. The purpose of this review is to raise awareness in different scientific communities about intriguing connections between wild animals and humans regarding infertility syndromes or improvement of fertility preservation. The objectives are to (1) highlight commonalities between wild species and human fertility, (2) demonstrate that research in wild species-assisted reproductive technologies can greatly enhance success in human reproductive medicine, and (3) recognize that human fertility preservation is highly inspiring and relevant to wild species conservation. In addition to having similar biological traits in some wild species and humans, the fact of sharing the same natural environment and the common needs for more options in fertility preservation are strong incentives to build more bridges that will eventually benefit both animal conservation and human reproductive medicine.
Collapse
Affiliation(s)
- P Comizzoli
- Smithsonian Conservation Biology Institute, National Zoological Park, Washington, DC, 20008, USA.
| | - E E Paulson
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - L K McGinnis
- Department of Obstetrics and Gynecology and USC-Norris Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
41
|
Karabulut S, Keskin İ, Kutlu P, Delikara N, Atvar Ö, Öztürk Mİ. Male infertility, azoozpermia and cryptozoospermia incidence among three infertility clinics in Turkey. Turk J Urol 2018; 44:109-113. [PMID: 29511578 PMCID: PMC5832370 DOI: 10.5152/tud.2018.59196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/02/2017] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Semen parameters are directly correlated with the infertility of the male. Incidence rates of male factor infertility, azoospermia and cryptozoospermia differ according to many factors such as geographic region, age, occupation and body weight. The aim of the present study is to determine the incidence of male factor infertility, azoospermia and cryptozoospermia among patients who have been admitted to three separate infertility clinics in Turkey for infertility investigation and analyze the outcomes of these patients. MATERIAL AND METHODS A total of 9733 men, who have been admitted to 3 infertility clinics in Turkey due to infertility between March 2011 and October 2016, were included in the study. Male infertility, azoozpermia and cryptozoospermia incidence were calculated according to WHO criteria. RESULTS Male factor infertility was determined in 3114 (32%) of the patients including cases with azoospermia and cryptozoospermia. Azoospermia cases were observed in 570 (5.85%) and cryptozoospermia in 850 (8.73%) men. Azoospermic, and cryptozoospermic patients constitute 18.3%, and 27.2% of the male infertility cases. Sperm retrieval rates in diagnostic or oocyte pick-up plus testicular sperm extraction groups were found to be comparable (16.39%, and 41.3%, respectively). CONCLUSION The data obtained may help to estimate the number of in vitro fertilization cycles and testicular sperm extraction cases, to determine social security policies, and reproductive potential, and in the light of these data to establish social insurance policies. These data will help patients to decide on treatment alternatives, and guide the urologists about the issue.
Collapse
Affiliation(s)
- Seda Karabulut
- Medipol University, International School of Medicine, REMER (Regenerative And Restorative Medicine Research Center), İstanbul, Turkey
| | - İlknur Keskin
- Medipol University School of Medicine, REMER (Regenerative And Restorative Medicine Research Center), İstanbul, Turkey
| | - Pelin Kutlu
- Medicana Çamlıca Hospital, IVF Center, İstanbul, Turkey
| | - Nuri Delikara
- Florence Nightingale Hospital, IVF Unit, İstanbul, Turkey
| | | | - Metin İ. Öztürk
- Department of Urology, University of Health Sciences, Haydarpaşa Numune Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
42
|
Abou Alchamat G, Madania A, Alhalabi M. Mild androgen insensitivity syndrome (MAIS): the identification of c.1783C>T mutation in two unrelated infertile men. BMJ Case Rep 2017; 2017:bcr-2017-220361. [PMID: 28659371 DOI: 10.1136/bcr-2017-220361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Two unrelated men complaining of primary male infertility presented to Orient Hospital in Damascus city. Physical examination showed moderate hypoandrogenic features. Both men were azoospermic. Hormone profiles revealed an elevation of follicle-stimulating hormone in one patient, but all the other hormones tested were within normal limits for both patients. Further genetic analyses, including karyotype and microdeletions in the AZF region of the Y chromosome, were normal in both patients. Mild androgen insensitivity syndrome was expected in the two patients. Sequencing analysis of the first exon in the androgen receptor (AR) gene have shown c.1783C>T mutation in the two patients with azoospermia. This paper sheds light on the need to screen for mutations in the AR gene, causing male infertility whenever mild hypoandrogenic features are present with unexplained male infertility.
Collapse
Affiliation(s)
- Ghalia Abou Alchamat
- Department of Biology, Faculty of Sciences, Damascus University, Damascus, Syrian Arab Republic
| | - Ammar Madania
- Department of Radiation Medicine, Atomic Energy Commission, Damascus, Syrian Arab Republic
| | - Marwan Alhalabi
- Department of Reproductive Medicine, Genetics and Embryology, Faculty of Medicine, Damascus University, Damascus, Syrian Arab Republic.,Assisted Reproduction Unit, Orient Hospital, Damascus, Syrian Arab Republic
| |
Collapse
|
43
|
Arafat M, Har-Vardi I, Harlev A, Levitas E, Zeadna A, Abofoul-Azab M, Dyomin V, Sheffield VC, Lunenfeld E, Huleihel M, Parvari R. Mutation in TDRD9 causes non-obstructive azoospermia in infertile men. J Med Genet 2017; 54:633-639. [PMID: 28536242 DOI: 10.1136/jmedgenet-2017-104514] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/15/2017] [Accepted: 04/02/2017] [Indexed: 11/04/2022]
Abstract
BACKGROUND Azoospermia is diagnosed when sperm cells are completely absent in the ejaculate even after centrifugation. It is identified in approximately 1% of all men and in 10%-20% of infertile males. Non-obstructive azoospermia (NOA) is characterised by the absence of sperm due to either a Sertoli cell-only pattern, maturation arrest, hypospermatogenesis or mixed patterns. NOA is a severe form of male infertility, with limited treatment options and low fertility success rates. In the majority of patients, the cause for NOA is not known and mutations in only a few genes were shown to be causative. AIM We investigated the cause of maturation arrest in five azoospermic infertile men of a large consanguineous Bedouin family. METHODS AND RESULTS Using whole genome genotyping and exome sequencing we identified a 4 bp deletion frameshift mutation in TDRD9 as the causative mutation with a Lod Score of 3.42. We demonstrate that the mutation results in a frameshift as well as exon skipping. Immunofluorescent staining with anti-TDRD9 antibody directed towards the N terminus demonstrated the presence of the protein in testicular biopsies of patients with an intracellular distribution comparable to a control biopsy. The mutation does not cause female infertility. CONCLUSION This is the first report of a recessive deleterious mutation in TDRD9 in humans. The clinical phenotype recapitulates that observed in the Tdrd9 knockout mice where this gene was demonstrated to participate in long interspersed element-1 retrotransposon silencing. If this function is preserved in human, our data underscore the importance of maintaining DNA stability in the human male germ line.
Collapse
Affiliation(s)
- Maram Arafat
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Iris Har-Vardi
- Fertility and IVF Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Avi Harlev
- Fertility and IVF Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eliahu Levitas
- Fertility and IVF Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Atif Zeadna
- Fertility and IVF Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Maram Abofoul-Azab
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Victor Dyomin
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Institute of Pathology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Val C Sheffield
- Department of Pediatrics, Division of Medical Genetics, University of Iowa, Iowa City, USA
| | - Eitan Lunenfeld
- Fertility and IVF Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ruti Parvari
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
44
|
Mou L, Gui Y. A novel variant of androgen receptor is associated with idiopathic azoospermia. Mol Med Rep 2016; 14:2915-20. [PMID: 27498682 PMCID: PMC5042743 DOI: 10.3892/mmr.2016.5587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 03/07/2016] [Indexed: 12/13/2022] Open
Abstract
A variety of genetic variants can lead to abnormal human spermatogenesis. The androgen receptor (AR) is an important steroid hormone receptor that is critical for male sexual differentiation and the maintenance of normal spermatogenesis. In the present study, each exon of AR in 776 patients diagnosed with idiopathic azoospermia (IA) and 709 proven fertile men were sequenced using use panel re‑sequencing methods to examine whether AR is involved in the pathogenesis of IA. Two synonymous variants and seven missense variants were detected. Of the missense variants, a luciferase assay demonstrated that the R630W variant reduced the transcriptional regulatory function of AR. This novel variant (p. R630W) of AR is the first to be identified in association with IA, thereby highlighting the importance of AR during spermatogenesis.
Collapse
Affiliation(s)
- Lisha Mou
- Shenzhen Domesticated Organ Medical Engineering Research and Development Center, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Biomedical Research Institute, Shenzhen PKU‑HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
45
|
Dhanoa JK, Mukhopadhyay CS, Arora JS. Y-chromosomal genes affecting male fertility: A review. Vet World 2016; 9:783-91. [PMID: 27536043 PMCID: PMC4983133 DOI: 10.14202/vetworld.2016.783-791] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/23/2016] [Indexed: 12/30/2022] Open
Abstract
The mammalian sex-chromosomes (X and Y) have evolved from autosomes and are involved in sex determination and reproductive traits. The Y-chromosome is the smallest chromosome that consists of 2-3% of the haploid genome and may contain between 70 and 200 genes. The Y-chromosome plays major role in male fertility and is suitable to study the evolutionary relics, speciation, and male infertility and/or subfertility due to its unique features such as long non-recombining region, abundance of repetitive sequences, and holandric inheritance pattern. During evolution, many holandric genes were deleted. The current review discusses the mammalian holandric genes and their functions. The commonly encountered infertility and/or subfertility problems due to point or gross mutation (deletion) of the Y-chromosomal genes have also been discussed. For example, loss or microdeletion of sex-determining region, Y-linked gene results in XY males that exhibit female characteristics, deletion of RNA binding motif, Y-encoded in azoospermic factor b region results in the arrest of spermatogenesis at meiosis. The holandric genes have been covered for associating the mutations with male factor infertility.
Collapse
Affiliation(s)
- Jasdeep Kaur Dhanoa
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana - 141 004, Punjab, India
| | - Chandra Sekhar Mukhopadhyay
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana - 141 004, Punjab, India
| | - Jaspreet Singh Arora
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana - 141 004, Punjab, India
| |
Collapse
|
46
|
Abstract
BACKGROUND: The number of couples that meet the definition of infertility at reproductive ages is increasing worldwide. One of the most known conditions of infertility in males is azoospermia, defined as complete absence of spermatozoa in the semen. Azoospermia manifests in two forms, namely obstructive and non-obstructive azoospermia. Although the presence of antisperm antibody (ASA) has been reported in 88% of the patients with obstructive azoospermia (OA), interestingly, there is no data regarding ASA targets in OA individuals. AIM: The present study aimed to identify sperm antibody targets in a group of OA men. SETTINGS AND DESIGN: The present study was carried out on 27 OA infertile men and 27 healthy fertile age-matched males as cases and controls, respectively. SUBJECTS AND METHODS: The sperm proteome was separated using two-dimensional gel electrophoresis technique, transferred onto the polyvinylidene fluoride membrane, and blotted with the sera of a group of OA men. Then, it was compared with the membranes blotted with the sera of a group of healthy fertile men. Matrix-assisted laser desorption/ionization time-of-flight/time-of-flight (MALDI TOF/TOF) mass spectrometry was used to identify the different blotted spots and finally the results of the mass analysis were confirmed using reverse transcriptase polymerase chain reaction method. RESULTS: The results indicated that OA patients might produce antibody against two sperm proteins, Tektin-2 and triose phosphate isomerase. Moreover, the expressions of the two targeted proteins were confirmed at RNA level. CONCLUSIONS: The findings of the present study revealed two functionally important sperm proteins as antibody targets in azoospermic men.
Collapse
Affiliation(s)
- Mohammad-Sadegh Soltani Zangbar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee of Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Keshtgar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jaleh Zolghadri
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Obstetrics and Gynecology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Gharesi-Fard
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department Reproductive Biology, Proteomics Laboratory, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
47
|
Ma Q, Li Y, Guo H, Li C, Chen J, Luo M, Jiang Z, Li H, Gui Y. A Novel Missense Mutation in USP26 Gene Is Associated With Nonobstructive Azoospermia. Reprod Sci 2016; 23:1434-41. [PMID: 27089915 DOI: 10.1177/1933719116641758] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate whether ubiquitin-specific peptidase 26 (USP26) gene variations were associated with nonobstructive azoospermia (NOA). METHODS Seven hundred and seventy-six patients diagnosed with NOA and 709 proven fertile men were included in this study. Genetic variations of infertility-related genes, including USP26, were identified by selected exonic sequencing. The effects of USP26 mutations on androgen receptor (AR) binding, ubiquitination, and transcriptional activity were detected by immunoprecipitation and luciferase assay in Hela and TM4 cells. RESULTS Six novel missense mutations and 1 novel synonymous mutation of USP26 unique to the patients with NOA were identified. Of these missense mutations, USP26 R344W remarkably reduced the binding affinity and deubiquitinating activity of USP26 to AR, thus eliminated the inhibitory effect of USP26 on transcriptional activity of AR in Hela and TM4 cells. CONCLUSION A novel USP26 variant p.R344W is associated with NOA probably through affecting AR function.
Collapse
Affiliation(s)
- Qian Ma
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, PR China
| | - Yuchi Li
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, PR China
| | - Huan Guo
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, PR China
| | - Cailing Li
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, PR China
| | - Jianbo Chen
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, PR China
| | - Manling Luo
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, PR China
| | - Zhimao Jiang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, PR China
| | - Honggang Li
- The Family Planning Research Institute/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, PR China
| |
Collapse
|
48
|
Refaat AM. Pilot study for early prognosis of Azoospermia in relation to Y-STR Profiling. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2016. [DOI: 10.1016/j.ejmhg.2015.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
49
|
Fourteen babies born after round spermatid injection into human oocytes. Proc Natl Acad Sci U S A 2015; 112:14629-34. [PMID: 26575628 DOI: 10.1073/pnas.1517466112] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During the human in vitro fertilization procedure in the assisted reproductive technology, intracytoplasmic sperm injection is routinely used to inject a spermatozoon or a less mature elongating spermatid into the oocyte. In some infertile men, round spermatids (haploid male germ cells that have completed meiosis) are the most mature cells visible during testicular biopsy. The microsurgical injection of a round spermatid into an oocyte as a substitute is commonly referred to as round spermatid injection (ROSI). Currently, human ROSI is considered a very inefficient procedure and of no clinical value. Herein, we report the birth and development of 14 children born to 12 women following ROSI of 734 oocytes previously activated by an electric current. The round spermatids came from men who had been diagnosed as not having spermatozoa or elongated spermatids by andrologists at other hospitals after a first Micro-TESE. A key to our success was our ability to identify round spermatids accurately before oocyte injection. As of today, all children born after ROSI in our clinic are without any unusual physical, mental, or epigenetic problems. Thus, for men whose germ cells are unable to develop beyond the round spermatid stage, ROSI can, as a last resort, enable them to have their own genetic offspring.
Collapse
|
50
|
Abstract
Secretory azoospermia is a severe form of male infertility caused by unknown factors. DAX-1 is predominantly expressed in mammalian reproductive tissues and plays an important role in spermatogenesis because Dax-1 knockout male mice show spermatogenesis defects. To examine whether DAX-1 is involved in the pathogenesis of secretory azoospermia in humans, we sequenced all of the exons of DAX-1 in 776 patients diagnosed with secretory azoospermia and 709 proven fertile men. A number of coding mutations unique to the patient group, including two synonymous mutations and six missense mutations, were identified. Of the missense mutations, our functional assay demonstrated that the V385L mutation caused the reduced functioning of DAX-1. This novel mutation (p. V385L) of DAX-1 is the first to be identified in association with secretory azoospermia, thereby highlighting the important role of DAX-1 in spermatogenesis.
Collapse
Affiliation(s)
- Lisha Mou
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Domesticated Organ Medical Engineering Research and Development Center, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Biomedical Research Institute, Shenzhen PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Nie Xie
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Domesticated Organ Medical Engineering Research and Development Center, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Biomedical Research Institute, Shenzhen PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Lihua Yang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Biomedical Research Institute, Shenzhen PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Yuchen Liu
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Domesticated Organ Medical Engineering Research and Development Center, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Ruiying Diao
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Domesticated Organ Medical Engineering Research and Development Center, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Zhiming Cai
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Domesticated Organ Medical Engineering Research and Development Center, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Honggang Li
- The Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- * E-mail: (YG); (HL)
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Biomedical Research Institute, Shenzhen PKU-HKUST Medical Center, Shenzhen, 518036, China
- * E-mail: (YG); (HL)
| |
Collapse
|